You are on page 1of 357

An imprint of Elsevier Inc.

FINE NEEDLE ASPIRATION CYTOLOGY


© 2007, Elsevier Inc. All rights reserved.

ISBN-13: 978-0-443-06731-0
ISBN-10: 0-443-06731-7

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 permission of the Publishers. Permissions may be sought directly from Elsevier’s Health
Sciences Rights Department, 1600 John F. Kennedy Boulevard, Suite 1800, Philadelphia, PA
19103–2899, USA: phone: (+1) 215 239 3804; fax: (+1) 215 239 3805; or, e-mail: healthpermissions@
elsevier.com. You may also complete your request on-line via the Elsevier homepage (http://www.
elsevier.com), by selecting ‘Support and contact’ and then ‘Copyright and Permission’.

NOTICE

Medical knowledge is constantly changing. 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 administration, and contraindications. It is the
responsibility of the practitioner, relying on experience and knowledge of the patient, to
determine dosages and the best treatment for each individual patient. Neither the Publisher nor
the author assume any liability for any injury and/or damage to persons or property arising
from this publication.
The Publisher

First published 2007

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

Commissioning Editor: Michael Houston


Project Development Manager: Claire Bonnet
Project Manager: Cheryl Brant
Design Manager: Louis Forgione
Marketing Manager(s) (USA/UK): Kathleen Neely/Leontine Treur Working together to grow
libraries in developing countries
Printed in China www.elsevier.com | www.bookaid.org | www.sabre.org

Last digit is the print number: 9 8 7 6 5 4 3 2 1

FM-F06731.indd iv 10/26/2006 10:18:19 AM


List of Contributors

Fadi W Abdul-Karim, MD Shahla Masood, MD


Professor of Pathology Professor and Associate Chair
Department of Pathology University of Florida
University Hospitals of Cleveland Chief of Pathology
Cleveland, OH Department of Pathology
USA Shands Jacksonville Medical Center
Jacksonville, FL
Syed Z Ali, MD USA
Associate Professor of Pathology and Radiology
Associate Director, Division of Cytopathology Ritu Nayar, MD
Johns Hopkins University School of Medicine Associate Professor and Director of Cytopathology
Baltimore, MD Department of Pathology
USA Feinberg School of Medicine
Northwestern University
Ema A Berbescu, MD Chicago, IL
Assistant Professor of Pathology
USA
Division of Anatomic Pathology
Medical College of Virginia Marlo M Nicolas, MD
Virginia Commonwealth University Health System Genitourinary Fellow
Richmond, VA Department of Pathology
USA University of Texas, MD Anderson Cancer Center
Houston, TX
Katherine Berezowski, MD USA
Assistant Professor
Department of Pathology Anil V Parwani, MD, PhD
The George Washington University Assistant Professor
Washington, DC Department of Pathology
USA University of Pittsburgh Medical Center
Pittsburgh, PA
Martha Bishop Pitman, MD USA
Associate Professor of Pathology
Harvard Medical School Celeste N Powers, MD, PhD
Assistant Director of Cytopathology Professor and Chair
Director of the Fine Needle Aspiration Biopsy Service Division of Anatomic Pathology
Massachusetts General Hospital Medical College of Virginia
Boston, MA Virginia Commonwealth University Health System
USA Richmond, VA
USA
Yener S Erozan, MD
Professor of Pathology Natasha Rekhtman, MD, PhD
Department of Pathology Assistant, Department of Pathology
The Johns Hopkins University School of Medicine Division of Surgical Pathology
Baltimore, MD Johns Hopkins University
USA Baltimore, MD
USA
Michael O Idowu, MD
Assistant Professor of Pathology Mary K Sidawy, MD
Division of Anatomic Pathology Professor of Pathology, Director of Anatomic Pathology
Medical College of Virginia Georgetown University
Virginia Commonwealth University Health System Washington, DC
Richmond, VA USA
USA
Sana O Tabbara, MD
Igor Jovanovic, MD, PhD Professor of Pathology
Cytopathology Fellow The George Washington University
Department of Pathology Washington, DC
George Washington University USA
Washington, DC
USA

FM-F06731.indd v 10/26/2006 10:18:19 AM


vi LIST OF CONTRIBUTORS

Paul E Wakely Jr, MD Maureen F Zakowski, MD


Department of Pathology Attending Pathologist
The Ohio State University Department of Pathology
College of Medicine Memorial Sloan-Kettering Cancer Center
Columbus, OH New York, NY
USA USA

FM-F06731.indd vi 10/26/2006 10:18:19 AM


Foreword

The study and practice of anatomic pathology is both only written extensively in these areas, but, more
exciting and overwhelming. Surgical pathology, with importantly, have been practicing cytopathologists with
all of the subspecialties it encompasses, and cytopathol- vast practical experience. As with other editions in the
ogy have become increasingly complex and sophisti- Foundations in Diagnostic Pathology series, the informa-
cated, and it is not possible for any individual to master tion is presented in a straightforward and accessible
the skills and knowledge required to perform all of manner, including numerous practical tables and
these tasks at the highest level. Simply being able to impressive photomicrographs. Where appropriate, the
make a correct diagnosis is challenging enough, but the authors seamlessly integrate ancillary diagnostic
standard of care has far surpassed merely providing a techniques including immunohistochemistry, electron
diagnosis. Pathologists are now asked to provide large microscopy and molecular techniques, which are also
amounts of ancillary information, both diagnostic and an important part of the cytopathologist’s diagnostic
prognostic, often on small amounts of tissue, a task armamentarium.
that can be daunting even to the most experienced This edition is organized into ten chapters covering
pathologist. the full spectrum of fine needle aspiration cytology in
Although large general surgical pathology textbooks an organ-by-organ fashion. The first three chapters
are useful resources, they by necessity could not possi- focus on aspiration cytology of salivary glands, thyroid
bly cover many of the aspects that pathologists needs to and lymph nodes. Drs. Abdul-Karim and Massood
know and include in their reports. As such, the concept provide superb discussions on the finer points of fine
behind Foundations in Pathology was born. This series needle aspiration of soft tissue tumors and tumors
is designed to cover the major areas of surgical and of the breast. Separate chapters covering the lung,
cytopathology, and each edition is focused on one major mediastinum, liver, pancreas, kidney and adrenal
topic. The goal of every book in this series is to provide gland are also provided. The chapters are uniformly
the essential information that any pathologist, whether well organized, concisely written and beautifully
general or subspecialized, in training or in practice, photomicrographed.
would find useful in the evaluation of virtually any type I wish to extend my sincerest gratitude to Drs. Sidawy
of specimen encountered. and Ali for leading this outstanding effort. Clearly, they
Dr. Mary Sidawy from Georgetown and Dr. Syed Ali poured their heart and soul into this edition of the
from Johns Hopkins Hospital, both outstanding and Foundations in Diagnostic Pathology series, and for this
renowned cytopathologists, have edited what I believe I am truly grateful. I would also like to extend my appre-
to be an outstanding state-of-the-art book covering the ciation to the authors who took time from their busy
essential aspects of fine needle aspiration cytology. This schedules to contribute their expertise and knowledge.
book cuts to the essentials of what all pathologists I sincerely hope you enjoy this volume of Foundations
who practice the art of cytopathology need to know in Diagnostic Pathology.
about this challenging field. The list of contributors is
truly impressive and includes authors who have not John R. Goldblum, M.D.

FM-F06731.indd vii 10/26/2006 10:18:19 AM


To our families,
Tony, Michelle, and Nicholas
Tehmina, Adeel, and Sabeen

FM-F06731.indd viii 10/26/2006 10:18:19 AM


Preface

In 1930, Martin and Ellis, recognizing the value of referencing the histopathologic characteristics with the
cytologic sampling, published a cornerstone article cytologic features reflects our conviction that the two
entitled “Biopsy by Needle Puncture and Aspiration.” disciplines are complementary.
Fine needle aspiration (FNA) began gaining momen- Cytopathologists enjoy a position at the forefront of
tum in Europe in the 1950s; however, it was not until diagnostic anatomic pathology; we intimately interact
the 1980s when it became widely accepted in the United with many disciplines of clinical medicine. Therefore,
States. FNA of palpable and deep-seated lesions has emphasis was placed on the interpretation of morpho-
quickly grown to become a powerful diagnostic tool in logic findings in the proper clinicoradiologic context
the diagnosis of neoplastic, reactive, and infectious pro- leading to a more meaningful and valid list of differen-
cesses. But as we acquired more experience and knowl- tial diagnoses of FNA interpretation.
edge we developed a better appreciation of the pitfalls The book is aimed primarily at pathology residents
and limitations in interpreting cytologic samples. and fellows, as well as practicing pathologists looking
There is no paucity of published scientific material for a practical overview of FNA cytopathology. Our
on aspiration cytopathology; we have seen an exponen- ultimate goal is the best patient care based on timely and
tial growth in the cytopathology literature paralleling its accurate FNA interpretation. It is our hope that this
widespread success as a medical discipline. This volume book will further the understanding of FNA and its role
in the series Foundations in Diagnostic Pathology, offers in patient management. We would like to thank whole-
several notable features. FNA of most commonly heartedly the authors that contributed time and effort to
sampled sites are presented in ten chapters authored by this project; our mentors who inspired us, and our resi-
experts in the field. The template format throughout the dents and fellows for constantly challenging us at the
book provides a high degree of consistency in style and microscope. We sincerely hope that this book will
presentation. Furthermore, the use of Facts and Fea- convey our enthusiasm to the discipline of cytopathol-
tures boxes allows easy access to key information. High ogy, an area that we find exciting, challenging and
resolution images are the cornerstone of morphologic fulfilling in our daily practice and teaching.
discussion of any given entity with the text revolving
around issues of practical diagnostic and clinical impor- Mary K. Sidawy, M.D.
tance. Differential diagnoses, potential pitfalls, and the Syed Z. Ali, M.D.
use of ancillary techniques are emphasized. Cross October 2006

FM-F06731.indd ix 10/26/2006 10:18:19 AM


1 Salivary Glands
Sana O Tabbara

plasm. Acinar cells are arranged in acinar clusters that


INTRODUCTION appear to have a grape-like configuration and are some-
times attached to sheets of ductal cells. Ductal cells are
less numerous than acinar cells. They are small, bland
Fine needle aspiration (FNA) of salivary glands is cuboidal cells with scant dense cytoplasm and are orga-
widely used in the initial diagnosis of salivary gland nized in small honeycomb sheets or occasionally branch-
swellings. It is a simple procedure with limited compli- ing tubules.
cations that provides rapid and valuable information This chapter will address mass-producing lesions of
for the subsequent planning of patient management. the salivary gland that are evaluated by FNA. These
The cytologic features of common salivary gland neo- lesions will be presented according to their line of
plasms and inflammatory processes are well recog- differentiation.
nized, and accurate diagnosis is possible in most cases.
Salivary gland lesions involve most commonly the
parotid glands, followed by the submandibular glands
and the minor salivary glands. FNA of masses in the NON-NEOPLASTIC LESIONS
salivary gland area serves multiple purposes: it helps
determine the origin of the mass (salivary versus non-
salivary), it sorts neoplastic from non-neoplastic pro- SIALADENOSIS
cesses, and in most neoplastic cases it distinguishes
benign from malignant tumors. Reported sensitivity for
presence of tumor and specificity for absence of neo- Sialadenosis is a condition associated with malnutri-
plasm are >90%, but accuracy for a specific diagnosis tion, diabetes, bulimia, alcoholism, cirrhosis, and some
is lower. False negative diagnoses result mostly from drugs such as antihypertensives. Sialadenosis occurs
sampling errors and inadequately sampled cystic lesions, mostly in the parotid and is often bilateral. Typically,
whereas false positive diagnoses are encountered mostly smears are cellular and comprise normal salivary gland
in benign neoplasms with cytologic atypia and in basa- elements. The features differentiating such from normal
loid lesions. In FNAs of salivary gland neoplasms where salivary gland tissue are the hypertrophic, enlarged
a specific diagnosis cannot be reached, it is important to acinar cells; however, the size difference from normal
indicate if the tumor is a low- or a high-grade neoplasm acinar cells is usually difficult to appreciate. When
in order to guide surgical management. normal tissue without an inflammatory background
Non-neoplastic salivary gland lesions range between is aspirated from an enlarged gland, a sampling error
50% and 60% in some large series. FNA truncates should be entertained, unless the patient is proven to
patient management in such instances and prevents have no mass lesion by additional evaluations, includ-
unnecessary surgery, resulting in a 30% decrease of ing re-aspiration, radiographic evaluation, and clinical
surgical excisions of salivary gland lesions. Eventually, follow-up. If all evaluations are negative, the diagnosis
10–15% of non-neoplastic cases on FNA will require of sialadenosis should be considered.
surgery for definitive diagnosis.
FNA of a normal salivary gland tissue yields a sparsely
cellular smear that contains a mixture of acinar cells,
SIALADENITIS
ductal cells, mature adipose tissue, and scattered small
acinar bare nuclei (Fig. 1-1). Serous acinar cells are seen
in FNAs from all salivary glands. They are pyramidal- CLINICAL FEATURES
shaped and have abundant granular or finely vacuolated
cytoplasm containing periodic acid–Schiff (PAS)-
positive diastase-resistant zymogen granules, and a Acute sialadenitis is usually related to infection (viral,
small eccentric round nucleus with an inconspicuous bacterial, fungal) and may develop as a postoperative
nucleolus. Outside the parotid, acinar cells show both complication or in relation to sialolithiasis. It presents
serous and mucinous differentiation. Mucinous acinar with diffusely enlarged, tender glands, most frequently
cells are columnar and have a pale vacuolated cyto- the parotids.
1

Ch001-F06731.indd 1 10/26/2006 10:19:23 AM


2 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-1
Normal salivary gland. An aggregate
of salivary gland acini surrounds
a portion of ductal epithelium
arranged as a branching tubular
structure. Acinar cells have abun-
dant granular or finely vacuolated
cytoplasm, and small, regular, round
nuclei located peripherally within
the individual cell. The ductal epi-
thelium consists of uniform cuboi-
dal cells with bland round nuclei
arranged in orderly pattern. Alcohol-
fixed, Papanicolaou stain, medium
power.

SIALADENITIS – DISEASE FACT SHEET SIALADENITIS – PATHOLOGIC FEATURES

Clinical Features Cytopathologic Findings


៉ Acute sialadenitis: infectious (viral, bacterial, fungal); enlarged ៉ Acute sialadenitis: neutrophils, fibrin, necrotic debris
tender gland ៉ Chronic sialadenitis: variable with duration of disease; mixed
៉ Chronic sialadenitis: duct stricture, obstruction, trauma; diffuse mononuclear infl ammatory cells; salivary gland epithelium,
gland enlargement or mass; exacerbations and remissions predominantly ductal; fibrous stroma; squamous and mucous cell
៉ Granulomatous sialadenitis: infectious, systemic autoimmune metaplasias; crystalline material
disease, rarely neoplasms ៉ Granulomatous sialadenitis: epithelioid histiocytes,
multinucleated giant cells, possible necrosis
Treatment
៉ Variable, treat etiology Differential Diagnosis and Pitfalls
៉ Predominance of lymphoid cells, lack of epithelium: intraparotid
lymph node
៉ Lymphocyte-rich lesions: lymphoepithelial lesion, Warthin
tumor, malignant lymphoma
Chronic sialadenitis is more commonly found in the ៉ Epithelial metaplasia: mucoepidermoid carcinoma (SCC not
submandibular glands and is often associated with discussed)
duct stricture, obstruction (e.g. sialolithiasis), or trauma.
Clinically, it is characterized by exacerbations and
remissions associated with eating. Chronic sialadenitis
typically presents with a diffuse gland enlargement
and cultures must be obtained and may identify the
but may produce a mass (Kuttner tumor) that must be
causative organisms. In chronic sialadenitis, the cellu-
differentiated from a neoplastic process. Granuloma-
lar composition of the FNA material varies with the
tous sialadenitis may result from a variety of infections,
duration of the disease. Smears contain a heteroge-
including mycobacterial, fungal, cat scratch, and toxo-
neous population of lymphocytes admixed with benign
plasmosis, or may be associated with systemic diseases
salivary gland tissue. Both acinar and ductal epithe-
such as sarcoidosis. Rarely, granulomatous sialadenitis
lium may be present. In disease of long duration, the
is a manifestation of malignant neoplasms such as
acinar epithelium may atrophy, decreasing the number
Hodgkin or T-cell lymphoma or metastatic carcinoma.
and size of the acinar cells and making the ductal
component more prominent (Fig. 1-2). The smear back-
ground may contain mucus, debris, and/or various
CYTOPATHOLOGIC FEATURES crystalloids (Fig. 1-3). Spindle-shaped fibroblasts and
collagenous stromal fragments may also be noted
(Fig. 1-4). Duct obstruction may cause squamous or
FNA smears of acute sialadenitis contain neutrophils, mucinous metaplasia (Fig. 1-5). Granulomatous sialad-
fibrin, and necrotic debris. Material for special stains enitis is characterized by the presence of aggregates of

Ch001-F06731.indd 2 10/26/2006 10:19:24 AM


CHAPTER 1 Salivary Glands 3

FIGURE 1-2
Chronic sialadenitis. Numerous lym-
phocytes and scattered epithelial
and stromal fragments are compat-
ible with chronic sialadenitis. Typi-
cally there is a decrease in acinar
cells and a predominance of ductal
epithelium. Alcohol-fixed, Papani-
colaou stain, low power.

FIGURE 1-3
Chronic sialadenitis. Rhomboid-
shaped crystalloids are present in a
background of mononuclear infl am-
matory cells including lymphocytes
and histiocytes. Fibrin and cellular
debris are also noted. Alcohol-fixed,
Papanicolaou stain, medium power.

Ch001-F06731.indd 3 10/26/2006 10:19:24 AM


4 FINE NEEDLE ASPIRATION CYTOLOGY

submandibular) and lacrimal glands. FNA smears


consist of an abundant mixed population of lympho-
cytes, plasma cells, and tingible-body macrophages with
rare epithelial elements arranged in cohesive clusters
and infiltrated by lymphocytes, forming the characteris-
tic lymphoepithelial islands. Because these lesions have
a propensity to develop into lymphomas, immunophe-
notypic studies and flow cytometry may be useful in
selected cases.
Aspirate smears from glands with patchy involve-
ment by chronic sialadenitis may look normal, with
benign salivary gland epithelial cells and little or no
inflammation.
Extensive squamous and mucinous metaplasia in
chronic sialadenitis may produce atypical cells that can
mimic necrotizing sialometaplasia or mucoepidermoid
carcinoma. Necrotizing sialometaplasia occurs almost
exclusively in the minor salivary glands, particularly the
palate, in contrast to the typical submandibular gland
presentation of chronic sialadenitis. Moreover, the rela-
tively low cellularity of the epithelial component and
the presence of an inflammatory background in sialad-
enitis should aid in its distinction from low-grade muco-
epidermoid adenocarcinoma. Radiation sialadenitis can
result in the presence of atypical epithelial cells, but the
cellularity of these aspirates is low, in contrast to the
high cellularity of malignant neoplasms.
Intraparotid lymph nodes yield a cellular aspirate
containing a polymorphous population of lymphoid
cells with large and small lymphocytes and immuno-
blasts with few or no epithelial cells and no stromal
fragments. Aspirate from Warthin tumors that are poor
in epithelium may mimic chronic sialadenitis. Care
must also be taken to distinguish sheets of oncocytes
FIGURE 1-4 from sheets of squamous metaplastic cells that can be
Chronic sialadenitis. A collagenous stromal fragment containing bland
seen in chronic sialadenitis. Lastly, malignant lym-
spindle cells is a common finding in long-standing chronic sialadenitis. phoma, if suspected, can be identified by ancillary
A rare acinar structure is noted in the vicinity. Alcohol-fixed, Papani- testing.
colaou stain, high power. Courtesy of Ms Jamie L Covell, The University
of Virginia Health Sciences Center.

NON-NEOPLASTIC CYSTIC LESIONS


epithelioid histiocytes and associated multinucleated
giant cells (Fig. 1-6). Epithelioid histiocytes are recog-
A cystic lesion is encountered in about 5% of FNAs of
nized by their elongated, finely vacuolated cytoplasm
salivary glands. A cystic component in a salivary gland
and bland, occasionally bent nuclei. Their presence is
mass may be neoplastic or non-neoplastic in nature.
an indication for the performance of special stains or
The two most common non-neoplastic cystic lesions
culture for the identification of microorganisms.
encountered are HIV-associated cystic lymphoepi-
thelial lesions and mucocele/retention cysts. HIV-
associated cystic lymphoepithelial lesions may be one
DIFFERENTIAL DIAGNOSIS AND PITFALLS of the presenting criteria for the HIV-related complex.
They occur almost exclusively in the parotid glands of
both adults and children and, in contrast to simple
The differential diagnosis for chronic sialadenitis lymphoepithelial cysts that are solitary and unilateral,
includes other patterns of sialadenitis and other sali- they are commonly multiple and bilateral. The patho-
vary gland lesions containing lymphocytes. genesis of these cysts may be due to duct obstruction,
Autoimmune sialadenitis or lymphoepithelial sialad- lymphoid hyperplasia, or duct destruction related to
enitis (LESA) is most commonly related to Sjögren cell-mediated immunity. FNA produces clear to turbid,
syndrome. The diagnosis is usually made by the unique yellow–brown fluid.
clinical presentation of bilateral enlargement of the Mucocele/retention cysts are common in the sub-
salivary glands (parotid and, in a minority of cases, mandibular and sublingual glands. A mucocele lacks a

Ch001-F06731.indd 4 10/26/2006 10:19:26 AM


CHAPTER 1 Salivary Glands 5

FIGURE 1-5
Chronic sialadenitis. Long-standing
infl ammation and metaplastic
changes may produce cytologic
atypia of the ductal epithelium
represented as nuclear enlarge-
ment, hyperchromasia, and nucleoli.
The isolated nature of the change
should preclude from making a
malignant diagnosis. Air-dried, Diff-
Quik stain, high power.

FIGURE 1-6
Granulomatous sialadenitis. In a
background of lymphocytic infil-
trate, clusters of epithelioid histio-
cytes and a multinucleated giant
cell are the highlights of granulo-
matous sialadenitis. The differen-
tial diagnosis includes infectious,
autoimmune, and, less likely, neo-
plastic diseases. Alcohol-fixed,
Papanicolaou stain, high power.

lining, whereas a retention cyst may have a mixture of mature or anucleated squamous cells in a thick protein-
squamous, columnar and oncocytic epithelial lining. aceous background (Fig. 1-7). Some cases may show
They both result from obstruction associated with squamous metaplastic cells or glandular cells, some cili-
sialolithiasis. ated or mucinous. Normal salivary gland tissue is scant
and degenerated or absent. Cholesterol crystals have
been noted in some cases. FNA of mucus-containing
cysts are usually scantly cellular and contain scattered
CYTOPATHOLOGIC FEATURES histiocytes and inflammatory cells in an abundant
mucinous background. Extracellular mucin may be
recognized as pale-green (Papanicolaou stain) to pink–
The cytologic findings in aspirates of HIV-associated red hyaline material (Romanowsky stain). Occasional
cystic lymphoepithelial lesions include a mixed popula- mucinous/metaplastic cell or normal salivary gland
tion of lymphoid cells, vacuolated and multinucleated elements may be present (Fig. 1-8). A ruptured cyst
histiocytes, lymphohistiocytic aggregates, and scattered can incite a fibroblastic reaction that shows numerous

Ch001-F06731.indd 5 10/26/2006 10:19:27 AM


6 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-7
HIV-associated cystic lymphoepi-
thelial lesion. Numerous small lym-
phocytes, histiocytes, some with
multinucleation, and a thick pro-
teinaceous and granular background
dominate the smears. Scattered
mature or anucleated squamous
cells are also present. Alcohol-fixed,
Papanicolaou stain, medium power.
Courtesy of Ms Jamie L Covell, The
University of Virginia Health Sci-
ences Center.

FIGURE 1-8
Mucocele/retention cyst. Abundant
thick mucoid material contains only
a rare, if any, group of benign epi-
thelial cells. Alcohol-fixed, Papani-
colaou stain, low power.

elongated spindle-shaped cells associated with abun- cellular; however, in scanty aspirates, these tumors
dant thick mucoid material. may be difficult to differentiate from non-neoplastic
conditions based on cytomorphology alone. The correct
diagnosis of a neoplastic lesion relies on the identifica-
tion of the characteristic cell type. Lymphoepithelial
DIFFERENTIAL DIAGNOSIS AND PITFALLS cysts are lymphocyte-rich and a malignant lymphoma
may enter the differential diagnosis. Flow cytometric
analysis will resolve that differential. Lymphangioma
The differential diagnosis for benign non-neoplastic can produce a lymphocyte-rich fluid on aspiration. The
cystic lesions includes both non-neoplastic conditions clinical presentation for this lesion is essential in its
such as LESA and chronic sialadenitis with cyst forma- recognition.
tion, as well as neoplastic diseases such as Warthin Retention cysts/mucoceles yield abundant mucoid
tumor. Typically, aspirates from neoplasms are highly material with very few cells. This pattern can overlap

Ch001-F06731.indd 6 10/26/2006 10:19:29 AM


CHAPTER 1 Salivary Glands 7

with that of an inadequately sampled mucoepidermoid


CYTOPATHOLOGIC FEATURES
carcinoma. A residual mass and the identification of
an occasional cluster of intermediate and squamous
cells may distinguish low-grade mucoepidermoid The cytologic image of benign mixed tumor is varied
carcinoma. and the smear appearance may differ from one FNA
Clinical correlation and follow-up are essential in the pass to another, emphasizing the importance of
management of these lesions. Furthermore, surgical adequate sampling of these tumors. Typically, highly
excision may be recommended to exclude malignancy cellular smears contain variable combinations of bland
in the event of an unresolving residual mass. epithelial cells, fibrillar stroma, and myoepithelial cells
(Figs 1-9 & 1-10). Epithelial cells are cuboidal, usually
arranged in honeycomb sheets and clusters; ducts,
trabeculae, glands, and papillae can be identified (Fig.
TUMORS WITH STROMAL/MESENCHYMAL 1-11). Fragments of chondromyxoid stroma, intensely
DIFFERENTIATION metachromatic in Diff-Quik-stained smears, and pale
blue–green in Papanicolaou-stained smears, are fibril-
lar with frayed and irregular borders and are the clue
to the diagnosis. Spindle or stellate myoepithelial cells
BENIGN MIXED TUMOR are embedded within the stroma. Epithelial-stromal
interdigitation and transition from spindle to epithelial
CLINICAL FEATURES cells are characteristic features of benign mixed tumor.
Single myoepithelial cells may be present in the back-
ground and may be plasmacytoid in appearance. Single,
Benign mixed tumor is the most common salivary clear or epithelioid myoepithelial cells are typically
gland neoplasm and accounts for up to 75% of the difficult to distinguish from single epithelial cells.
neoplasms in some series. They are benign, slow- Minor variations may occur in a tumor with the
growing tumors that occur predominantly in the super- typical background and include isolated cytologic atypia,
ficial lobe of the parotid (90%), involving the tail or the mucinous, squamous, and, less frequently, oxyphilic
anterior portion. The submandibular gland and minor and sebaceous metaplasia (Fig. 1-12) of ductal epithe-
salivary glands are less frequently affected. Benign lium, and variability in the stromal cellularity and
mixed tumor occurs in all age groups, including chil- appearance from myxoid to fibrocollagenous, chondroid,
dren, but are seen more frequently in women (M : F = or osteoid. Crystal and crystalloid deposition and intra-
1 : 4) in their fourth and fi fth decades. Benign mixed nuclear inclusions may be seen. Occasionally, mixed
tumors are treated with superficial parotidectomy with tumor may harbor a cystic component (7%). The cystic
a good margin of normal salivary gland, since enucle-
ation may leave small tumor extensions behind and
may lead to recurrences (0–4%). Recurrences are slow
growing, require a lengthy follow-up, and may be diffi- BENIGN MIXED TUMOR – PATHOLOGIC FEATURES
cult to eradicate. With deep-seated tumors, multifocal
tumors, and scarring, the facial nerve may be compro- Cytopathologic Findings
mised, resulting in injuries. If not excised, benign ៉ Interdigitating ductal epithelial cells, some with metaplastic
mixed tumors may reach an enormous size and poten- changes
tially undergo malignant transformation. ៉ Myoepithelial cells
៉ Chondromyxoid stroma

Histopathologic Findings
៉ Gross: rubbery, bosselated, well-circumscribed mass with small
BENIGN MIXED TUMOR – DISEASE FACT SHEET extensions into the surrounding normal tissue
៉ Microscopic: biphasic appearance with varied epithelial and
Incidence and Location stromal components in variable proportions
៉ Most common salivary gland neoplasm (75%)
៉ Parotid (90%) > submandibular > minor salivary glands Ancillary Studies
៉ Immunoreactivity with glial fibrillary acidic protein (GFAP) is
Gender and Age Distribution useful in differentiating benign mixed tumor from adenoid
៉ Male to female ratio is 1 : 4 cystic carcinoma and basal cell adenoma
៉ Age: 40 years
Differential Diagnosis and Pitfalls
Prognosis and Treatment ៉ Cylindromatous areas: adenoid cystic carcinoma
៉ Slow growing, may reach enormous size if not removed ៉ Scant stroma: basal cell adenoma
៉ Potential for malignant transformation if not excised ៉ Squamous and/or mucinous metaplasia: mucoepidermoid
៉ Surgical excision with good margins carcinoma
៉ Enucleation associated with high recurrence rate ៉ Cytologic atypia: carcinoma ex-pleomorphic adenoma

Ch001-F06731.indd 7 10/26/2006 10:19:30 AM


8 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-9
Benign mixed tumor. Cellular smears
show the characteristic biphasic
cellular component. Epithelial and
stromal elements interdigitate with
each other. The stroma is pale blue–
green and has irregular edges and
a fibrillar nature. Courtesy of Ms
Jamie L Covell, The University of
Virginia Health Sciences Center.
Alcohol-fixed, Papanicolaou stain,
low power.

FIGURE 1-10
Benign mixed tumor. The stroma
appears metachromatic and can
vary from dense to more diffuse in
the smear background. A number of
single epithelial and myoepithelial
cells are typically present and are
not a sign of malignancy. Air-dried,
Diff-Quik stain, low power.

proteinaceous background is thin and should not be tumors have a distinctive low-power pattern composed
misinterpreted as mucin. predominantly of loosely cohesive branching and arbo-
rizing groups of cells having indistinct community
borders. Frequently, the stroma associated with cellular
mixed tumor is fibrocollagenous, dense, and courses
DIFFERENTIAL DIAGNOSIS AND PITFALLS within the branching epithelial clusters (Fig. 1-13). The
cells have scant cytoplasm, oval to round nuclei with
finely granular chromatin, and lack distinct nucleoli.
Cellular mixed tumors account for about 30% of mixed Occasional spindling of the cells is observed (Fig. 1-14).
tumors and FNA may sample the predominant epithe- Nuclear overlap and haphazard arrangement is a
lial/myoepithelial component but not the scant stromal/ common finding. Scattered plasmacytoid single cells
mesenchymal component, leading to misclassifying with moderate amount of cytoplasm are seen in most
the tumor as basal cell adenoma. These cellular mixed cases.

Ch001-F06731.indd 8 10/26/2006 10:19:31 AM


CHAPTER 1 Salivary Glands 9

FIGURE 1-11
Benign mixed tumor. Bland epithe-
lial cells are embedded into the
fibrillar mesenchymal stroma and
there is gradual transition into
spindle and stellate mesenchymal
cells that also populate the stroma.
Alcohol-fixed, Papanicolaou stain,
high power.

FIGURE 1-12
Benign mixed tumor. Sebaceous
metaplasia is rare in salivary gland
neoplasms. The metaplastic cells
resemble histiocytes. Round uniform
nuclei are centrally located in an
abundant vacuolated cytoplasm.
The vacuoles are small and punched
out and distend the cytoplasm.
Single myoepithelial cells and a
stromal fragment are present in
the background. Air-dried, Diff-Quik
stain, high power.

Increased cellularity coupled with focal cytologic may mimic the intermediate cells of mucoepidermoid
atypia is encountered in about 20% of tumors and carcinoma. Mucoepidermoid carcinoma is a difficult
should not be considered a sign of malignancy when the diagnosis to make in FNA and requires the identifica-
overall picture is that of benign mixed tumor. A cylin- tion of squamous, intermediate polygonal cells and
dromatous pattern occurs in 5% of mixed tumors and vacuolated mucinous cells, in an acellular mucinous
should not elicit a diagnosis of adenoid cystic carcinoma stringy background.
if occasional (Fig. 1-15); and scattered squamous and Tumors rich in plasmacytoid/hyaline myoepithelial
mucinous metaplasia should not result in a diagnosis cells may also be interpreted as myoepitheliomas, acinic
of mucoepidermoid carcinoma. In some tumors, these cell carcinoma, or oncocytic. Predominance of spindle
metaplastic changes may be present in association with myoepithelial cells elicits the differential diagnosis of
a highly myxoid background mimicking a mucinous spindle cell proliferation including benign reactive pro-
background, which may further confound the situation. cesses such as nodular fasciitis, pseudotumors, periph-
A predominant plasmacytoid/hyaline cell population eral nerve sheath tumors, and spindle cell sarcomas.

Ch001-F06731.indd 9 10/26/2006 10:19:32 AM


10 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-13
Cellular mixed tumor. Branching and
arborizing clusters of epithelial
cells are found amidst numerous
single cells. The stroma appears
dense, fibrocollagenous rather than
the typical fibrillar stroma seen in
benign mixed tumor, and courses
within the epithelial clusters.
Alcohol-fixed, Papanicolaou stain,
medium power.

FIGURE 1-14
Cellular mixed tumor. Single cells in
the background show variability in
shape and size. Some are plasmacy-
toid in appearance, while others are
more spindled, suggesting myo-
epithelial differentiation. The pres-
ence of variable nuclear atypia
including dark but smudgy chroma-
tin is not sufficient for a diagnosis
of carcinoma. Alcohol-fixed, Papani-
colaou stain, high power.

Myoepithelial cells have a characteristic immunoprofile standing, slow-growing mass. The malignant changes
that allows their recognition and helps in the classifica- occur in a background of mixed tumor, and the diagno-
tion of such tumors. sis requires microscopic documentation of previous
benign mixed tumor. The malignant tumor is usually
a high-grade carcinoma, and could show differentiation
toward any of the usual primary carcinomas of the
MALIGNANT MIXED TUMOR salivary gland. The carcinoma is characterized by high
cellularity with cellular groups or sheets, and papillary
clusters of large cells with pleomorphic nuclei, promi-
Carcinoma ex-pleomorphic adenoma is rare and devel- nent nucleoli, and finely vacuolated cytoplasm domi-
ops in up to 10% of cases of benign mixed tumor. nating the smears. Necrosis may be prominent in the
Malignant changes are suggested by sudden rapid background and mitoses are increased (Fig. 1-16). As
growth, pain, and facial paralysis in a patient with a long- opposed to the focal nature of metaplasia and atypia

Ch001-F06731.indd 10 10/26/2006 10:19:34 AM


CHAPTER 1 Salivary Glands 11

FIGURE 1-15
Benign mixed tumor. A focal cylin-
dromatous pattern in this tissue
fragment is characterized by the
small metachromatic globules of
extracellular matrix embedded
within the epithelial group. These
findings overlap with those of basal
cell adenoma and resemble archi-
tectural features of adenoid cystic
carcinoma. The background protein-
aceous material reflects the cystic
nature of this lesion. Air-dried,
Diff-Quik stain, medium power.

A B

FIGURE 1-16
Malignant mixed tumor. A, Superimposed on findings of mixed tumor is a carcinoma with atypical discohesive cells with hyperchromatic pleomorphic
nuclei, coarsely granular chromatin, and a high N/C ratio. Alcohol-fixed, Papanicolaou stain, high power. B, Similar malignant cytologic features
are noted in addition to a mitotic figure. Air-dried, Diff-Quik stain, high power.

Ch001-F06731.indd 11 10/26/2006 10:19:35 AM


12 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 1-17
Carcinosarcoma. A, A malignant epithelial cluster consists of crowded overlapping cells with cytologic atypia. B, Mesenchymal cells with atypical
nuclei are embedded in a chondromyxoid matrix material and establish the presence of a biphasic malignant neoplasm. Alcohol-fixed, Papanicolaou
stain, high power.

associated with cellular mixed tumor, features of malig- ponent is ductal and the mesenchymal component is
nancy are cytologically obvious in malignant mixed chondrosarcomatous. These tumors are highly aggres-
tumor. These tumors have a good prognosis if the sive and rapidly fatal.
changes are confined to the pre-existing mixed tumor.
The prognosis depends on the histologic type, grade,
proliferative index, and extent of invasion beyond the
capsule of the pre-existing tumor. MYOEPITHELIAL NEOPLASMS

CARCINOSARCOMA Myoepithelioma may be considered a variant of pleo-


morphic adenoma and is thought to consist solely of
myoepithelial cells. Myoepithelial cells have three dif-
True malignant mixed tumor is extremely rare and ferent appearances to include plasmacytoid or hyaline
has a biphasic composition of malignant epithelial cells with dense, moderate to abundant, well-
and mesenchymal elements. The aspirate is cellular demarcated cytoplasm, glycogen-rich clear cells, and
and contains large cell clusters and dissociated cells spindle cells. Myoepitheliomas can display a mixture
with marked atypia and a background amorphous sub- of those cell types. Most hyaline cell-type tumors occur
stance (Fig. 1-17). The diagnosis can be established on in minor salivary glands such as in the palate, whereas
cytology only if both sarcomatous and carcinomatous spindle and clear cell myoepitheliomas are primarily
elements are sampled. Frequently, the epithelial com- encountered in the parotid gland. Most hyaline cell

Ch001-F06731.indd 12 10/26/2006 10:19:38 AM


CHAPTER 1 Salivary Glands 13

FIGURE 1-18
Myoepithelioma. A loosely cohesive
sheet and single cells with plasma-
cytoid features have eccentrically
placed nuclei and a moderate
amount of dense well-demarcated
cytoplasm. Mild nuclear variability
is noted. Courtesy of Ms Jamie L
Covell, The University of Virginia
Health Sciences Center. Alcohol-
fixed, Papanicolaou stain, high
power.

myoepitheliomas will behave as benign neoplasms, nucleoli, and demonstrate S-100 positivity characteris-
whereas some spindle and clear cell tumors will present tic of myoepithelial differentiation. Some tumor cell
with malignant infi ltrative features and may have clusters may be surrounded by amorphous hyaline
malignant potential with possible local recurrence, material. Cellular arrangements with acellular basement
metastases, and tumor-related death. membrane material reminiscent of adenoid cystic carci-
Cytologically, myoepitheliomas differ from mixed noma may also be found (Fig. 1-19). These tumors may
tumors by the absence of stroma and epithelial cells and share features with both mixed tumors and basaloid
by the presence of a dominant myoepithelial cell popula- neoplasms, as they consist of basaloid cells and stroma
tion arranged in loose groups and single cells (Fig. 1-18). that forms spherules or has an irregular, somewhat
Hyaline cell myoepithelioma can be distinguished from fibrillar appearance.
plasma cell neoplasm by the nuclear chromatin pattern
and lack of perinuclear Hoff and plasma cell markers
(CD79a and CD138). Spindle cell neoplasms such as
peripheral nerve sheath tumors and low-grade spindle
cell sarcomas should be considered in the differential ONCOCYTIC NEOPLASMS
diagnosis of spindle cell myoepitheliomas. Lastly, clear
cell myoepithelioma enters the differential diagnosis of
other clear cell neoplasms occurring in the salivary WARTHIN TUMOR
glands, such as acinic cell carcinoma, mucoepidermoid
carcinoma, epithelial-myoepithelial carcinoma, and
metastatic renal cell carcinoma. The myoepithelial CLINICAL FEATURES
nature of such tumors can be confirmed by the demon-
stration of staining with one or more of the following:
S-100 protein, smooth muscle actin, calponin, cytokera- Warthin tumors occur almost exclusively in the parotid
tin, and glial fibrillary acidic protein (GFAP). Addition- and are thought to arise from entrapped salivary duct
ally, a myoepithelial-rich mixed tumor should always remnants within intra- or periparotid lymph nodes.
be considered in the evaluation of an aspirate from a They are more common in men in the sixth decade and
myoepithelial-dominant tumor. older. The incidence in women is on the rise, as this
Epithelial-myoepithelial carcinomas are low-grade tumor shows a statistical relationship with smoking.
myoepithelial-rich tumors that are locally aggressive. Warthin tumors are commonly multicentric (10–15%),
They consist of two cell populations: the small dark and bilateral; they constitute 70% of all bilateral sali-
ductal cells arranged in tight clusters and the larger clear vary gland neoplasms. The treatment is surgical exci-
myoepithelial cells with moderate to abundant cyto- sion, and recurrences may occur but are uncommon.
plasm, vesicular nuclei, and conspicuous nucleoli, which Malignant transformation in Warthin tumors is a rare
predominate. The clear cells are large, polygonal, and event, and may occur in either the epithelial or
glycogen-rich, with mildly atypical nuclei and small lymphoid component.

Ch001-F06731.indd 13 10/26/2006 10:19:39 AM


14 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 1-19
Myoepithelial carcinoma. A, Metachromatic stromal fragment with features similar to those seen in benign mixed tumor contains a large number
of basaloid cells. Air-dried, Diff-Quik stain, high power. B, Oval hyperchromatic nuclei with finely granular chromatin and scant cytoplasm with
short tapered cytoplasmic ends suggest a myoepithelial rather than epithelial differentiation of the basaloid cells. Courtesy of Ms Jamie L Covell,
The University of Virginia Health Sciences Center. Alcohol-fixed, Papanicolaou stain, high power.

WARTHIN TUMOR – DISEASE FACT SHEET WARTHIN TUMOR – PATHOLOGIC FEATURES

Incidence and Location Cytopathologic Findings


៉ Second most common salivary gland neoplasm (5–10%) ៉ Mixed population of lymphocytes
៉ Most common bilateral salivary gland neoplasm (70%) ៉ Small sheets of oncocytes
៉ Exclusively in the parotid ៉ Granular proteinaceous background
៉ Multicentric ៉ Metaplastic cells

Gender and Age Distribution Histopathologic Findings


៉ Male to female ratio is 5 : 1 ៉ Gross: lobulated mass, multicystic with fl uid-filled spaces
៉ Age >50 years separated by grayish septae; hemorrhagic infarcts post FNA are
common
Prognosis and Treatment ៉ Microscopic: typically, cystic lesion with prominent lymphoid
៉ Surgical excision with good margins tissue containing frequent germinal centers covered by
៉ Potential for recurrence oncocytes arranged in two layers, resulting in a papillary-like
architecture

Differential Diagnosis and Pitfalls


៉ Cystic lesions: lymphoepithelial cyst, branchial cleft cyst,
papillary cystadenoma
៉ Lesions with lymphoid component: chronic sialadenitis, benign
CYTOPATHOLOGIC FEATURES
lymphoepithelial cyst, lymph node, lymphoma, acinic cell
carcinoma
៉ Oncocytic neoplasms
Characteristically, Warthin tumors are multicystic and
៉ Intraductal papilloma
yield a turbid brown fluid. Microscopically, a classic ៉ Other neoplasm such as squamous carcinoma or mucoepidermoid
dirty proteinaceous background (Fig. 1-20), and a carcinoma in association with epithelial metaplasia
mixed but maturing population of lymphocytes and ៉ Infarct-like necrosis: malignant neoplasms
scattered groups of oncocytes characterize these tumors
(Fig. 1-21). The lymphoid population is comprised pre-
dominantly of small lymphocytes and fewer larger cells
showing an ordered maturational sequence and por-
tions of germinal centers. A variable amount of uniform groups. Occasionally, pseudopapillary arrangements
oncocytic epithelium is present in small sheets without may be present, consisting of two oncocytic cell layers
crowding. The oncocytes have abundant granular cyto- surrounding lymphoid aggregates.
plasm, round uniform nuclei, even chromatin, and con- Cytologic variants may include the presence of
spicuous nucleoli (Fig. 1-22). Frequently, the two cell focal squamous or mucinous metaplasia similar to that
populations are intermixed within the same cellular seen in benign mixed tumor. Granulomas formed by

Ch001-F06731.indd 14 10/26/2006 10:19:40 AM


CHAPTER 1 Salivary Glands 15

FIGURE 1-20
Warthin tumor. Large pools of
thick proteinaceous material are
easily identified among numerous
lymphocytes and scattered small
epithelium groups. The protein-
aceous material, which may have
some similarity to mucin pools, cor-
responds to the aspirated cyst
fluid. Air-dried, Diff-Quik stain,
high power.

FIGURE 1-21
Warthin tumor. Small fl at sheets of
oncocytes and lymphocytes in all
stages of maturation are the two
cell populations characteristic of
this tumor. Alcohol-fixed, Papanico-
laou stain, medium power.

both epithelioid histiocytes and multinucleated giant DIFFERENTIAL DIAGNOSIS AND PITFALLS
cells (sarcoid type) can be seen in some cases. The
pathogenesis of granulomas is unknown, and could be
related to a toxic effect of the cyst content. Non-tyrosine Potential sources of diagnostic pitfalls in Warthin
crystalloids similar to those seen in sialolithiasis and tumors may be caused by the presence of dominant
sialadenitis, as well as corpora amylacea have been morphologic features or atypical features. Such factors
described in association with Warthin tumors, and are may involve the epithelial component or the lymphoid
found in highest concentration in cystic and luminal component. A scanty lymphoid component with a pre-
spaces. dominance of oncocytic cells may suggest a neoplasm

Ch001-F06731.indd 15 10/26/2006 10:19:41 AM


16 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-22
Warthin tumor. Uniform oncocytes
are orderly arranged in this sheet
and appear to line up at the edge,
suggesting a papillary configura-
tion. The cells have the character-
istic round nuclei with conspicuous
nucleoli and abundant dense but
granular cytoplasm. Air-dried, Diff-
Quik stain, high power.

such as oncocytoma or a bland oncocytic carcinoma. In show vacuolization and granularity and will be more
oncocytic neoplasms, oncocytes are organized in large delicate than the cytoplasm of oncocytes. Architectur-
sheets rather than in the small clusters seen in Warthin ally, attempt at acinar formation can be found in acinic
tumors; there is also a lack of the background protein- cell carcinoma, whereas acini or glandular aggregates
aceous material. Intraductal papilloma of the salivary are lacking in Warthin tumors.
gland is another cystic neoplasm that may have a pre- Patients with Warthin tumors are at increased risk
dominance of oncocytic epithelium. In these rare cases, of developing malignant lymphoma. When malignant
the oncocytes are arranged in a striking papillary con- lymphoma is suspected, clonality of the cytologic
figuration with a total absence of lymphocytes in the material can be proven by immunocytochemistry, flow
background. cytometry, or molecular studies to demonstrate gene
Squamous metaplasia can occur in Warthin tumors. rearrangement.
Such metaplastic changes have been reported in patients
with a previous history of FNA and in patients with
spontaneous infarction of a Warthin tumor. Infarcted
Warthin tumors may yield necrotic material that may ONCOCYTOMA AND ONCOCYTIC
suggest a malignant neoplasm. CARCINOMA
Also included in the differential diagnosis are lesions
of the salivary glands with lymphoid component. Benign
lymphoepithelial cysts overlap with Warthin tumors. Oncocytoma is a rare benign neoplasm that involves
The lymphoid population is similar in both lesions. mainly the parotid gland in patients over 70 years of
Separating the two entities requires focusing on the age. Oncocytomas produce a well-defined, solid, par-
epithelial component, and identifying oncocytes for tially encapsulated mass and yield a pure population of
Warthin tumor, or occasional mature and parakeratotic oncocytes by FNA. Oncocytes have abundant dense
squamous cells or columnar cell ghosts for lymphoepi- granular cytoplasm, central round nuclei, and conspic-
thelial cysts. Variants of chronic sialadenitis or benign uous nucleoli. They occur in large sheets in a clean
lymphoepithelial lesions may mimic Warthin tumor but background devoid of lymphocytes and proteinaceous
are typically characterized by an intense inflammatory material. The differential diagnosis of oncocytoma is
response and a different mix of epithelial cells, includ- oncocytosis and Warthin tumor. The malignant coun-
ing ductal and some acinar epithelium. Lymph nodes terpart of oncocytoma is oncocytic carcinoma, which
present within salivary glands constitute another diag- may not always be easy to diagnose based on cytologic
nostic pitfall, but may be easily recognized when an features alone. Oncocytic carcinoma is a diagnosis of
almost complete absence of an epithelial component is exclusion after ruling out oncocytic variants of other
noted. Acinic cell carcinoma with lymphoid component more common tumors such as mucoepidermoid carci-
will also enter the differential diagnosis. Acinic cell car- noma. Oncocytic carcinoma may be suspected in cyto-
cinoma will typically have a more abundant epithelial logic preparations in the presence of atypia, mitosis,
component. The cytoplasm in acinic cell carcinoma will and necrosis.

Ch001-F06731.indd 16 10/26/2006 10:19:42 AM


CHAPTER 1 Salivary Glands 17

BASALOID NEOPLASMS BASAL CELL ADENOMA – DISEASE FACT SHEET

Location
៉ Parotid
This section addresses a group of salivary gland neo-
plasms that share cytologic features and cellular compo-
Gender and Age Distribution
sition, therefore resulting in overlapping findings on
៉ Slight predilection for females
FNA and creating diagnostic dilemmas and pitfalls.
៉ Age >50 years
These tumors consist of epithelial and myoepithelial
cells with bland nuclear features and scant cytoplasm,
Clinical Features
giving them a basaloid appearance. They also have vari-
៉ Presentation similar to benign mixed tumor
able amounts of extracellular matrix that may be hyalin-
ized, forming globules and cylinders enrobed within the
Prognosis and Treatment
cells. This group of tumors includes basal cell adenoma,
៉ Surgical excision with good margins
basal cell adenocarcinoma, adenoid cystic carcinoma,
៉ Potential for malignant transformation to adenoid cystic
polymorphous low-grade adenocarcinoma, and some carcinoma or basal cell adenocarcinoma
myoepithelial neoplasms. As a result of their shared
features, they also share their differential diagnoses.

BASAL CELL ADENOMA CYTOPATHOLOGIC FEATURES

CLINICAL FEATURES Smears are moderately cellular and consist of variably


sized, dark-appearing, densely packed clusters of mono-
morphic basaloid cells. Some clusters may display irreg-
Basal cell adenomas are tumors of the parotid gland ular branching and trabecular patterns. Peripheral
that have a slight female predilection. Their clinical palisading of basaloid cells may be noted. A variable
presentation is similar to that of benign mixed tumors, number of single cells and naked nuclei are present in
although they tend to occur in an older age group. the background (Fig. 1-23). The basaloid cells are
Specific subtypes have been recognized, such as the uniform with round nuclei, scant cytoplasm, well-
membranous type for its more aggressive clinical course defined cell borders, and a high nuclear to cytoplasmic
and association with skin tumors (cylindromas), and (N/C) ratio. The chromatin is bland and nucleoli are
the canalicular type for its location in the upper lip. inconspicuous. Single basaloid cells lack the character-
Treatment of these tumors is by complete excision. istic plasmacytoid appearance of single myoepithelial

FIGURE 1-23
Basal cell adenoma. Single basaloid
cells with bland uniform oval nuclei
and scanty well-defined cytoplasm
lack the plasmacytoid appearance
of myoepithelial cells of benign
mixed tumor. Some cells are
arranged in a pseudoacinar forma-
tion surrounding the extracellular
matrix. Courtesy of Ms Jamie L
Covell, The University of Virginia
Health Sciences Center. Air-dried,
Diff-Quik stain, high power.

Ch001-F06731.indd 17 10/26/2006 10:19:43 AM


18 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-24
Basal cell adenoma. Cluster of basa-
loid cells containing small uniform
metachromatic stromal globules of
extracellular matrix. The globule
edges are poorly defined and
interdigitate with the surrounding
cells. Air-dried, Diff-Quik stain,
high power.

identification by FNA. Solid nests and tubular struc-


BASAL CELL ADENOMA – PATHOLOGIC FEATURES tures formed by basaloid cells are surrounded by a
thick ribbon of hyalinized basement membrane-
Cytopathologic Findings like material. The neoplastic cells show peripheral
៉ Monomorphic bland basaloid cells palisading at the periphery of the clusters (Fig. 1-25).
៉ Variably sized clusters, branching and trabecular pattern Scattered hyaline globules can be seen within the cell
៉ Peripheral palisading groups.
៉ Single cells and bare nuclei
៉ Small hyaline globule and cylinders of extracellular matrix
៉ Thick ribbon of extracellular matrix around nests and tubules
(membranous type)
៉ Poorly defined stromal–epithelial interface DIFFERENTIAL DIAGNOSIS AND PITFALLS
Histopathologic Findings
៉ Gross: encapsulated, often cystic See page 22.
៉ Microscopic:
៉ Tubular, trabecular, or solid
៉ Peripheral palisading of cells at the edge of epithelial nests
៉ Abundant extracellular matrix at periphery and within nests
BASAL CELL ADENOCARCINOMA
(cylindromatous pattern) in membranous type (unclear)

Basal cell adenocarcinoma is a rare, recently recog-


nized entity. It affects patients in the sixth decade of
life and occurs predominantly in the parotid. It is cyto-
cells seen in pleomorphic adenoma. Within the clus- logically similar to basal cell adenoma. The presence of
ters, metachromatic/clear (Romanowsky/Papanicolaou mitoses and nuclear atypia including prominent nucle-
stains) stromal hyaline globules and cylinders sur- oli may point to the malignant nature of the tumor.
rounded by basaloid cells are easily identified. Typi- However, the malignant nature of the process may be
cally, the globules are small and the stromal edge is difficult to ascertain, as it is impossible to determine
poorly defined and interdigitates with the surrounding from FNA material the invasive nature of the tumor.
cells (Fig. 1-24). Irregularly shaped stromal fragments Invasion is primarily a histologic feature that sets it
may be focally encountered within or at the periphery apart from basal cell adenoma. Basal cell adenocarci-
of the clusters and represent the fibrous supporting noma is associated with perineural invasion and tends
stroma. The fibrillar stroma of benign mixed tumor is to overexpress p53, bcl2, and EGFR. Its clinical course
absent. may include local recurrences, and lymph node and
The cytologic findings in the membranous type of distant metastases, primarily to the lung. For the dif-
basal cell adenoma are distinctive and allow for its ferential diagnosis, see page 22.

Ch001-F06731.indd 18 10/26/2006 10:19:44 AM


CHAPTER 1 Salivary Glands 19

FIGURE 1-25
Basal cell adenoma. Basaloid cells
show peripheral palisading at the
edge of this large solid cluster of
monomorphic cells. A ribbon of
stroma is noted surrounding the
cluster, a feature that suggests a
membranous pattern of basal cell
adenoma. Alcohol-fixed, Papanico-
laou stain, medium power.

ADENOID CYSTIC CARCINOMA achieve. Radiation therapy is not curative but may
produce temporary regression in unresectable cases.
Prognostic factors include tumor grade and stage, status
CLINICAL FEATURES of surgical margins, anatomic site, and tumor size.
Adenoid cystic carcinoma has a high recurrence rate
even in well-differentiated tumors and is associated
Adenoid cystic carcinoma is rare, and occurs with less with frequent lung and bone metastases. It has a pro-
frequency than mucoepidermoid and acinic cell carci- tracted course and a low 15–20-year survival.
noma in the parotid gland. It is, however, the most
common malignancy in minor salivary glands. Adenoid
cystic carcinoma occurs more frequently in women in
the fi fth to sixth decade and has a slow, relentless CYTOPATHOLOGIC FEATURES
growth. It may present with a painful mass or facial
nerve paralysis. Radical resection is the treatment of
choice, and cure following recurrence is difficult to Well-differentiated tumors are composed of small
basaloid cells, both ductal and myoepithelial, in solid
ADENOID CYSTIC CARCINOMA – DISEASE FACT SHEET

ADENOID CYSTIC CARCINOMA – PATHOLOGIC FEATURES


Incidence
៉ Rare in major salivary glands
Cytopathologic Findings
៉ Most common malignancy in minor salivary gland
៉ Small basaloid cells
៉ Solid clusters, spherules, cylinders
Gender and Age Distribution
៉ Hyalinized, acellular, large stromal fragments
៉ More frequent in women in the 5th and 6th decade ៉ Well-defined stromal edges
៉ Sharp stromal–epithelial interface
Clinical Features
៉ Slow relentless growth Histopathologic Findings
៉ Pain and facial nerve paralysis ៉ Gross: solid mass, infiltrative borders
៉ Microscopic:
Prognosis and Treatment ៉ Cribriform nests with pseudocysts
៉ Radical surgical excision with clear margins ៉ Tubular and solid patterns
៉ High recurrence rate and metastases to lung and bone ៉ Perineural invasion
៉ Low survival rate ៉ Strong staining with CD117 (c-kit protein)

Ch001-F06731.indd 19 10/26/2006 10:19:45 AM


20 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-26
Adenoid cystic carcinoma. Large
variably sized metachromatic stro-
mal globules and small clusters of
bland basaloid cells are the classic
finding. Some of the stromal glob-
ules are rimmed by a row of basaloid
cells that are barely touching the
stromal fragment. Note the size dif-
ference of the stromal globules and
the lack of cell/stroma interdigita-
tion in comparison with basal cell
adenoma. Air-dried, Diff-Quik stain,
medium power.

FIGURE 1-27
Adenoid cystic carcinoma. The cells
are uniform with a high N/C ratio,
round to oval nuclei, and an occa-
sional angulated nuclear membrane.
Chromatin is fine and even, and
nucleoli are small but conspicuous.
ThinPrep, Papanicolaou stain, oil.

clusters, spherules, and cylinders, or in cribriform or cylindrical stromal fragments lying bare or sur-
groups (Fig. 1-26). The cells are uniform with round to rounded by a single discontinuous rim of basaloid
oval, sometimes angulated nuclei, scant cytoplasm, and cells that seem to hardly touch the stromal surface.
a high N/C ratio. The chromatin varies from fine to Conversely, the poorly differentiated or solid form of
coarse and nucleoli are small but conspicuous (Fig. adenoid cystic carcinoma will have few stromal glob-
1-27). The basaloid cells surround intensely metachrom- ules, a predominance of basaloid cells, and more promi-
atic (Romanowsky stain) or pale and glassy (Papanico- nent nuclear atypia (Fig. 1-29). Special stains and
laou stain) stromal spheres and cylinders. The stromal immunocytochemistry do not provide significant dif-
fragments are larger than those seen in monomorphic ferentiating features, except for CD117 (c-kit), which is
adenoma; they are variably sized, acellular, and sharply strongly immunoreactive in adenoid cystic carcinoma
demarcated with smooth rounded edges (Fig. 1-28). In in comparison with other basaloid tumors and may be
some cases, the predominant finding is that of spherical of diagnostic consideration.

Ch001-F06731.indd 20 10/26/2006 10:19:46 AM


CHAPTER 1 Salivary Glands 21

DIFFERENTIAL DIAGNOSIS AND PITFALLS

See page 22.

POLYMORPHOUS LOW-GRADE
ADENOCARCINOMA

Polymorphous low-grade adenocarcinoma is a neoplasm


of minor salivary glands. It has rarely been reported in
the parotid gland and affects women more often than
men. Its most common location is the palate, where it
follows adenoid cystic carcinoma in frequency. Smears
are characterized by the presence of tubules, cords,
linear groupings, and branching papillary sheets and

POLYMORPHOUS LOW-GRADE ADENOCARCINOMA –


DISEASE FACT SHEET

Incidence and Location


៉ Second most common salivary gland neoplasm in palate
៉ Minor salivary glands, palate

Gender and Age Distribution


៉ More common in females
៉ Age: adult

Prognosis and Treatment


FIGURE 1-28 ៉ Comprehensive surgical excision with clear margins
Adenoid cystic carcinoma. A bare stromal fragment may be the predominant ៉ Potential for recurrence and lymph node metastases
finding. Note the dense hyalinized appearance of the stroma and the sharply ៉ No distant metastases
demarcated contour. Globules, as well as cylindrical and more complex
structures, are common. Air-dried, Diff-Quik stain, medium power.

FIGURE 1-29
Adenoid cystic carcinoma. Poorly
differentiated tumors consist of
densely packed basaloid cells with
more obvious nuclear atypia and a
scant stromal component. The lack
of nuclear molding and neuroendo-
crine chromatin pattern helps dif-
ferentiate this tumor from a small
cell undifferentiated carcinoma.
Air-dried, Diff-Quik stain, medium
power.

Ch001-F06731.indd 21 10/26/2006 10:19:47 AM


22 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-30
Polymorphous low-grade adenocar-
cinoma. Large tissue fragments
composed of monotonous uniform
cells display a branching papillary
architecture, one of the patterns
encountered in this tumor. Air-
dried, Diff-Quik stain, low power.

FIGURE 1-31
Polymorphous low-grade adenocar-
cinoma. Within the sheet of uniform
basaloid-appearing cells are small
stromal globules that bear resem-
blance to those seen in basal cell
adenoma and adenoid cystic carci-
noma and help differentiate this
lesion from other papillary neo-
plasms. Air-dried, Diff-Quik stain,
high power.

clusters (Fig. 1-30). Hyaline globules are identified DIFFERENTIAL DIAGNOSIS AND PITFALLS
within the sheets of basaloid epithelium. Cytologic
uniformity or monomorphism is key; the tumor is com-
posed of basaloid, bland, uniform cells with round to Tumors comprised of basaloid cells, such as basal cell
oval nuclei, dispersed chromatin, inconspicuous or adenoma, adenoid cystic carcinoma, polymorphous
absent nucleoli, and scant cytoplasm (Fig. 1-31). Mild low-grade adenocarcinoma, and basal cell adenocarci-
nuclear variability can be seen, and mitoses are absent. noma, share cytologic features, and therefore all enter
Treatment is by surgical excision with clean margins, the same differential diagnosis. Distinction between
as recurrences are difficult to eradicate. Like adenoid basal cell adenoma and cellular mixed tumor may be
cystic carcinoma, this tumor has a predilection for difficult, but is of no significant consequence since both
perineural invasion; however, it typically is a favor- are benign tumors that are treated in a similar fashion.
able actor, with local recurrences and regional lymph Cells of basal cell adenoma show a uniform orderly cel-
nodes metastases described, but overall absent distant lular arrangement with no loss of polarity, as opposed
metastases. to the haphazard arrangement in cellular pleomorphic

Ch001-F06731.indd 22 10/26/2006 10:19:49 AM


CHAPTER 1 Salivary Glands 23

logic grounds, and with a high degree of certainty, the


POLYMORPHOUS LOW-GRADE ADENOCARCINOMA – benign from the malignant neoplasms. In the individual
PATHOLOGIC FEATURES case, it is important to recognize the limitation of the
technique and classify the difficult lesions as ‘salivary
Cytopathologic Findings gland neoplasm’, listing a differential diagnosis, and
៉ Monomorphous bland basaloid cells deferring classification to histologic examination.
៉ Tubules, cords, linear groupings, and branching papillary sheets
៉ Hyaline globules within sheets of epithelial cells

Histopathologic Findings
៉ Uniformity of cell type MUCOEPIDERMOID CARCINOMA
៉ Architectural variability: tubular, cribriform, papillary, solid,
and fascicular
៉ Infiltrative border
CLINICAL FEATURES
៉ Perineural invasion

Differential Diagnosis and Pitfalls (for all basaloid neoplasms) Mucoepidermoid carcinoma comprises 5–10% of all
៉ Basal cell adenoma salivary gland neoplasms. It is the second most common
៉ Cellular pleomorphic adenoma primary neoplasm of the salivary glands in adults
៉ Adenoid cystic carcinoma (30%), and the most common primary malignancy in
៉ Polymorphous low-grade adenocarcinoma
children. It is mostly located in the parotid, but may
៉ Basal cell adenocarcinoma
៉ Myoepithelial carcinoma
also involve other major and minor salivary glands.
៉ Metastatic basal cell carcinoma In adults, these tumors typically occur in the third to
fifth decades. Mucoepidermoid carcinomas are divided
according to grade and are most often classified as
low-grade and high-grade tumors, although some clas-
sification schemes include an intermediate grade. Their
adenoma. The individual cells in basal cell adenoma presentation, treatment, and prognosis are dependent
have a small rim of cytoplasm and lack the plasma- on grade. Low-grade tumors tend to develop slowly over
cytoid appearance of myoepithelial cells seen in benign a period of years. They may recur locally, but distant
mixed tumor. metastases are rare. The 5-year survival for low-grade
Basal adenoma closely resembles adenoid cystic carci- tumors approaches 98%. The tumors tend to be well
noma and may be difficult to distinguish from it cytologi- circumscribed and may become cystic, containing
cally in the individual case. The unique appearance of the mucinous material. These tumors are treated and may
cell–stroma interface is useful in distinguishing between be cured with limited excision. High-grade tumors
basal adenomas and adenoid cystic carcinoma. In basal grow rapidly, recur locally, and metastasize to regional
cell adenoma the collagenous stroma interdigitates with lymph nodes, lung, and bone. Their 5-year survival rate
the adjacent cells, whereas in adenoid cystic carcinoma is around 56%. High-grade tumors are infi ltrative and
there is a sharp demarcation of cells and stroma. The tend to be solid. They are treated aggressively with
spherules in basal cell adenoma are small and somewhat surgery, and possible lymph node dissection and
uniform in size, whereas in adenoid cystic carcinoma radiation therapy. Most of these tumors manifest their
they are large and variable in size and shape. The stroma poor behavior within the first year after surgery.
of basal adenoma may contain some spindle cells and
capillaries, whereas the stroma of adenoid cystic carci-
noma is acellular. The stroma of solid adenoid cystic
carcinoma may mimic that seen in cellular mixed tumor.
MUCOEPIDERMOID CARCINOMA – DISEASE FACT SHEET
It, however, represents a desmoplastic stroma. Any
abnormality of the chromatin and the presence of promi-
Incidence and Location
nent nucleoli favor a diagnosis of adenoid cystic carci-
៉ Most common salivary gland malignancy in children and adults
noma over basal cell adenoma. The solid variant of
៉ Predominantly parotid
adenoid cystic carcinoma may also resemble small cell
undifferentiated carcinoma or basaloid squamous carci- Age Distribution
noma. Basal cell adenocarcinoma, as previously stated,
៉ 20–40 years
cannot be differentiated cytologically from basal cell
adenoma since presence of invasion is the determining Clinical Features
factor. Polymorphous low-grade adenocarcinoma may
៉ Slow-growing mass for low-grade tumors, rapidly growing for
be suspected because of its location and the detection high-grade tumors
of architectural patterns that are not characteristic of
adenoid cystic carcinoma, such as papillary features. Treatment
Although less common, other tumors such as myoepithe- ៉ Wide local excision, regional node dissection if nodes clinically
lial carcinoma and metastatic basal cell carcinoma should involved
also be considered in the differential diagnosis. The major
dilemma in this group of tumors is to separate, on cyto-

Ch001-F06731.indd 23 10/26/2006 10:19:50 AM


24 FINE NEEDLE ASPIRATION CYTOLOGY

Extraglandular extension, vascular invasion, necrosis,


and high mitotic rate are poor prognostic features. MUCOEPIDERMOID CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Variable combination of mucous, intermediate, squamous, and
CYTOPATHOLOGIC FEATURES clear cells
៉ Low grade: abundant mucoid background with predominance of
bland mucous and intermediate cells
The FNA diagnosis of mucoepidermoid carcinoma is ៉ High grade: predominance of squamous and intermediate cells,
one of pattern recognition. Aspirates from these tumors sparse mucous cells, overtly malignant features
contain a variety of cell types: mucin-producing, squa-
mous, intermediate, and clear. Intermediate cells are Histopathologic Findings
small and bear resemblance to squamous metaplasia. ៉ Gross:
In low-grade mucoepidermoid carcinoma, extracellular ៉ Low grade: well circumscribed, cystic
stringy mucin is seen in the smear background of a ៉ High grade: solid, infiltrative growth pattern
៉ Microscopic:
majority of cases. The cellular components are quanti-
៉ Low grade: cysts containing mucinous material, lined by well-
tatively variable and include epithelial cell groups that
differentiated mucinous cells
are multilayered with overlapping nuclei, mucin- ៉ High grade: solid, infiltrative sheets of squamous intermediate
containing cells, intermediate cells without identifiable and clear cells with scattered mucin-producing cells
keratin, squamous cells, and finely vacuolated clear
cells. In cellular clusters, a gradual transition from Differential Diagnosis and Pitfalls
intermediate to mucus-producing cells is readily iden- ៉ Low grade: mucocele, Warthin tumor with squamous/mucinous
tifiable (Fig. 1-32). The nuclei of these cells are gener- metaplasia, pleomorphic adenoma with squamous/mucinous
ally small, uniform, round or oval, and not overtly metaplasia
malignant. Mucin-producing cells typically predomi- ៉ High grade: salivary duct carcinoma, carcinoma ex-pleomorphic
nate. They may have a columnar or signet-ring appear- adenoma, adenosquamous carcinoma, oncocytic carcinoma,
ance or may resemble histiocytes when present singly. metastatic squamous cell carcinoma
Aspirates from high-grade tumors demonstrate poorly
differentiated cells with frankly malignant features
including pleomorphic nuclei, hyperchromasia, coarse
chromatin, and prominent nucleoli; glandular/mucous
cells may be few in number and difficult to recognize,
requiring a special stain for mucin for identification DIFFERENTIAL DIAGNOSIS AND PITFALLS
(Fig. 1-33). The squamous cells are more easily recog-
nized and, together with intermediate cells, predomi-
nate. Predominant bizarre forms of squamous cells, Low-grade mucoepidermoid carcinoma is commonly
pearl formation, extensive necrosis, and frequent cystic, and may be difficult to diagnose by FNA. Ade-
mitoses are not classic features of high-grade muco- quate sampling of these tumors is critical to accuracy
epidermoid carcinoma. in interpretation.

A B

FIGURE 1-32
Mucoepidermoid carcinoma, low grade. A, Intermediate and mucin-producing cells are mingled within the same cluster. Intermediate cells, centrally
located in the cluster, are small with round uniform nuclei and moderate to scant cytoplasm, whereas mucinous cells have similar nuclear features
but ample vacuolated mucin-containing cytoplasm and are noted at the periphery of the cluster. Cells are multilayered and overlapping, a clue to
their neoplastic nature, and display a gradual transition from intermediate to mucous cells. Wisps of mucinous material are present in the back-
ground. Alcohol-fixed, Papanicolaou stain, medium power. B, A sheet of streaming, elongated cells with moderate amount of cytoplasm suggests
squamous differentiation. Alcohol-fixed, Papanicolaou stain, medium power.

Ch001-F06731.indd 24 10/26/2006 10:19:50 AM


CHAPTER 1 Salivary Glands 25

FIGURE 1-33
Mucoepidermoid carcinoma, high
grade. Nuclear pleomorphism, hy-
perchromasia, nucleoli, cellular
crowding, and discohesion are easily
recognizable features of malignancy.
An occasional mucin-producing cell
is identified within the cluster.
Alcohol-fixed, Papanicolaou stain,
medium power.

Aspiration of these tumors may yield mucoid cyst glandular differentiation may be scattered mucin vacu-
fluid containing inflammatory cells, histiocytes (which oles in sheets of intermediate and squamous cells. Sali-
may mimic glandular cells), and few epithelial cells. vary duct carcinoma has extensive necrosis and may
This presentation must be distinguished from a mucous have a papillary architecture. The presence of extensive
retention cyst or mucocele, which is typically less cel- keratinization and prominent pearl formation favors
lular and lacks the intermediate or squamous cell com- the diagnosis of squamous cell carcinoma over muco-
ponent seen in low-grade mucoepidermoid carcinoma. epidermoid carcinoma. Finally, the clinical history is
Another clue in separating a benign cyst from a neo- helpful in excluding a metastasis or identifying a pre-
plasm is the persistence in neoplastic processes of a existing mass such as in carcinoma ex-pleomorphic
mass post FNA. Squamous and mucinous metaplasia adenoma. In the final evaluation, all primary high-grade
accompanied by inspissated mucus or thick protein- carcinomas of the salivary gland are treated similarly
aceous fluid can be seen in chronic sialadenitis, sialo- and a definitive classification is not essential for appro-
lithiasis, and Warthin tumor, and may mimic the cells priate therapy.
seen in low-grade mucoepidermoid carcinoma. These
cells are usually found focally and do not constitute the
predominant cell population. Rarely, mucoepidermoid
carcinoma may present with oncocytic features that
renders its distinction from Warthin tumor or onco- ACINIC CELL CARCINOMA
cytoma problematic.
The thick mucoid material in the smear background
of low-grade mucoepidermoid carcinoma may be meta- CLINICAL FEATURES
chromatic with Diff-Quik staining and resemble the
myxoid matrix characteristic of benign mixed tumor.
This material, however, lacks the spindle cells seen in Acinic cell carcinoma is an uncommon neoplasm, com-
the stroma of mixed tumors. prising only 1–6% of all salivary gland neoplasms.
High-grade mucoepidermoid carcinoma shows However, it is the second most common primary sali-
obvious cytologic features of malignancy and can mimic vary gland malignancy, occurs mostly in the parotid
other poorly differentiated primary or metastatic tumors gland, and can be bilateral in approximately 3% of
such as carcinoma ex-pleomorphic adenoma, salivary cases. It is most frequently encountered in the third to
duct carcinoma, adenosquamous carcinoma, and meta- fourth decade, and shows prevalence in women. Acinic
static squamous cell carcinoma. The correct diagnosis cell carcinoma forms a well-circumscribed, mobile,
of mucoepidermoid carcinoma relies on the identifica- slowly growing mass, and although it appears as a low-
tion of both squamous and glandular components. In grade malignancy, it often demonstrates a protracted
high-grade tumors, squamous differentiation is more course and has a propensity for local recurrence and
easily recognized, but the mucous glandular cells are regional and distant metastases. Treatment requires
few in number or difficult to identify without the use wide local excision with regional node dissection if
of special stains for mucin. The only indication of necessitated by the presence of clinically involved

Ch001-F06731.indd 25 10/26/2006 10:19:51 AM


26 FINE NEEDLE ASPIRATION CYTOLOGY

ACINIC CELL CARCINOMA – DISEASE FACT SHEET ACINIC CELL CARCINOMA – PATHOLOGIC FEATURES

Incidence and Location Cytopathologic Findings


៉ Uncommon salivary gland neoplasm (1–6%) ៉ Sheets and tissue fragments with loose acinar formation
៉ Predominantly parotid ៉ Polygonal cells with bland nuclei and abundant granular/
vacuolated cytoplasm, indistinct cell border
Gender and Age Distribution ៉ Background bare nuclei
៉ Female predominance ៉ Vascular component
៉ 30–40 years ៉ Possible lymphoid background

Clinical Features Histopathologic Findings


៉ Slow-growing mass ៉ Gross: encapsulated round mass, solid, friable, grey–white cut
៉ Occasionally painful surface; occasionally cystic
៉ Microscopic: variable growth patterns (solid microcystic,
papillary cystic, follicular) and variable appearance of
Treatment
neoplastic cells (acinic, intercalated duct, vacuolated, clear,
៉ Wide local excision, regional node dissection if nodes clinically
glandular NOS)
involved
Differential Diagnosis and Pitfalls
៉ Non-neoplastic salivary gland
៉ Cytoplasmic granularity: oncocytic neoplasms
៉ Lymphoid background: Warthin tumor
lymph nodes. The role of radiation therapy is contro-
៉ Clear/vacuolated cytoplasm: mucoepidermoid carcinoma,
versial. Markers of poor prognosis include pain or fi xa- epi-myoepithelial carcinoma, metastatic renal cell carcinoma,
tion, gross invasion, desmoplasia, cytologic atypia, and sebaceous neoplasms
increased mitotic activity. ៉ Cytologic atypia: other high-grade carcinomas

CYTOPATHOLOGIC FEATURES
granular to foamy cytoplasm, small dark nuclei, and
small nucleoli (Fig. 1-34). The cytoplasmic granules
Aspiration smears are cellular and contain cell frag- (zymogen granules) are variable in quantity and char-
ments with vague, irregular acinar configurations. acteristically PAS-positive, diastase-resistant. Ductal
Occasionally, a papillary configuration may be present. structures are absent. Cells with foamy cytoplasm have
Some tissue fragments will include portions of vascular few, if any, granules and have a clear cell appearance.
structures surrounded by neoplastic cells, reflecting the Although usually bland, nuclei can show variable
vascular nature of the neoplasm. Within sheets and degrees of atypia, including a more granular chromatin
clusters, cell borders are not distinct. When well dif- pattern and prominent nucleoli. The smear background
ferentiated, the tumor cells resemble normal acinar contains numerous bare nuclei that must be differenti-
cells. They have a bland appearance with abundant ated from lymphocytes. Lymphocytes are usually

A B

FIGURE 1-34
Acinic cell carcinoma. A, A loose cellular aggregate consists of bland-appearing cells with delicate granular and finely vacuolated cytoplasm. Cyto-
plasmic borders are indistinct and true acinar formation is absent. Bare nuclei of neoplastic cells are present in the background. Alcohol-fixed,
Papanicolaou stain, medium power. B, Irregular acinar formation, lack of ductal epithelium, and granular nature of the cytoplasm are characteristic
features. Air-dried, Diff-Quik stain, high power.

Ch001-F06731.indd 26 10/26/2006 10:19:52 AM


CHAPTER 1 Salivary Glands 27

absent; however, in approximately 10% of acinic cell abundant cytoplasm, the cytoplasm of oncocytes in
carcinomas (follicular variant), the stroma is rich in Warthin tumor is very dense in comparison to the
lymphoid cells with germinal centers. These tumors granular, foamy cytoplasm of acinic cell carcinoma
produce aspirates with abundant lymphoid elements in cells. This distinction would also aid in the differential
addition to the tumor cells. Poorly differentiated acinic diagnosis with oncocytic neoplasms. The cytoplasmic
cell carcinoma is uncommon and demonstrates easily granularity of oncocytes results from the abundance
recognizable features of malignancy and may be diffi- of mitochondria which stain positively with phospho-
cult to differentiate from other high-grade malignan- tungstic acid–hematoxylin (PTAH).
cies involving the parotid. The papillary cystic variant The presence of clear cells can raise the possibility of
may present with sheet or papillary clusters of neoplas- clear cell tumors, both primary and metastatic, espe-
tic cells resembling ductal epithelium. They will also cially mucoepidermoid carcinoma and metastatic renal
be associated with material from cyst content, such as cell carcinoma. Although aspirates from mucoepider-
debris and histiocytes, and may yield laminated, calci- moid carcinoma can contain cells with abundant foamy
fied structures that resemble psammoma bodies. This cytoplasm resembling acinar cells, the presence of back-
variant may be diagnostically challenging and difficult ground mucin and mucin-producing cells would exclude
to recognize as acinic cell carcinoma. a diagnosis of acinic cell carcinoma, which is negative
for mucin. Metastatic renal cell carcinoma is charac-
terized by the presence of clear cells showing nuclear
pleomorphism and prominent nucleoli. This tumor may
DIFFERENTIAL DIAGNOSIS AND PITFALLS show numerous small vessels within the tissue frag-
ments, a finding that is compatible with the diagnosis
of acinic cell carcinoma. Specific immunomarkers
Well-differentiated acinic cell carcinoma must be dif- for renal cell carcinoma, such as CD10, and clinical
ferentiated from normal salivary gland tissue or sialad- correlation will help resolve this differential. The rare
enosis. The absence of ductal structures and associated epithelial-myoepithelial carcinoma will present with
adipose tissue seen in normal salivary gland tissue clear cells; however, it may be distinguished by the iden-
can help in the diagnosis of neoplasia. In acinic cell tification of a biphasic cell population. The presence
carcinoma, there is discohesion of neoplastic cells, the of small ductal cells forming tubules within sheets of
acinar-like arrangements are loose, the groups are clear cells is a useful clue to the correct diagnosis. Acinic
poorly formed, and crowded sheets are seen. The grape- cell carcinoma with vacuolated cytoplasm may
like arrangement of acini in association with ductal mimic tumors with sebaceous differentiation, such as
structures seen in normal salivary gland tissue and the sebaceous lymphadenoma and sebaceous carcinoma
orderly array of cells within acini showing basally (Fig. 1-35).
located nuclei are lacking. Acinic cell carcinoma with The absence of acinar structures typical of acinic cell
lymphocyte-rich stroma must be differentiated from carcinoma in all of the tumors considered in the differ-
Warthin tumor. Although cells from both lesions have ential diagnosis can aid in the accurate interpretation.

FIGURE 1-35
Acinic cell carcinoma. Punched-out
small vacuoles are similar to those
seen in cells with sebaceous differ-
entiation and are present in asso-
ciation with cytoplasmic granularity.
Courtesy of Ms Jamie L Covell, The
University of Virginia Health Sci-
ences Center. Air-dried, Diff-Quik
stain, high power.

Ch001-F06731.indd 27 10/26/2006 10:19:53 AM


28 FINE NEEDLE ASPIRATION CYTOLOGY

SALIVARY DUCT CARCINOMA SALIVARY DUCT CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Branching and cribriform sheets and clusters of cells
CLINICAL FEATURES ៉ Background necrosis
៉ Frequent marked cytologic atypia: polygonal cells with granular
or vacuolated cytoplasm and prominent nucleoli
Salivary duct carcinoma is an uncommon and highly
៉ Rare low-grade cytologic features
aggressive neoplasm that arises in Stensen’s duct of the
parotid gland or in the submandibular gland, and has Histopathologic Findings
a predilection for men (M : F = 8 : 1). Although it can
៉ Similar to ductal carcinoma of the breast, comedo
occur at any age, patients are usually older and range ៉ Solid, cribriform, papillary, mucinous, infiltrating, and
in age from 62 to 89 years (median, 69 years). Treat- sarcomatoid patterns
ment consists of surgical excision followed by radiation
therapy. The prognosis of this tumor is dependent on Ancillary Studies
the proportion of invasive and in-situ component and ៉ Immunoreactivity for low and high molecular weight
on the histologic grade. Overall, the tumor has a poor cytokeratins, GCDFP-15, and androgen receptor (>90%)
prognosis, and approximately two-thirds of the patients ៉ Moderate positivity for PAP and PSA
die within 4 years of initial diagnosis. Tumor size, ៉ Rare positivity for ER and PR
presence of distant metastases, and c-erbB-2 amplifica- ៉ Overexpression of p53 and c-erbB-2
tion are independent prognostic parameters in patients
with salivary duct carcinoma. Salivary duct carcino- Differential Diagnosis and Pitfalls
ma’s immunophenotypic profi le is more closely related ៉ High-grade cytologic features: high-grade mucoepidermoid
to prostate carcinoma. A rare case report describes carcinoma, carcinoma ex-pleomorphic adenoma, adenosquamous
its association with elevated serum prostate-specific carcinoma, intraductal papillary adenocarcinoma, metastatic
antigen (PSA) levels. squamous cell carcinoma, malignant melanoma
៉ Low-grade cytologic features: oncocytoma, Warthin tumor

CYTOPATHOLOGIC FEATURES

Smears are variably cellular but obviously malignant.


The most characteristic features of salivary duct carci- prominent nucleoli (Fig. 1-37). The cytoplasm varies
noma are broad flat and branching sheets of cells with from dense granular to vacuolated. Cases with bland or
a cribriform, papillary, or gland-like pattern in a vari- mildly pleomorphic nuclei are reported. Salivary duct
ably necrotic background (Fig. 1-36). Dense fibrous carcinoma mostly resembles breast carcinoma in all its
fragments may also be seen in the background. The variable patterns and features, but shares an immuno-
tumor cells are large and polygonal with round to oval, phenotypic profi le with both ductal-type adenocarci-
pleomorphic, eccentric or centrally placed nuclei, and noma and prostatic adenocarcinoma. Malignant cells
are diffusely immunoreactive for low and high molecu-
lar weight cytokeratins. Salivary duct carcinoma will
show positivity for estrogen receptor (ER) and proges-
terone receptor (PR) in a minority of cases only – 1.3%
and 6%, respectively – but immunoreactivity to GCDFP-
SALIVARY DUCT CARCINOMA – DISEASE FACT SHEET
15 is present in most cases. Androgen receptor, a marker
expressed in prostate carcinoma, is expressed in over
Incidence and Location
90% of salivary duct carcinomas, whereas 60% of cases
៉ Uncommon salivary gland neoplasm
show scattered moderate positivity for prostatic acid
៉ Parotid > submandibular
phosphatase (PAP), and 17% diffuse moderate positiv-
Gender and Age Distribution
ity for PSA. Overexpression of p53 is reported in 53%
of cases and c-erbB-2 in 63%.
៉ Male to female ratio is 8 : 1
៉ Wide age range, more often 60–90 years
A rare sarcomatoid variant has been described in
association with usual-type salivary duct carcinoma. It
Clinical Features presents as a spindle and anaplastic cell tumor. Another
៉ Aggressive growth
rare variant of salivary duct carcinoma is small cell
undifferentiated carcinoma. While some immunohisto-
Prognosis and Treatment chemical studies have demonstrated luminal, basal, and
៉ Regional and distant metastases frequent, 70% mortality
myoepithelial differentiation compatible with origin of
៉ Surgical excision and radiation therapy
the tumor from the salivary duct, other studies have
demonstrated neuroendocrine differentiation of this
tumor.

Ch001-F06731.indd 28 10/26/2006 10:19:53 AM


CHAPTER 1 Salivary Glands 29

FIGURE 1-36
Salivary duct carcinoma. Broad
complex epithelial sheets with a
branching papillary and cribriform
appearance usually occur against a
necrotic background. Courtesy of Ms
Jamie L Covell, The University of
Virginia Health Sciences Center. Air-
dried, Diff-Quik stain, low power.

FIGURE 1-37
Salivary duct carcinoma. Large
polygonal cells with pleomorphic
round nuclei, prominent nucleoli,
and abundant dense granular cyto-
plasm are crowded in this duct-
like structure with central debris.
Although the cytologic features are
somewhat similar to those of onco-
cytes, the nuclear atypia and necro-
sis should preclude a benign
diagnosis. Courtesy of Ms Jamie L
Covell, The University of Virginia
Health Sciences Center. Air-dried,
Diff-Quik stain, high power.

dominate, an intraductal papillary adenocarcinoma


DIFFERENTIAL DIAGNOSIS AND PITFALLS should be considered. Intraductal papillary adenocarci-
noma, a low-grade carcinoma and the malignant coun-
terpart of intraductal papilloma, shows cytologic atypia
FNAs of salivary duct carcinoma are difficult to inter- and microinvasion, and has a better prognosis than
pret because of the morphologic spectrum of this tumor. salivary duct carcinoma. Metastatic tumors such as
In high-grade tumors where pleomorphism is evident, squamous cell carcinoma and malignant melanoma
other high-grade malignant tumors enter the differen- should also be excluded. Low-grade tumors are a source
tial diagnosis. Such group of malignant tumors includes of false negative diagnoses. When bland features are
high-grade carcinoma not otherwise specified (NOS), present, the neoplastic cells may show similarities to
high-grade mucoepidermoid carcinoma, and carcinoma oncocytes, and the tumor may be mistaken for onco-
ex-pleomorphic adenoma. If papillary structures pre- cytoma or Warthin tumor.

Ch001-F06731.indd 29 10/26/2006 10:19:53 AM


30 FINE NEEDLE ASPIRATION CYTOLOGY

arise in a background of LESA, Sjögren syndrome, or


MALIGNANT LYMPHOMA Warthin tumor. The MALT/marginal zone lympho-
mas, frequently encountered in the salivary glands,
have an indolent clinical course and an excellent
CLINICAL FEATURES prognosis.

Most patients with malignant lymphoma will present


with a unilateral salivary gland mass. Primary malig- CYTOPATHOLOGIC FEATURES
nant lymphoma can arise in the gland itself, mostly
involving the parotid gland, and occasionally the Most commonly, salivary glands are involved by B-cell
submandibular gland. In the area of the parotid gland, lymphomas, which in the majority of cases comprise
malignant lymphoma also arises in intra- or periparotid small lymphocytes and belong to the extranodal mar-
lymph nodes and has the features or clinical course of ginal zone B-cell lymphoma of MALT type. Typically
a nodal lymphoma. Salivary glands may also be second- in such cases, smears show a population of small to
arily involved by lymphoma as an expression of dis- intermediate lymphocytes with round to slightly angu-
seminated disease. Most malignant lymphomas in the lated nuclei, and occasional larger cells with features
salivary gland are of B-cell lineage, and Hodgkin lym- of immunoblasts (Fig. 1-38).
phoma is unusual. Malignant lymphoma is known to Follicular lymphomas occur in the salivary gland
with a high frequency as well (35–50%) and are com-
posed of small and large cleaved cells and large non-
cleaved cells that impart a polymorphous appearance to
MALIGNANT LYMPHOMA – DISEASE FACT SHEET the aspirate material. Conversely, diffuse large B-cell
lymphomas consist of large centroblastic and immuno-
Incidence
blastic lymphoid cells that are overtly malignant.
៉ 2–5% of salivary gland neoplasms
T-cell lymphoma in the salivary gland is rare and
cannot be distinguished from a B-cell tumor on a mor-
Clinical Features
phologic basis only. Another rare entity is plasma-
៉ Primary or secondary
cytoma that can present as a salivary gland mass with
៉ Unilateral mass, most commonly involving parotid or lymph nodes
cytologic features varying from mature to anaplastic
within area
៉ Behavior and treatment similar to that of nodal lymphoma
plasma cells, similar to those seen in other sites (Fig.
1-39). Patients with plasmacytoma can later develop
multiple myeloma.

FIGURE 1-38
Malignant lymphoma, marginal
zone. Small to intermediate cell
lymphocytes with round to slightly
angulated nuclei are the predomi-
nant cell population. Scattered
larger cells and a mitotic figure are
present. Air-dried, Diff-Quik stain,
high power.

Ch001-F06731.indd 30 10/26/2006 10:19:55 AM


CHAPTER 1 Salivary Glands 31

FIGURE 1-39
Plasmacytoma. Mature plasma cells
with round eccentric nuclei, clock-
face chromatin, and a moderate
amount of cytoplasm may resemble
hyaline myoepithelial cells. The
chromatin pattern and perinuclear
Hoff are distinguishing features.
Alcohol-fixed, Papanicolaou stain,
medium power.

flow cytometry is necessary to support the diagnosis of


MALIGNANT LYMPHOMA – PATHOLOGIC FEATURES lymphoma. A predominantly lymphoid infi ltrate can be
aspirated from neoplasms such as Warthin tumor, seba-
Cytopathologic Findings ceous lymphadenoma, and acinic cell carcinoma in
៉ Single discohesive cells which the epithelial cells have not been sampled. A
៉ Monotonous population of lymphocytes thorough search for epithelial cells or identification of
៉ Variable cell size and degree of atypia, dependent on cell type such cells by immunocytochemistry may be helpful in
៉ B-cell lymphomas most common identifying a non-lymphoid neoplasm. Lymphoepithe-
lial carcinoma that consists of a mixture of large
Pathologic Findings
anaplastic-appearing cells in a background of small
៉ Follicular diffuse or sclerosing growth pattern lymphocytes is also a consideration in the differential
៉ Commonly, cleaved or small cells
diagnosis of large cell lymphoma. Malignant melanoma
៉ Immunoblastic, plasma cell, or T-cell less frequent
៉ T-cell associated with prominent lymphoepithelial lesions
may need to be excluded if the lymphoma is immuno-
៉ Hodgkin lymphoma rare blastic in nature. The last two non-lymphoid malignant
neoplasms can be identified by immunocytochemical
Ancillary Studies staining for cytokeratin and for S-100 protein, Melan
៉ Flow cytometric studies
A, or HMB-45, respectively.

Differential Diagnosis and Pitfalls


៉ Chronic sialadenitis
៉ Lymphoepithelial lesion RARE TUMORS
៉ Poorly sampled lymphocyte-rich lesions (Warthin tumor,
sebaceous lymphadenoma)
៉ Lymphoepithelial carcinoma in large cell lymphoma
៉ Malignant melanoma in immunoblastic lymphoma The following section addresses in brief both benign
and malignant tumors that are only exceptionally
encountered.
Benign tumors such as vascular tumors, lymphangi-
oma, lipomas, schwannomas, skin adnexal-type tumors
DIFFERENTIAL DIAGNOSIS AND PITFALLS such as pilomatrixoma, and solitary fibrous tumors have
all been described in the salivary glands. Malignant
neoplasms include the following:
The differential diagnosis of malignant lymphoma in Primary small cell carcinoma of the salivary gland is
the salivary gland includes a number of lymphocyte- extremely rare and can be diagnosed only after exclud-
rich conditions, both neoplastic and non-neoplastic. ing other primary sources. It affects predominantly men
Low-grade lymphomas have to be differentiated from in their sixth decade and has morphologic and cytologic
chronic sialadenitis, LESA, lymphoepithelial cysts, features similar to small cell undifferentiated carcinoma
and intraparotid lymph nodes. Immunophenotyping by in other locations. It also has a similar aggressive course

Ch001-F06731.indd 31 10/26/2006 10:19:55 AM


32 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-40
Small cell undifferentiated carci-
noma. Poorly cohesive clusters and
syncytia of small blue cells with a
high N/C ratio. The nuclei are angu-
lated and molded, the chromatin is
granular, and nucleoli are incon-
spicuous. Alcohol-fixed, Papanico-
laou stain, high power.

and poor prognosis. Cellular smears contain poorly in-situ hybridization or by serology. Cytologically, it
cohesive clusters and syncytia of small blue cells with a consists of syncytial sheets of large, highly pleomorphic
high N/C ratio. The nuclei are angulated and molding, cells with vesicular chromatin, prominent nucleoli, and
the chromatin is granular, and nucleoli are inconspicu- delicate poorly defined cytoplasm, in an abundant back-
ous (Fig. 1-40). Mitotic figures are easily found. Ultra- ground of mixed lymphocytes and plasma cells (Fig.
structurally, it is characterized by neurosecretory 1-42). The main differential diagnosis is malignant lym-
granules and demonstrates neuroendocrine features by phoma and can be resolved by a positive cytokeratin
immunocytochemistry, including dot-like positivity for immunostain in the large atypical cells of lymphoepithe-
cytokeratin and staining with chromogranin, synapto- lial carcinoma.
physin, neuron-specific enolase, and CD56. Similar to Clear cell carcinomas do not constitute a homoge-
Merkel cell tumors and unlike lung primaries, small cell neous group of tumors but comprise tumors with abun-
carcinoma of salivary gland origin is positive for CK20. dant cytoplasmic glycogen, lipid, or mucin. Such tumors
The main differential diagnostic consideration in the include clear cell myoepitheliomas, neoplasms with
salivary gland, apart from a metastasis from lung or sebaceous differentiation or oncocytic features, muco-
skin, is a solid variant of adenoid cystic carcinoma. epidermoid carcinoma, acinic cell carcinoma, and meta-
Adenocarcinoma not otherwise specified is a diagnostic static renal cell carcinoma. If all such possibilities are
category that is used for high-grade tumors without excluded, the diagnosis of hyalinizing clear cell carci-
recognizable specific features. noma should be considered, particularly in a tumor
Squamous cell carcinoma as a primary tumor in the occurring in the oral cavity.
salivary gland is unusual. Some squamous-appearing Sarcomas may arise in salivary glands. In adults,
tumors may represent adenosquamous carcinoma, as those are malignant spindle cell neoplasms that should
demonstrated by scattered mucin-containing cells. be distinguished from metaplastic carcinomas, myo-
These tumors are highly aggressive and are treated with epithelial tumors, or spindle cell melanoma. Sarcomas
radical surgery and radiation therapy. More commonly, described in that location include malignant peripheral
squamous carcinoma results from secondary involve- nerve sheath tumor, synovial sarcoma, Kaposi sarcoma,
ment of the salivary gland either by direct extension or malignant fibrous histiocytoma, primitive neuroectoder-
by metastasis to an intra- or periglandular lymph node mal tumors/Ewing sarcoma, and, in children, embryo-
from a head and neck or skin primary. Cystic metastases nal rhabdomyosarcoma.
are frequently aspirated and show keratinizing and
non-keratinizing atypical squamous cells in a necrotic
background (Fig. 1-41).
Lymphoepithelial carcinoma of the salivary gland
has an incidence of less than 0.5% of salivary gland SECONDARY TUMORS
neoplasms, and occurs predominantly in Greenlanders,
Inuits, and Southern Chinese. It presents as a solitary
mass of the parotid or submandibular gland, and, like Metastatic tumors to the region of the salivary glands
its nasopharyngeal counterpart, is associated with occur not infrequently, and involve intraparotid and
Epstein-Barr virus, which can be demonstrated by submandibular lymph nodes. If metastasis is suspected,

Ch001-F06731.indd 32 10/26/2006 10:19:56 AM


CHAPTER 1 Salivary Glands 33

FIGURE 1-41
Squamous cell carcinoma. Keratin-
izing and non-keratinizing atypical
squamous cells in a necrotic back-
ground are compatible with a cystic
tumor of the head and neck primary.
The definite nuclear atypia and
cellularity separate this lesion from
non-neoplastic branchial cleft cysts
and cystic lymphoepithelial lesions.
Alcohol-fixed, Papanicolaou stain,
high power.

FIGURE 1-42
Lymphoepithelial carcinoma. Syncy-
tial sheets of large highly pleomor-
phic cells with vesicular chromatin,
prominent nucleoli, and delicate,
poorly defined cytoplasm in an
abundant background of mixed
lymphocytes and plasma cells may
mimic a large cell malignant
lymphoma. Courtesy of Ms Jamie
L Covell, The University of Virginia
Health Sciences Center. Alcohol-
fixed, Papanicolaou stain, high
power.

a clinical history of a primary tumor elsewhere, and plasm, eccentrically placed nuclei, and prominent
comparison of aspirate to pathology material from the nucleoli. A pronounced discohesive pattern, intra-
primary are always helpful. Most frequently, head and nuclear pseudoinclusions, binucleation, and melanin
neck squamous cell carcinoma forms a cystic necrotic pigment are diagnostic clues. In the absence of melanin
mass that may mimic some of the primary benign pigment, positive immunostaining for S-100 protein,
squamous-lined cystic lesions. Identifying atypia HMB-45, and Melan A confirms the diagnosis.
within the squamous cell is the way to a correct Tumors in the lung, kidney, breast, and, to a lesser
diagnosis. Malignant melanoma metastasizes to the extent, prostate and colon metastasize to the salivary
salivary gland and may mimic some of the primary glands. Lung, breast, and prostate carcinomas may
high-grade carcinomas or lymphomas. Malignant mela- mimic high-grade adenocarcinomas arising in the sali-
noma consists of large polygonal cells arranged in vary gland. However, these metastases may be distin-
loosely cohesive clusters and single cells. Cells typically guished from salivary gland primaries by a thorough
have a moderate to abundant amount of dense cyto- clinical history and immunostains that are site-specific.

Ch001-F06731.indd 33 10/26/2006 10:19:57 AM


34 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 1-43
Renal cell carcinoma. Large neo-
plastic cells with abundant translu-
cent, granular, or vacuolated
cytoplasm have large round cen-
trally located nuclei and are
arranged around and in between
capillaries. These findings may
overlap with those of an acinic cell
carcinoma; however, cells of renal
cell carcinoma may be larger and
display a more abundant delicate
and vacuolated cytoplasm. Air-
dried, Diff-Quik stain, medium
power.

TTF-1 stains a significant number of lung adenocarci- diagnosis by fine-needle aspiration biopsy. Diagn Cytopathol 1997;17:
noma; PSA and PAP, and ER and PR may help detect 183–190.
Cheuk W, Chan JK. Kuttner tumor of the submandibular gland: fine-needle
prostate and breast carcinoma, respectively. A tall aspiration cytologic findings of seven cases. Am J Clin Pathol 2002;
columnar glandular population associated with mucin 117:103–108.
production or a necrotic background may suggest a Lopez-Rios F, Diaz-Bustamante T, Serrano-Egea A, Jimenez J, de Agustin
colonic primary. Immunostains for the main mucin P. Amylase crystalloids in salivary gland lesions: report of a case with
a review of the literature. Diagn Cytopathol 2001;25:59–62.
protein cores (MUC1, MUC2, MUC3, MUC5AC), CDX2,
and villin may help confirm the site of origin of the
tumor. Metastases from renal cell carcinoma may pose
Cystic Lesions
a challenge to the cytopathologist, as they mimic acinic
cell or mucoepidermoid carcinoma. Neoplastic cells are Chhieng DC, Argosino R, McKenna BJ, Cangiarella JF, Cohen JM. Utility of
large with abundant translucent, granular or vacuolated fine-needle aspiration in the diagnosis of salivary gland lesions in
patients infected with human immunodeficiency virus. Diagn Cytopathol
cytoplasm. Nuclei are large, round, and may display 1999;21:260–264.
prominent nucleoli. Renal cell carcinoma is rich in Elliot JN, Oertel YC. Lymphoepithelial cysts of the salivary glands –
vessels that may be easily visualized in the FNA histologic and cytologic features. Am J Clin Pathol 1990;93:39–43.
material (Fig. 1-43). Layfield LJ, Gopez EV. Cystic lesions of the salivary glands: cytologic
features in fine-needle aspiration biopsies. Diagn Cytopathol 2002;27:
A positive CD10 immunostain will help support the 197–204.
renal origin of the tumor. Stewart CJR, MacKenzie K, McGarry GW, Mowatt A. Fine-needle aspiration
cytology of salivary gland: a review of 341 cases. Diagn Cytopathol
2000;22:139–146.

SUGGESTED READINGS
Non-neoplastic Lesions Tumor with Mesenchymal Differentiation

Sialadenosis Benign Mixed Tumor

Henrey-Stanley MJ, Beneke J, Bardales HR, Stanley MW. Fine-needle Chhieng DC, Cohen JM, Cangiarella JF. Fine-needle aspiration of spindle
aspiration of normal tissue from enlarged salivary glands: sialosis or cell and mesenchymal lesions of the salivary gland. Diagn Cytopathol
missed target? Diagn Cytopathol 1995;13:300–303. 2000;23:253–259.
Young NA, Mody DR, Davey DD. Misinterpretation of normal cellular ele- Das DK, Anim JT. Pleomorphic adenoma of salivary gland: to what extent
ments in fine-needle aspiration biopsy specimens: observations from does fine needle aspiration cytology reflect histopathological features?
the College of American Pathologists Interlaboratory Comparison Cytopathology 2005;16:65–70.
Program in Non-Gynecologic Cytopathology. Arch Pathol Lab Med 2002; Elsheikh TM, Bernacki EG. Fine needle aspiration cytology of cellular
126:670–675. pleomorphic adenoma. Acta Cytol 1996;40:1165–1175.
McLeod CB, Frable WJ. Fine-needle aspiration biopsy of the salivary gland:
problem cases. Diagn Cytopathol 1993;9:216–225.
Sialadenitis Mooney EE, Dodd LG, Layfield LJ. Squamous cells in fine-needle aspiration
biopsies of salivary gland lesions: potential pitfalls in cytologic diag-
Allen EA, Ali SZ, Mathew S. Lymphoid lesions of the parotid. Diagn nosis. Diagn Cytopathol 1996;15:447–452.
Cytopathol 1999;21:170–173. Viguer JM, Vicandi B, Jimenez-Heffernan JA, Lopez-Ferrer P, Limeres MA.
Chai C, Dodd LG, Glasgow BJ, Layfield LJ. Salivary gland lesions with a Fine needle aspiration of pleomorphic adenoma. An analysis of 212
prominent lymphoid component: cytologic findings and differential cases. Acta Cytol 1997;41:786–794.

Ch001-F06731.indd 34 10/26/2006 10:19:59 AM


CHAPTER 1 Salivary Glands 35

Malignant Mixed Tumor Stanley MW, Horwitz CA, Henry MJ, Burton LG, Lowhagen T. Basal cell
adenoma of the salivary gland: a benign adenoma that cytologically
Anand A, Brockie ES. Cytomorphologic features of salivary duct carcinoma mimics adenoid cystic carcinoma. Diagn Cytopathol 1988;4:342–
ex-pleomorphic adenoma: diagnosis by fine needle aspiration biopsy 346.
with histologic correlation. Diagn Cytopathol 1999;20:375–378. Stanley MW, Horwitz CA, Rollins SD, et al. Basal cell (monomorphic) and
De la Torre M, Larsson E. Fine-needle aspiration cytology of carcinosar- minimally pleomorphic adenomas of the salivary glands. Distinction
coma of the parotid gland: cytohistological and immunohistochemical from the solid type of adenoid cystic carcinoma in fine-needle aspira-
findings. Diagn Cytopathol 1995;12:350–353. tion. Am J Clin Pathol 1996;106:35–41.
Nigam S, Kumar N, Jain S. Cytomorphologic spectrum of carcinoma ex
pleomorphic adenoma. Acta Cytol 2004;48:309–314.
Pitman MB. Mucoepidermoid carcinoma ex-pleomorphic adenoma of the Adenoid Cystic Carcinoma
parotid gland. Acta Cytol 1995;39:604–606.
Nagel H, Hotze HJ, Laskawi R, Chilla R, Droese M. Cytologic diagnosis of
adenoid cystic carcinoma of salivary glands. Diagn Cytopathol 1999;20:
Myoepithelial Neoplasms 358–366.
Stanley MW, Horwitz CA, Rollins SD, et al. Basal cell (monomorphic) and
Darvishian F, Lin O. Myoepithelial cell-rich neoplasms: cytologic features minimally pleomorphic adenomas of the salivary glands. Distinction
of benign and malignant lesions. Cancer 2004;102:355–361. from the solid type of adenoid cystic carcinoma in fine-needle aspira-
Das DK, Haji BE, Ahmed MS, Hossain MN. Myoepithelioma of the parotid tion. Am J Clin Pathol 1996;106:35–41.
gland initially diagnosed by fine needle aspiration cytology and immuno- Yu GH, Caraway NP. Poorly-differentiated adenoid cystic carcinoma: cyto-
cytochemistry: a case report. Acta Cytol 2005;49:65–70. logic appearance in fine-needle aspirates of distant metastases. Diagn
Gupta RK, Naran S, Dowle C, Simpson JS. Coexpression of vimentin, cyto- Cytopathol 1996;15:296–300.
keratin and S-100 in monomorphic adenoma of salivary gland; value of
marker studies in the differential diagnosis of salivary gland tumors.
Cytopathology 1992;3:303–309. Polymorphous Low-Grade Adenocarcinoma
Kumar PV, Sobhani SA, Monabati A, Hashemi SB, Eghtadari F, Hamidi SA.
Myoepithelioma of the salivary glands. Fine needle aspiration biopsy Frierson HF Jr, Covell JL, Mills SE. Fine-needle aspiration cytology of ter-
findings. Acta Cytol 2004;48:302–308. minal duct carcinoma of minor salivary gland. Diagn Cytopathol 1987;3:
Ng W, Choy C, Ip P, Shek W, Collins RJ. Fine needle aspiration cytology 159–162.
of epithelial-myoepithelial carcinoma of salivary gland – a report of Gibbons D, Saboorian MH, Vuitch F, Gokaslan ST, Ashfaq R. Fine-needle
three cases. Acta Cytol 1999;43:675–680. aspiration findings in patients with polymorphous low grade adenocar-
Schultenover SJ, McDonald EC, Ramzy I. Hyaline-cell pleomorphic adenoma: cinoma of salivary glands. Cancer 1999;87:31–36.
diagnosis by fine needle aspiration biopsy. Acta Cytol 1984;28:593– Saenz, Santmaria J, Catalina-Fernandez I. Polymorphous low grade adeno-
597. carcinoma of the salivary gland. Diagnosis by fine needle aspiration
cytology. Acta Cytol 2004;48:52–56.

Warthin Tumor/Oncocytic Neoplasms


Mucoepidermoid Carcinoma
Allen EA, Ali SZ, Mathew S. Lymphoid lesions of the parotid. Diagn Cyto-
pathol 1999;21:170–173. Al-Khafaji BM, Nestok BR, Katz RL. Fine-needle aspiration of 154 parotid
Ballo MS, Shin HJ, Sneige N. Sources of diagnostic error in fine-needle masses with histologic correlation: ten-year experience at the Univer-
aspiration diagnosis of Warthin’s tumor and clues to a correct diagnosis. sity of Texas M.D. Anderson Cancer Center. Cancer (Cancer Cytopathol)
Diagn Cytopathol 1997;17:230–234. 1998;84:153–159.
Chai C, Dodd LG, Glasgow BJ. Salivary gland lesions with a promi- Cajulis RS, Gokaslan ST, Yu GH, Frias-Hidvegi D. Fine needle aspiration
nent lymphoid component: cytologic findings and differential diagnosis biopsy of the salivary glands – a five-year experience with emphasis on
by fine-needle aspiration biopsy. Diagn Cytopathol 1997;17:183– diagnostic pitfalls. Acta Cytol 1997;41:1412–1420.
190. Chan MKM, McGuire LJ, King W, Li AKC, Lee JCK. Cytodiagnosis of 112
Chhieng DC, Cangiarella JF, Cohen JM. Fine-needle aspiration cytology of salivary gland lesions – correlation with histologic and frozen section
lymphoproliferative lesions involving the major salivary glands. Am J diagnosis. Acta Cytol 1992;36:353–363.
Clin Pathol 2000;113:563–571. Cohen MB, Fisher PE, Holly EA, Ljung BM, Lowhagen T, Bottles K. Fine
Klijanienko J, Vielh P. Fine-needle sampling of salivary gland lesions II. needle aspiration biopsy diagnosis of mucoepidermoid carcinoma –
Cytology and histology correlation of 71 cases of Warthin’s tumor statistical analysis. Acta Cytol 1990;34:43–49.
(adenolymphoma). Diagn Cytopathol 1997;16:221–225. Jakobson PA , Blanck C, Eneroth C-M. Mucoepidermoid carcinoma of the
Mooney EE, Dodd LG, Layfield LJ. Squamous cells in fine-needle aspiration parotid gland. Cancer 1968;22:111–124.
biopsies of salivary gland lesions: potential pitfalls in cytologic diag- Klijianienko J, Vielh P. Fine-needle sampling of salivary gland lesions.
nosis. Diagn Cytopathol 1996;15:447–452. IV. Review of 50 cases of mucoepidermoid carcinoma with histologic
Parwani AV, Ali SZ. Diagnostic accuracy and pitfalls in fine-needle aspira- correlation. Diagn Cytopathol 1997;17:92–98.
tion interpretation of Warthin tumor. Cancer 2003;99:166–171. Kumar N, Kapila K, Verma K. Fine needle aspiration cytology of mucoepi-
Ryska A, Seifert G. Adenolymphoma (Warthin’s tumor) with multiple dermoid carcinoma. A diagnostic problem. Acta Cytol 1991;35:357–
sarcoid like granulomas. Pathol Res Pract 1999;195:835–839. 359.
Sherman ME, Magro C, Berry Y, Syzyfelbein WM. Oncocytic nodule. An Layfield LJ, Glasgow BJ. Aspiration cytology of clear-cell lesions of the
unusual case of a submaxillary gland mass in an elderly patient. Acta parotid gland: morphologic features and differential diagnosis. Diagn
Cytol 1990;34:827–830. Cytopathol 1993;9:705–712.
Williamson JD, Simmons BH, EL-Naggar A, Medeiros LJ. Mucoepidermoid MacLeod CB, Frable WJ. Fine-needle aspiration biopsy of salivary gland:
carcinoma involving Warthin tumor. A report of five cases and review problem cases. Diagn Cytopathol 1993;9:216–225.
of the literature. Am J Clin Pathol 2000;114:564–570. Palombini L. Challenges in the interpretation of FNAs from the salivary
glands. Diagn Cytopathol 1997;17:417–421.

Basaloid Neoplasms
Acinic Cell Carcinoma
Basal Cell Adenoma/Basal Cell Adenocarcinoma
Ali SZ. Acinic-cell carcinoma, papillary-cystic variant: a diagnostic dilemma
Pisharodi LR. Basal cell adenocarcinoma of the salivary gland. Diagnosis in salivary gland aspiration. Diagn Cytopathol 2002;27:244–250.
by fine-needle aspiration. Am J Clin Pathol 1995;103:603–608. Al-Khafaji BM, Nestok BR, Katz RL. Fine-needle aspiration of 154 parotid
Stanley MW, Horwitz CA, Bardales RH, Stern SJ, Korourian S. Basal cell masses with histologic correlation: ten-year experience at the Univer-
carcinoma metastatic to the salivary glands: differential diagnosis in sity of Texas M.D. Anderson Cancer Center. Cancer (Cancer Cytopathol)
fine-needle aspiration cytology. Diagn Cytopathol 1997;16:247–252. 1998;84:153–159.

Ch001-F06731.indd 35 10/26/2006 10:19:59 AM


36 FINE NEEDLE ASPIRATION CYTOLOGY

Baccato P, Altavilla G, Blandamura S. Fine-needle aspiration of salivary Chhieng DC, Cangiarella JF, Cohen JM. Fine-needle aspiration cytology of
gland lesions – a reappraisal of pitfalls and problems. Acta Cytol lymphoproliferative lesions involving the major salivary glands. Am J
1998;42:888–898. Clin Pathol 2000;113:563–571.
Cajulis RS, Gokaslan ST, Yu GH, Frias-Hidvegi D. Fine needle aspiration Crapanzano JP, Lin O. Cytologic findings of marginal zone lymphoma.
biopsy of the salivary glands – a five-year experience with emphasis on Cancer 2003;99:301–309.
diagnostic pitfalls. Acta Cytol 1997;41:1412–1420. MacCallum PL, Lampe HB, Cramer H, Matthews TW. Fine-needle aspiration
Klijanienko J, Vielh P. Fine-needle sampling of salivary gland lesions V: cytology of lymphoid lesions of the salivary gland: a review of 35 cases.
cytology of 22 cases of acinic cell carcinoma with histologic correlation. J Otolaryngol 1996;25:300–304.
Diagn Cytopathol 1997;17:347–352. Ustun MO, Ekinci N, Payzin B. Extramedullary plasmacytoma of the
Layfield LJ, Glasgow BJ. Aspiration cytology of clear-cell lesions of the parotid gland. Report of a case with extensive amyloid deposition
parotid gland: morphologic features and differential diagnosis. Diagn masking the cytologic and histopathologic picture. Acta Cytol 2001;45:
Cytopathol 1993;9:705–712. 449–453.
Lewis JE, Olsen KD, Weiland LH. Acinic cell carcinoma – clinicopathologic
review. Cancer 1991;67:172–179.
Nagel H, Laskawi R, Buter JJ, Schroder M, Chilla R, Droese M. Cytologic Rare Tumors
diagnosis of acinic-cell carcinoma of salivary glands. Diagn Cytopathol
1997;16:402–412. Chhieng DC, Cohen JM, Cangiarella JF. Fine-needle aspiration of spindle
Palombini L. Challenges in the interpretation of FNAs from the salivary cell and mesenchymal lesions of the salivary glands. Diagn Cytopathol
glands. Diagn Cytopathol 1997;17:417–421. 2000;23:253–259.
Sheyn I, Yassin R, Seiden A, Nestok BR. Papillary-cystic variant of acinic Gnepp DR, Wick MR. Small cell carcinoma of the major salivary glands: an
cell carcinoma of the salivary gland diagnosed by fine needle aspiration immunohistochemical study. Cancer 1990;66:185–192.
biopsy. A case report. Acta Cytol 2000;44:1073–1076. Mair S, Philips JI, Cohen R. Small cell undifferentiated carcinoma of the
parotid gland. Cytologic, histologic, immunohistochemical and ultra-
structural features of a neuroendocrine variant. Acta Cytol 1989;33:
Salivary Duct Carcinoma 164–168.
Milchgrub S, Vuitch F, Saboorian MH, Hameed A, Wu H, Albores-Saavedra
Dee S, Masood S, Isaacs JH Jr, Hardy NM. Cytomorphologic features of J. Hyalinizing clear-cell carcinoma of salivary glands in fine-needle
salivary duct carcinoma on fine needle aspiration biopsy. A case report. aspiration. Diagn Cytopathol 2000;23:333–337.
Acta Cytol 1993;37:539–542. Nagao T, Sugano I, Matsuzaki O, Hara H, Kondo Y, Nagao K. Intra-
Domson KK, Wakely PE Jr. Aspiration and imprint cytopathology of salivary ductal papillary tumors of the major salivary glands: case reports of
duct carcinoma. Cancer 1997;81:281–286. benign and malignant variants. Arch Pathol Lab Med 2000;124:
Elsheikh TM, Bernacki EG, Pisharodi L. Fine-needle aspiration cytology of 291–295.
salivary duct carcinoma. Diagn Cytopathol 1994;11:47–51. Thompson MB, Nestok BR, Gluckman JL. Fine needle aspiration cytology
Ersoz C, Cetik F, Aydin O, Cosar EF, Talas DU. Salivary duct carcinoma ex- of lymphoepithelioma-like carcinoma of the parotid gland. A case
pleomorphic adenoma: analysis of the findings in fine needle aspiration report. Acta Cytol 1994;38:782–786.
cytology and histology. Diagn Cytopathol 1998;19:201–204. Toyosawa S, Ohnishi A, Ito R, et al. Small cell undifferentiated carcin-
Fyrat P, Cramer H, Feczko JD, et al. Fine needle aspiration biopsy of oma of the submandibular gland: immunohistochemical evidence of
salivary duct carcinoma: report of five cases. Diagn Cytopathol 1997;16: myoepithelial, basal and luminal cell features. Pathol Int 1999;49:887–
526–530. 892.
Gilcrease MZ, Guzman-Paz M, Froberg K, Pambiccian S. Salivary duct
carcinoma. Is a specific diagnosis possible by fine needle aspiration
cytology? Acta Cytol 1998;42:1389–1396. Secondary Tumors
James GK, Pudek M, Berean KW, Diamandis EP, Archibald BL. Salivary duct
carcinoma secreting prostate-specific antigen. Am J Clin Pathol 1996; Hughes JH, Jensen CS, Donnelly AD, et al. The role of fine-needle aspira-
106:242–247. tion cytology in the evaluation of metastatic clear cell tumors. Cancer
Kapadia SB, Barnes L. Expression of androgen receptor, gross cystic 1999;87:380–389.
disease fluid protein, and CD44 in salivary duct carcinoma. Mod Pathol Lussier C, Klijanienko J, Vielh P. Fine-needle aspiration of metastatic
1998;11:1033–1038. nonlymphomatous tumors to the major salivary glands: a clinicopatho-
Khurana KK, Pitman MB, Powers CN, Korourian S, Bardales RH, Stanley logic study of 40 cases cytologically diagnosed and histologically
MW. Diagnostic pitfalls of aspiration cytology of salivary duct carci- correlated. Cancer 2000;90:350–356.
noma. Cancer 1997;81:373–378. Stanley MW, Bardales RH, Farmer CE, et al. Primary and metastatic high-
Moriki T, Ueta S, TakahashiT, Mitani M, Ichien M. Salivary duct carcinoma: grade carcinomas of the salivary glands: a cytologic-histologic study of
cytologic characteristics and application of androgen receptor immuno- twenty cases. Diagn Cytopathol 1995;13:37–43.
staining for diagnosis. Cancer 2001;93:344–350. Yang B, Ali SZ, Rosenthal DL. CD10 facilitates the diagnosis of metastatic
renal cell carcinoma from primary adrenal cortical neoplasm in adrenal
fine-needle aspiration. Diagn Cytopathol 2002;27:149–152.
Lymphoproliferative Disorders Zhang C, Cohen JM, Cangiarella JF, Waisman J, McKenna BJ, Chhieng
DC. Fine-needle aspiration of secondary neoplasms involving the
Allen EA, Ali SZ, Mathew S. Lymphoid lesions of the parotid. Diagn Cyto- salivary glands. A report of 36 cases. Am J Clin Pathol 2000;113:21–
pathol 1999;21:70–73. 28.

Ch001-F06731.indd 36 10/26/2006 10:19:59 AM


2 Thyroid
Katherine Berezowski
Mary K Sidawy
• Igor Jovanovic •

technology. Each method offers different advantages,


INTRODUCTION and the choice depends upon the familiarity and per-
sonal preference of the cytopathologist. The advantages
of air-dried smears include enhancement of background
Clinically detectable thyroid nodules occur in 4% to and cytoplasmic features and they are ideal for evaluat-
10% of the population, of which only 5% are malig- ing lymphoid infiltrates. Alcohol-fixed smears are better
nant. Fine needle aspiration (FNA) is a powerful diag- for preserving nuclear details, showing the chromatin
nostic tool recognized to have the best predictive value distribution and delineating nucleoli.
in the presurgical evaluation of the thyroid. FNA is Liquid-based preparations are advocated as an adjunct
regarded as a screening test aimed at distinguishing to conventional smears. Their exclusive use has not
nodules that require surgery from those that do not. Its gained popularity. Following each pass, one or two con-
introduction has resulted in a decrease in the number ventional smears are prepared and the remaining sample
of thyroidectomies performed on ‘cold’ nodules, and is rinsed in the appropriate liquid-based medium.
an increase in the yield of malignancy for excised The main advantages of liquid-based cytology are the
nodules. decrease in the number of slides to be screened, the
The sensitivity of thyroid FNA ranges from 83% to ability to perform immunocytochemical and other
99%, and its specificity from 70% to 91%. The reported special stains, and the enhancement of nuclear details
false negative rate ranges from 1% to 8%, but its true and irregularities in papillary carcinoma. Limitations of
frequency is difficult to assess, since only 10% of liquid-based preparation include loss of colloid during
patients with benign cytologic findings undergo processing, impacting the ability to assess the colloid to
surgery. cell ratio (Fig. 2-1). In addition, the diagnosis of Hashi-
Since FNA is considered a screening test, particular moto’s thyroiditis may be missed due to the dispersed
attention should be given to minimize false negative appearance of lymphoid cells in these preparations
results at the expense of accepting some false positive (Fig. 2-2).
diagnoses. Repeat FNA of benign lesions and periodic
clinical follow-up are advocated to decrease the false
negative diagnoses.
Accurate diagnostic interpretation depends upon an
adequate sample and optimally prepared slides. Although CHRONIC LYMPHOCYTIC
it is difficult to define what constitutes an adequate (HASHIMOTO’S) THYROIDITIS
specimen, proposed criteria require five to ten groups of
well-visualized follicular cells with each group contain-
ing 10–20 cells. Rendering a definite diagnosis on sub- CLINICAL FEATURES
optimal samples that barely meet the adequacy criteria
is a source of pitfall.
A good aspiration technique is paramount. The pro- Chronic lymphocytic (Hashimoto’s) thyroiditis is the
cedure is performed using a 23- or 25-gauge needle. The most prevalent form of autoimmune thyroid disease,
thyroid is a vascular organ, thus a few milliliters or no and is considered to be the most common cause of
suction is recommended. The needle is moved rapidly goitrous hypothyroidism in areas of the world in which
in the same needle track, and the procedure is stopped dietary iodine is sufficient. In the United States, Hashi-
as soon as the sample begins to fill the clear hub of the moto’s thyroiditis affects approximately 3% to 4% of
needle. Three or four passes are usually necessary to the population, has a female to male ratio of 8–20 : 1,
obtain an adequate sample. When cyst fluid is obtained, and is most frequently diagnosed between the third and
the cyst is drained. If any palpable lesion remains, re- fi fth decade of life. Although primarily a disease of
aspiration should be performed. Ultrasonographic guid- middle-aged women, it also occurs in children. Genetic
ance is necessary for sampling non-palpable nodules. predisposition is a risk factor. Hashimoto’s thyroiditis
The smears may be air-dried and stained with the occurs in clusters within families, and is associated
Diff-Quik stain, alcohol-fixed and stained with the with HLA-DR3 and HLA-DR5 major histocompatibil-
Papanicolaou method, or prepared using a liquid-based ity antigens.
37

Ch002-F06731.indd 37 10/26/2006 10:21:09 AM


38 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 2-1
Adenomatoid nodule: colloid to cell ratio in conventional versus liquid-based preparations. A, Conventional smear demonstrating abundant colloid
with only few scattered clusters of follicular cells (high colloid to cell ratio). Diff-Quik stain, low power. B, Liquid-based preparation of the same
case, illustrating loss of colloid during processing and concentration of the follicular cells. The colloid to cell ratio is relatively low as compared
to the conventional smear. Papanicolaou stain, low power.

A B

FIGURE 2-2
Hashimoto’s thyroiditis: lymphoid cells in conventional versus liquid-based preparations. A, A cellular streak of lymphoid cells composed of a poly-
morphous population of lymphoid cells, tingible-body macrophages, germinal centers, and plasma cells. Diff-Quik stain, high power. B, Liquid-based
preparation from the same case, showing the dispersed nature of the lymphoid cells. A close-up evaluation of the cells is necessary to distinguish
the polymorphous lymphoid nature from follicular epithelial cells and peripheral blood elements. Papanicolaou stain, high power.

The thyroid gland is diffusely slightly enlarged, non-


HASHIMOTO’S THYROIDITIS – DISEASE FACT SHEET tender to palpation, and has a rubbery to firm consis-
tency. Although the disease affects the entire gland, the
Incidence process may accentuate any asymmetry of the normal
៉ Affects 3–4% of the US population thyroid, and some patients present with an asymmetri-
cally enlarged, nodular thyroid. The majority of the
Gender, Race, and Age Distribution patients are either euthyroid or hypothyroid. In some
៉ Female predominance cases, however, the disease may present with a transient
៉ More prevalent in Caucasians hyperthyroidism.
៉ Affects adults and children Patients with Hashimoto’s thyroiditis are at increased
risk of developing non-Hodgkin B-cell lymphomas, par-
Clinical Features ticularly large B-cell type. Also, of note is the association
៉ Diffusely enlarged gland, occasionally nodular of Hashimoto’s thyroiditis and papillary carcinoma of
៉ Painless the thyroid; however, the causal relationship between
៉ Transient hyperthyroidism, euthyroid, or hypothyroid
the two has not been established.
៉ Increased risk for non-Hodgkin B-cell lymphoma

Ch002-F06731.indd 38 10/26/2006 10:21:10 AM


CHAPTER 2 Thyroid 39

CYTOPATHOLOGIC FEATURES defined cytoplasmic borders. Their nuclei are enlarged


with variable degrees of pleomorphism. Nucleoli vary
from inconspicuous to prominent (Fig. 2-4). The lym-
FNA is a reliable technique in the initial diagnosis of phoid cell population is polymorphous, consisting of
Hashimoto’s thyroiditis. It is also instrumental in small, mature lymphocytes, centroblasts, centrocytes,
establishing the diagnosis in patients with negative or and immunoblasts. Germinal centers, tingible-body
low autoantibody titers, which occurs in up to 10% of macrophages, plasma cells, and multinucleated giant
cases, and in investigating potentially neoplastic domi- cells may be present (Fig. 2-5). Epithelioid histiocytes
nant nodules. may be seen in small aggregates and may resemble gran-
The aspirates are typically cellular and contain an ulomas. They occasionally exhibit mitoses. Lymphoid
admixture of lymphoid and epithelial cells in variable cells are delicate and may appear as crushed tangles of
proportions depending on sampling and the stage of the twisted chromatin material – ‘lymphoid tangles’ (Fig.
disease (Fig. 2-3). The follicular cells demonstrate a 2-6). Blue cytoplasmic fragments (lymphoglandular
spectrum of nuclear and cytoplasmic changes ranging bodies) derived from the breakdown of lymphoid cells
from regular follicular cells to Hürthle cells. They are are noted in the background. It is important to distin-
arranged in aggregates, sheets, and occasionally in a guish bare follicular cell nuclei from lymphocytes. The
microfollicular pattern. Hürthle cells are characterized latter retain a rim of blue cytoplasm, while the follicular
by abundant granular, dense cytoplasm with well- cells appear as well-preserved stripped nuclei. The
amount of colloid is variable. When fibrosis occurs, the
aspirates tend to be dry and hypocellular.

HASHIMOTO’S THYROIDITIS – PATHOLOGIC FEATURES

Cytopathologic Findings ANCILLARY STUDIES


៉ Moderately to highly cellular aspirates
៉ Mixture of Hürthle cells and regular follicular cells
៉ Polymorphous population of lymphoid cells The diagnosis of Hashimoto’s thyroiditis is confirmed
៉ Germinal center, tingible-body macrophages, lymphohistiocytic by the presence of antithyroid autoantibodies (antithy-
aggregates, plasma cells, multinucleated giant cells roglobulin and antithyroid peroxidase) in the serum.
Antithyroid peroxidase is more specific.
Ancillary Studies
៉ Antithyroid autoantibodies in the serum

Differential Diagnosis and Pitfalls DIFFERENTIAL DIAGNOSIS AND PITFALLS


៉ Lymphoma
៉ Hürthle cell neoplasm
The differential diagnosis of Hashimoto’s thyroiditis
depends on the cellular composition of the smears. In

FIGURE 2-3
Hashimoto’s thyroiditis. A polymor-
phous lymphoid population admixed
with Hürthle cells. Diff-Quik stain,
high power.

Ch002-F06731.indd 39 10/26/2006 10:21:11 AM


40 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-4
Hashimoto’s thyroiditis. Follicular
cells with Hürthle cell change. The
cells are arranged in sheets and
display abundant granular cyto-
plasm and nuclear pleomorphism.
Diff-Quik stain, high power.

FIGURE 2-5
Hashimoto’s thyroiditis. Liquid-
based preparation illustrating a
multinucleated giant cell. Note the
scattered lymphocytes in the back-
ground. The presence of giant cells
is a useful clue to the diagnosis of
Hashimoto’s in liquid-based prepa-
rations, since the lymphoid popula-
tion is dispersed. Papanicolaou
stain, high power.

cases where the lymphoid component predominates, and a Hürthle cell neoplasm may be problematic.
lymphoma needs to be considered. Lymphomas yield a Multiple passes increase the chance of sampling the
monotonous population of lymphoid cells, unlike the lymphoid component. Moreover, the relatively
heterogenous population of Hashimoto’s thyroiditis. cohesive nature of the Hürthle cells and the nuclear
Whenever the cytologic distinction is in question, flow pleomorphism are features more in keeping with Hashi-
cytometric analysis should be performed. The sample moto’s thyroiditis. The presence of uniform, loosely
can be obtained by simply rinsing the needle in the cohesive Hürthle cells with macronucleoli favors a
appropriate medium. neoplasm.
When the aspirates show a predominance of Hürthle
cells, the distinction between Hashimoto’s thyroiditis

Ch002-F06731.indd 40 10/26/2006 10:21:12 AM


CHAPTER 2 Thyroid 41

FIGURE 2-6
Hashimoto’s thyroiditis. Lymphoid
tangles that represent twisted chro-
matin of crushed lymphocytes, and
a cluster of Hürthle cells. Diff-Quik
stain, high power.

involves stimulation of the thyroid by TSH due to low


NODULAR GOITER levels of the thyroid hormones, which leads to follicular
cell hyperplasia and involution. Nodular goiter is the
end-stage of a diffuse goiter and is caused by the cyclic
CLINICAL FEATURES changes taking place during hyperplasia and
involution.
Nodular goiter is a disease of adults and shows a
Goiter is a clinical term that denotes enlargement of female predominance. Most patients are asymptomatic
the thyroid, which occurs due to impaired synthesis and, by definition, euthyroid. Multinodular goiter devel-
of thyroid hormones. Clinically, goiter presents in a ops over many years and is detected on routine physical
nodular or diffuse form, and is divided by the func- examination or by the patient noticing an enlargement
tional activity of the thyroid into the non-toxic and in the neck. If the goiter is large enough, it can lead to
toxic variants. Non-toxic nodular goiter is the most compressive symptoms. Patients may complain of
common form in the United States, affecting approxi- sudden pain caused by hemorrhage into a nodule.
mately 5% of the population. In essence, it is a com-
pensatory response of the gland for a decrease in
hormone secretion. The specific cause of this form of
goiter is usually unknown. The basic mechanism CYTOPATHOLOGIC FEATURES

Cytologic nomenclature of nodular goiter includes


adenomatoid, cellular adenomatoid, adenomatous,
NODULAR GOITER (ADENOMATOID NODULE) – hyperplastic, and non-neoplastic nodule. The aspirates
DISEASE FACT SHEET from nodular goiter are of low to moderate cellularity.
They demonstrate an admixture of colloid and follicu-
Incidence lar cells in variable proportion, reflecting the different
៉ Affects 5% of population phases of evolution of the disease. During the hyper-
plastic stage, follicular cells are abundant and colloid is
Gender and Age Distribution scant. As the disease progresses to the involutional
៉ Female predominance stage, follicular cells become fewer and colloid becomes
៉ Occurs in adults
abundant.
The presence of abundant colloid and a high colloid
Clinical Features
to cell ratio are extremely helpful in the cytologic diag-
៉ Asymptomatic, euthyroid nosis of goiter. Macroscopically, smeared unstained
៉ Slow growing
colloid resembles varnish. Microscopically, colloid can
៉ Sudden growth as result of hemorrhage
៉ Compressive symptoms
appear as thick amorphous material with sharply cir-
cumscribed edges or as a thin translucent film in the
background, often with folds and cracks. When diluted

Ch002-F06731.indd 41 10/26/2006 10:21:13 AM


42 FINE NEEDLE ASPIRATION CYTOLOGY

and unevenly spaced, elongated nuclei (Fig. 2-8). Tissue


NODULAR GOITER (ADENOMATOID NODULE)– fragments with supporting vascular stroma may be mis-
PATHOLOGIC FEATURES taken for papillary structures and suggest papillary car-
cinoma. However, attention to the arrangement of the
Cytopathologic Findings follicular cells within the tissue fragments (spherules
៉ High colloid to cell ratio and honeycomb sheets with maintained nuclear polar-
៉ Follicular cells arranged in honeycomb sheets, spherules, tissue ity) should prevent such a pitfall (Fig. 2-9). The follicu-
fragments, or singly lar cells may also be found dispersed singly and stripped
៉ Small round nuclei, naked nuclei, delicate cytoplasm
of their cytoplasm (Fig. 2-10). Hürthle cells with
៉ Hürthle cells
៉ Cystic change: macrophages (pigmented, multinucleated),
enlarged nuclei showing variable degrees of pleomor-
cyst-lining cells phism are seen in nodular goiter.
In Diff-Quik-stained smears, the follicular cells may
Differential Diagnosis and Pitfalls reveal abundant intracytoplasmic blue granules, which
៉ Cystic papillary carcinoma can obscure the nuclei. These granules are seen in cystic
៉ Follicular neoplasm and hemorrhagic lesions, and represent hemosiderin
៉ Follicular variant of papillary carcinoma pigments. They are not specific and may be seen in
៉ Parathyroid cyst goiter, as well as in benign and malignant neoplasms
(Fig. 2-11). These non-specific granules should be dis-
tinguished from a different type of granules, ‘paravacu-
olar granules’ (Fig. 2-12). The latter consist of small
blue granules grouped within a vacuole close to the
nucleus. They are frequently observed in non-lesional
by blood, its appearance overlaps with serum. Colloid thyroid tissue and occasionally in Hashimoto’s thyroid-
may be lost during processing, particularly with liquid- itis. When the majority of aspirated follicular cells
based preparations (see Fig. 2-1). display paravacuolar granules, the cytopathologist needs
Follicular cell nuclei are 1.5–2 times the size of to consider that the targeted lesion (especially when
mature lymphocytes. The chromatin is finely granular small) was missed and only adjacent non-lesional
and uniformly dispersed, with inconspicuous nucleoli. thyroid tissue was sampled.
The nuclei are round and may show enlargement and Hemorrhage and cystic change are quite common in
variability in size. The cytoplasm is delicate with indis- aspirates from nodular goiters. Cytologically, they mani-
tinct borders (Fig. 2-7). The follicular cells are arranged fest by the presence of histiocytes and hemosiderin-
in groups, honeycomb sheets, spherules, and tissue frag- laden macrophages (many multinucleated), cholesterol
ments. A spherule represents an intact non-neoplastic crystals, and cyst-lining cells (Figs 2-13 & 2-14). Cyst-
macrofollicle with its basement membrane. It appears lining cells appear as flat sheets of spindled to polygonal
as a round structure, with smooth borders and evenly (squamoid) cells with abundant, dense cytoplasm,
spaced nuclei. A spherule may mimic a giant cell, but enlarged pleomorphic nuclei, and prominent nucleoli
the latter can be differentiated by its irregular outline (Fig. 2-15). Bi- and multinucleation are common.

FIGURE 2-7
Adenomatoid nodule. Follicular
cells arranged in a honeycomb
sheet. The cells are evenly spaced,
with uniform round nuclei and
fine evenly dispersed chromatin.
The cytoplasm is delicate with ill-
defined borders. Papanicolaou stain,
high power.

Ch002-F06731.indd 42 10/26/2006 10:21:14 AM


A B

FIGURE 2-8
Adenomatoid nodule: spherule. A spherule represents an intact non-neoplastic macrofollicle with its basement membrane. It is a tridimensional
round structure with smooth borders and evenly spaced nuclei. The latter feature helps distinguishing a spherule from a giant cell (see Fig. 2-5).
A, Diff-Quik stain, high power. B, Papanicolaou stain, high power.

FIGURE 2-9
Adenomatoid nodule. Tissue frag-
ment with supporting vascular
stroma. Note the presence of intact
macrofollicles (spherules). Their
presence along with the maintained
nuclear polarity are helpful clues
for the diagnosis of adenomatoid
nodule. Diff-Quik stain, low power.

FIGURE 2-10
Adenomatoid nodule. Follicular
epithelial cells dispersed singly and
stripped of their delicate cyto-
plasm. The absence of a small rim
of blue cytoplasm helps distinguish
follicular cells from lymphocytes.
Diff-Quik stain, high power.

Ch002-F06731.indd 43 10/26/2006 10:21:15 AM


44 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-11
Adenomatoid nodule. Follicular
cells displaying non-specific intra-
cytoplasmic blue granules. These
granules are seen in cystic and
hemorrhagic lesions and represent
hemosiderin pigments. Diff-Quik
stain, high power.

FIGURE 2-12
Paravacuolar granules. Small blue
granules clustered within a vacuole
adjacent to the nucleus. These
granules are frequently seen in
aspirates of non-lesional thyroid
and in Hashimoto’s thyroiditis. Diff-
Quik stain, high power.

Ch002-F06731.indd 44 10/26/2006 10:21:17 AM


CHAPTER 2 Thyroid 45

FIGURE 2-13
Adenomatoid nodule with hemor-
rhage and cystic change. Liquid-
based preparation illustrating
regular follicular cells and hemo-
siderin-laden macrophages (some
multinucleated). Papanicolaou stain,
high power.

FIGURE 2-14
Cholesterol crystals. Cholesterol
crystals in a Diff-Quik-stained prep-
aration appear as negative images.
Diff-Quik stain, high power.

Ch002-F06731.indd 45 10/26/2006 10:21:19 AM


46 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-15
Adenomatoid nodule: cyst-lining
cells. Flat sheet of polygonal cells
with abundant dense cytoplasm,
enlarged pleomorphic nuclei, and
distinct nucleoli. These cells are a
source of false positive diagnosis;
they can be mistaken for ‘squamoid
cells’ seen in papillary carcinoma.
Papanicolaou stain, high power.

ANCILLARY STUDIES may lead to the erroneous diagnosis of papillary


carcinoma.

Ancillary techniques including morphometry, image FOLLICULAR NEOPLASMS


analysis, DNA measurements by flow cytometry, telom-
erase activity (by polymerase chain reaction [PCR]), The cytologic features of cellular adenomatoid
and immunocytochemistry for a variety of antigens nodules overlap with those of follicular neoplasms,
have been advocated for discriminating between benign making the distinction between the two entities unreli-
and malignant nodules; however, none of these methods able. High cellularity, scant colloid, and follicular cells
is sufficiently reliable. arranged in acini and occasional microfollicles are
shared features of both entities. When the distinction is
difficult, the aspirates fall in the indeterminate category
and are classified as ‘cellular follicular lesions’. The risk
DIFFERENTIAL DIAGNOSIS AND PITFALLS of malignancy with indeterminate thyroid cytology is
reported as 15–20%.
The differential diagnosis of nodular goiter depends on FOLLICULAR VARIANT OF PAPILLARY CARCINOMA
the stage of the disease. Cystic nodules must be differ-
entiated from cystic papillary carcinomas, while the This is a well-known pitfall, and one of the major
differential diagnosis of cellular adenomatoid (hyper- sources of false negative diagnoses in thyroid FNA. This
plastic) nodules includes follicular neoplasms and variant of papillary carcinoma reveals neoplastic cells
follicular variant of papillary carcinomas. arranged in syncytial clusters and microfollicles, and
should be considered in the differential diagnosis
CYSTIC PAPILLARY CARCINOMA of cellular follicular lesions. Attention to the nuclear
features may help in establishing the correct diagnosis,
Thirty per cent of thyroid nodules are cystic, most of or at least including papillary carcinoma in the differ-
which are nodular goiter. It is also important to point ential diagnosis.
out that a third of papillary carcinomas are cystic, while
cystic degeneration is rare in follicular, medullary, and PARATHYROID CYST
anaplastic carcinomas. The gross appearance of the
fluid (yellow or hemorrhagic) is not reliable in distin- When crystal-clear fluid is aspirated, the possibility of
guishing goiter from papillary carcinoma. To the unini- a parathyroid cyst should be considered. Measuring the
tiated, the prominent cytologic atypia and squamoid level of C-terminal/midmolecule parathyroid hormone
appearance of the cyst-lining cells of nodular goiter in the fluid helps in confirming the diagnosis.

Ch002-F06731.indd 46 10/26/2006 10:21:20 AM


CHAPTER 2 Thyroid 47

and RAS mutations are also known risk factors. This


FOLLICULAR NEOPLASMS disease commonly presents as a painless thyroid nodule
which appears ‘cold’ on radionuclide scanning. Com-
pressive symptoms such as dyspnea, dysphagia, hoarse-
CLINICAL FEATURES ness, and cough are rare at presentation and may occur
in patients with the widely invasive type. Follicular
carcinoma spreads hematogenously, to the lungs and
Follicular neoplasms include follicular adenomas and bones most commonly.
carcinomas. Follicular adenoma is defined as a benign Prognosis is related to the tumor grade, stage, and
thyroid neoplasm showing follicular cell differentia- clinical presentation. Poor prognostic indicators are
tion, which is completely encapsulated and lacks cap- high histologic grade (poorly differentiated carcinoma),
sular and vascular invasion. Follicular adenoma is the age above 45 years, tumors more than 4.0 cm in size,
most common thyroid neoplasm and may be found in extrathyroid extension, and distant metastases. Well-
up to 4% of thyroid glands at autopsy. It occurs in differentiated follicular carcinomas that are confined to
adults and shows female predominance. It presents as the thyroid have an excellent prognosis, with a 10-year
a painless, solitary thyroid nodule, rarely greater than survival rate approaching 90%, whereas widely inva-
3 cm. Most appear as ‘cold’ nodules on radionuclide sive, poorly differentiated follicular carcinomas with
scanning, but functioning or ‘hot’ adenomas do occur. distant metastases have a much worse prognosis, with
Follicular carcinoma is the second most common a 10-year survival rate of approximately 50%.
form of thyroid malignancies. It accounts for 5% to
15% of all thyroid carcinomas. Follicular carcinomas
are divided histologically into two groups: minimally
invasive (encapsulated type) and widely invasive. Fol- CYTOPATHOLOGIC FEATURES
licular carcinoma has a predilection for women, with a
peak incidence in the fifth decade of life. It is more FNA cannot separate follicular adenomas from well-
common in iodine-deficient areas. Exposure to radiation differentiated follicular carcinomas. Both entities
display similar cytologic features, and the diagnosis of
malignancy relies on the histologic demonstration of
either capsular or vascular invasion. Thus, the inclu-
FOLLICULAR NEOPLASMS – DISEASE FACT SHEET sive term follicular neoplasm is recommended in the
cytologic literature. The cytologic features of follicular
Definition neoplasms are variable, reflecting their histologic types.
៉ Term includes follicular adenomas and carcinomas However, they have in common high cellularity, scant
or absent colloid, and follicular cells with enlarged
Incidence nuclei arranged in syncytial groups displaying nuclear
៉ Adenoma: most common thyroid neoplasm crowding and overlapping. The cytoplasm varies from
៉ Carcinoma: 5–15% of thyroid carcinomas ill-defined to dense. A prominent repetitive microaci-
nar arrangement is a reliable (although non-diagnostic)
Gender and Age Distribution
៉ Female predominance
៉ Adenoma: middle age
៉ Carcinoma: older age FOLLICULAR NEOPLASMS – PATHOLOGIC FEATURES

Risk Factors for Carcinoma Cytopathologic Findings


៉ Iodine deficiency ៉ Cellular aspirates
៉ Exposure to radiation ៉ Low colloid to cell ratio
៉ Activating RAS mutations ៉ Repetitive acinar arrangement
៉ Microfollicles
Clinical Features ៉ Cytoplasm delicate, ill-defined, or dense
៉ Solitary ‘cold’ nodule ៉ Nuclei:
៉ Carcinoma: hematogenous spread (lungs, bones) ៉ Enlarged, round
៉ Loss of polarity with crowding and overlapping
Prognosis ៉ Prominent nucleoli
៉ Minimally invasive carcinoma: excellent prognosis
៉ Widely invasive carcinoma: mortality rate 80% Differential Diagnosis and Pitfalls
៉ Poor prognostic indicators of carcinoma: ៉ Cellular adenomatoid nodule versus adenoma/encapsulated
៉ Poorly differentiated carcinoma
៉ Age >45 years ៉ Adenomatoid nodule versus macrofollicular adenoma
៉ Tumor >4.0 cm ៉ Follicular variant of papillary carcinoma
៉ Extrathyroid extension ៉ Insular carcinoma versus poorly differentiated follicular
៉ Distant metastases carcinoma

Ch002-F06731.indd 47 10/26/2006 10:21:21 AM


48 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-16
Follicular neoplasm (histologically
confirmed follicular adenoma).
Repetitive microacinar pattern.
Note the nuclear crowding and
overlapping. The smear is cellular
with no colloid in the background.
Diff-Quik stain, high power.

FIGURE 2-17
Follicular neoplasm. Microfollicular
arrangement characterized by the
presence of acini with central drops
of colloid. Note the nuclear enlarge-
ment, crowding, and overlapping.
Diff-Quik stain, high power.

criterion for a neoplasm (Fig. 2-16). Acini with a central chromatin, prominent nucleoli, and the presence of
lumen containing a drop of colloid represent microfol- mitotic figures. Adenomas very rarely reveal mitoses.
licles and are characteristic of follicular neoplasms
with a microfollicular growth pattern (Figs 2-17 &
2-18). A trabecular pattern is represented by parallel
rows of follicular epithelial cells. ANCILLARY STUDIES
Poorly differentiated follicular carcinoma may be
recognized as malignant cytologically. Cytologic features
that should alert the pathologist to the possibility of There are currently no ancillary tests or markers
malignancy include nuclear pleomorphism, marked that can reliably distinguish follicular adenomas
crowding and overlapping, hyperchromasia, coarse from follicular carcinomas. Though showing initial

Ch002-F06731.indd 48 10/26/2006 10:21:21 AM


CHAPTER 2 Thyroid 49

FIGURE 2-18
Follicular neoplasm. Liquid-based
preparation illustrating microacinar
arrangement with nuclear enlarge-
ment, crowding, and overlapping.
Note the similarities with Fig. 2-16.
Based on cytologic features, this
case cannot be further classified
as benign or malignant. Surgical
follow-up demonstrated a follicular
carcinoma with capsular invasion.
Papanicolaou stain, high power.

promising results, telomerase activity (detected by


PCR), and immunostains for RET/PTC, CK19, galec- HÜRTHLE CELL NEOPLASMS
tin-3, HBME-1, and thyroid-specific transcription
factors have limited predictive value in distinguishing
follicular-patterned lesions owing to a lack of sensitiv- CLINICAL FEATURES
ity and/or specificity.

Hürthle cell neoplasms, also known as oncocytic or


oxyphilic thyroid neoplasms, are comprised exclusively
DIFFERENTIAL DIAGNOSIS AND PITFALLS or predominantly (more than 75%) of follicular cells

Overlapping cytologic features limit the distinction


between follicular neoplasms and cellular adenomatoid
(hyperplastic) nodules. Up to 15–25% of FNAs reported HÜRTHLE CELL NEOPLASMS – DISEASE FACT SHEET
as follicular neoplasms correlate with hyperplastic
nodules in subsequent thyroidectomies. Definition
Macrofollicular follicular neoplasms are difficult to ៉ Term includes Hürthle cell adenomas and carcinomas
recognize as neoplastic and may be misinterpreted as
adenomatoid nodules. The cytologic features of both Incidence
entities overlap and are characterized by the presence ៉ Accounts for 5–10% of thyroid neoplasms
of abundant colloid and regular follicular cells arranged
in honeycomb sheets. Fortunately, this misclassification Gender and Age Distribution
bears no negative impact on patient management, ៉ Female predominance
since most macrofollicular follicular neoplasms are ៉ Occurs in adults
adenomas.
Follicular variant of papillary carcinomas may be Clinical Features
misdiagnosed as follicular neoplasms. The syncytial ៉ Solid nodule
arrangement and microfollicular architectural pattern ៉ ’Cold’ on radionuclide scan
are shared by both entities. Attention to the nuclear ៉ Carcinomas: cervical lymph nodes and hematogenous spread
features, i.e. elongated nuclei with irregular nuclear
membranes and powdery chromatin, is key and should Prognosis
alert the pathologist to the possibility of papillary ៉ Poor prognostic indicators of carcinoma:
៉ Age >45 years
carcinoma.
៉ Tumor >4.0 cm
Poorly differentiated follicular carcinoma may be dif-
៉ Extrathyroid extension
ficult to distinguish from insular carcinoma. Both tumors ៉ Lymph node and visceral metastases
are positive for thyroglobulin and thyroid transcription
factor 1 (TTF-1).

Ch002-F06731.indd 49 10/26/2006 10:21:23 AM


50 FINE NEEDLE ASPIRATION CYTOLOGY

displaying cytoplasmic granularity due to the accumu-


lation of mitochondria. Hürthle cell neoplasms account HÜRTHLE CELL NEOPLASMS – PATHOLOGIC FEATURES
for 5% to 10% of all thyroid neoplasms, and include
Hürthle cell adenomas and carcinomas. According Cytopathologic Findings
to the World Health Organization classification, they ៉ Cellular aspirates
are considered as a variant of follicular thyroid ៉ Scant to absent colloid
neoplasms. ៉ Monomorphic neoplastic cells
៉ Loosely cohesive and dispersed cells
Hürthle cell neoplasms occur in adults, and have a
៉ Polygonal and ovoid cells
female predilection, with a female to male ratio of 2–
៉ Large round nuclei with prominent macronucleoli
7 : 1. Hürthle cell adenomas have a peak incidence in the ៉ Abundant granular cytoplasm with distinct borders
late forties and early fifties, whereas carcinomas tend to
affect a slightly older patient population, with a peak Ancillary Studies
incidence in the late fifties. The disease presents as a ៉ Positive for thyroglobulin, TTF-1
painless thyroid nodule which appears ‘cold’ on radio- ៉ Negative for calcitonin
nuclide scanning. Compared with conventional follicu-
lar carcinomas, Hürthle cell carcinomas show a more Differential Diagnosis and Pitfalls
aggressive clinical course. Extrathyroid extension, and ៉ Dominant Hürthle cell nodule in Hashimoto’s thyroiditis
cervical lymph node and distant metastases are com- ៉ Dominant Hürthle cell nodule in nodular goiter
monly seen at the time of diagnosis. Local recurrence ៉ Papillary carcinoma
and distant metastases occur in up to 50% of patients. ៉ Medullary carcinoma
Prognosis is related to the stage and clinical presenta-
tion. Poor prognostic indicators are: age above 45 years,
size of the tumor (more than 4.0 cm), extrathyroid
extension, and lymph node and visceral metastases. The
10-year survival rate ranges from 56% to 88%.
monotonous population of Hürthle cells arranged in
loosely cohesive clusters or dispersed singly (Fig. 2-19).
CYTOPATHOLOGIC FEATURES A microfollicular pattern may be seen. The cells are
polyhedral and have abundant granular cytoplasm with
well-defined cell borders. The nuclei are round to oval,
Similar to follicular neoplasms, FNA cannot separate enlarged, and have fine chromatin and a central promi-
Hürthle cell adenomas from carcinomas, since the diag- nent macronucleolus. Binucleation is seen frequently.
nosis of malignancy is based on the histologic demon- Although enlarged, the nuclei appear monotonous with
stration of either capsular or vascular invasion. relatively little pleomorphism (Fig. 2-20). Lymphoid
Hürthle cell neoplasms yield cellular aspirates with cells and an admixture of regular follicular cells are
scant or no colloid. The smears are composed of a notably absent.

FIGURE 2-19
Hürthle cell neoplasm. Cellular
smear composed of a monotonous
population of Hürthle cells arranged
in loosely cohesive clusters and
singly. Compare the relatively
monotous nuclei to nuclear pleo-
morphism noted in non-neoplastic
conditions (Fig. 2-4). Diff-Quik
stain, high power.

Ch002-F06731.indd 50 10/26/2006 10:21:23 AM


CHAPTER 2 Thyroid 51

A B

FIGURE 2-20
Hürthle cell neoplasm. Loosely cohesive Hürthle cells with abundant granular cytoplasm, uniformly enlarged nuclei, and prominent macronucleoli.
Binucleation is frequently seen. Surgical follow-up demonstrated Hürthle carcinoma. A, Liquid-based preparation, Papanicolaou stain, high power.
B, Diff-Quik stain, high power.

ANCILLARY STUDIES
PAPILLARY CARCINOMA

Hürthle cell neoplasms are immunoreactive for thyro-


globulin and TTF-1, and negative for calcitonin. CLINICAL FEATURES

Papillary carcinoma is the most common thyroid malig-


nancy, accounting for up to 80% of malignant thyroid
DIFFERENTIAL DIAGNOSIS AND PITFALLS

PAPILLARY CARCINOMA – DISEASE FACT SHEET


The differential diagnosis of Hürthle cell neoplasms
includes non-neoplastic Hürthle cell nodule in the
Incidence
setting of Hashimoto’s thyroiditis and nodular goiter,
៉ Most common malignant thyroid neoplasm
papillary carcinoma, and medullary carcinoma. In
Hashimoto’s thyroiditis, the Hürthle cells are cohesive,
Gender and Age Distribution
usually forming sheets, and reveal nuclear pleomor-
៉ Female predominance
phism. In addition, the aspirates typically contain a
៉ Peak incidence 35–40 years; occurs in children
mixture of Hürthle cells, regular follicular cells, and
a polymorphous population of lymphoid cells. Similar Risk Factors
to Hashimoto’s thyroiditis, aspirates from nodular
៉ Exposure to radiation
goiter yield cohesive Hürthle cells arranged in ៉ Activating BRAF mutations
clusters and sheets. Nuclear pleomorphism and ៉ RET/PTC gene rearrangement
prominent nucleoli are present, but distinct macro-
nucleoli are not present in every cell. An admixture of Clinical Features
regular follicular cells and colloid also favors the diag- ៉ Slow growing
nosis of nodular goiter. Papillary carcinoma is an ៉ Solid or cystic
important differential diagnostic consideration, since ៉ ’Cold’ on radionuclide scanning
the granular cytoplasm and the well-defined cytoplas- ៉ Cervical lymph node metastases
mic borders of Hürthle cells may easily be confused ៉ Hematogenous dissemination uncommon
with the dense cytoplasm of papillary carcinoma.
However, in contrast to papillary carcinomas, a fine Prognosis
powdery chromatin pattern, intranuclear cytoplasmic ៉ Overall prognosis excellent
inclusions, nuclear grooves, and irregular nuclear mem- ៉ Less favorable prognostic factors:
branes are not features of Hürthle cell neoplasms. ៉ Age >40 years
៉ Tumor size >4.0 cm
Medullary carcinoma is also a consideration, since a
៉ Extrathyroid extension
dispersed cellular pattern is a feature seen in both
៉ Distant metastases
neoplasms. The nuclei of medullary carcinomas display ៉ Male gender
coarsely granular, ‘salt and pepper’ chromatin and lack
macronucleoli.

Ch002-F06731.indd 51 10/26/2006 10:21:24 AM


52 FINE NEEDLE ASPIRATION CYTOLOGY

neoplasms. It occurs in adults, with a peak incidence


between 35 and 40 years. It also occurs in children, and PAPILLARY CARCINOMA – PATHOLOGIC FEATURES
accounts for 90% of all childhood thyroid malignan-
cies. Papillary carcinoma is more common in women, Cytopathologic Findings
with a female to male ratio of 3–4 : 1. Its predisposing ៉ Cellular aspirates
risk factor is exposure to radiation. ៉ Architectural features:
Papillary carcinoma presents as a solid or cystic, ៉ Papillary with/without fibrovascular cores
៉ Monolayers
slow-growing, ‘cold’ thyroid nodule. It spreads via lym-
៉ Cytologic features:
phatics, resulting in a high incidence of cervical lymph
៉ Enlarged, elongated nuclei
node metastases and multifocal involvement of the ៉ Nuclear pseudoinclusions, grooves
gland. Cervical lymphadenopathy may be the initial ៉ Marginated micronucleoli
manifestation of the disease. The involved lymph nodes ៉ Nuclear membrane irregularity
may be cystic. The incidence of lymph node metastases ៉ Pale, powdery chromatin
is inversely related to age, being more common in ៉ Dense cytoplasm, septate vacuoles
younger patients. The presence of lymph node metasta- ៉ Background features:
ses does not correlate with a worse outcome, but is ៉ ‘Bubble-gum’, ropy colloid, pink ‘blobs’
associated with an increased risk of recurrences. Infre- ៉ Multinucleated giant cells
៉ Psammoma bodies
quently, papillary carcinoma disseminates hematoge-
nously to the lungs, bones, and central nervous
Ancillary Studies
system.
៉ Immunostains: positive for TTF-1, thyroglobulin, CK19, galactin-
The overall prognosis is excellent, with a 10-year
3, and HMBE-1
survival rate exceeding 90%. Less favorable prognostic
indicators are: age above 40 years, tumor size more than
Differential Diagnosis and Pitfalls
4.0 cm, extrathyroid extension, distant metastases, and
៉ Cystic adenomatoid nodule
male gender.
៉ Adenomatoid nodule
៉ Papillary hyperplasia
៉ Follicular neoplasm
៉ Hürthle cell neoplasm
CYTOPATHOLOGIC FEATURES ៉ Hyalinizing trabecular adenoma

With adequate sampling, the diagnostic accuracy of


papillary carcinoma by FNA is over 90%. The aspi-
rates are highly cellular. The neoplastic cells are ground-glass appearance of the nuclei seen in histologic
arranged in papillary clusters, flat/folded monolayered sections is an artifact of formalin fixation and is not
sheets, cohesive groups with finger-like projections, recognized in cytologic preparations. The cytoplasm is
syncytial clusters, microfollicles, or dispersed singly. dense with well-defined borders, and may have a squa-
The distinctive papillary architecture may be difficult moid appearance (Fig. 2-25). In some cases, the cyto-
to recognize cytologically, since the classic papillae plasm contains septate vacuoles, which appear as multiple
with accompanying fibrovascular cores is not frequently small vacuoles and resemble soap bubbles (Fig. 2-26).
present. Features of papillary architecture include Colloid is dense, stringy, and intimately associated with
branching groups of cells with or without fibrovascular the neoplastic cells, with the so-called ‘bubble-gum’
cores, three-dimensional dome-shaped aggregates, appearance (Fig. 2-27). Variably sized, round pink blobs
finger-like projections, densely cohesive follicular cells of colloid may be noted. In about half of cases, multi-
with smooth external contour or peripheral palisading nucleated giant cells are present; they contain few or
(Figs 2-21 & 2-22). It is important to note that a tissue numerous nuclei and their cytoplasm contains no pig-
fragment with central branching vascular stroma does ments or vacuoles (Fig. 2-28). Their presence in the
not necessarily mean that the architecture is papillary. absence of cystic degeneration should raise the suspi-
Nodular goiter and follicular neoplasms frequently cion for papillary carcinoma. Psammoma bodies, defined
yield tissue fragments that display central branching as non-polarizable concentric calcified laminations, are
vascular stroma. seen in one-third of cases (Fig. 2-29). Dystrophic calci-
The nuclei are enlarged, ovoid, and crowded with fications need to be distinguished from psammoma
small nucleoli adherent to the nuclear membrane. They bodies, since they do not carry the same diagnostic
display longitudinal grooves and intranuclear cytoplas- significance.
mic pseudoinclusions. The latter are the result of cyto- It is important to emphasize that none of the above-
plasmic invaginations; they have well-defined margins described architectural and cytomorphologic or back-
and occupy two-thirds of the nucleus (Fig. 2-23). Pseu- ground features are unique to or diagnostic of papillary
doinclusions must be distinguished from artifactual carcinoma, since they may be present in various non-
vacuoles and red blood cells overlying nuclei. Nuclear neoplastic and neoplastic thyroid diseases. The defini-
membrane irregularities, an important characteristic tive diagnosis should be based on a constellation of
feature, are best appreciated in liquid-based prepara- features, rather than on an individual cytologic
tions (Fig. 2-24). Chromatin is pale and powdery. The criterion.

Ch002-F06731.indd 52 10/26/2006 10:21:24 AM


CHAPTER 2 Thyroid 53

FIGURE 2-21
Papillary carcinoma. Papillary struc-
ture with branching central fibro-
vascular core. A characteristic
finding, but not necessary for the
diagnosis of papillary carcinoma.
Diff-Quik stain, low power.

FIGURE 2-22
Papillary carcinoma. Three-
dimensional cohesive group of fol-
licular cells with smooth external
contour and peripheral palisading,
representing a papillary tip or ‘cap’
in papillary carcinoma. Papanico-
laou stain, high power.

Ch002-F06731.indd 53 10/26/2006 10:21:25 AM


54 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-23
Papillary carcinoma. The nuclei are
enlarged, round to oval, and display
intranuclear cytoplasmic pseudo-
inclusions. The cytoplasm is dense
with well-defined borders. Diff-Quik
stain, high power.

FIGURE 2-24
Papillary carcinoma. Neoplastic fol-
licular cells arranged in a fl at mono-
layered sheet. This arrangement
must be distinguished from the
honeycomb sheet of adenomatoid
nodule. Note the presence of nuclear
grooves and nuclear membrane
irregularities, features helpful in
establishing the correct diagnosis
of papillary carcinoma. The latter
feature is well appreciated in this
liquid-based preparation. Papanico-
laou stain, high power.

Ch002-F06731.indd 54 10/26/2006 10:21:26 AM


CHAPTER 2 Thyroid 55

FIGURE 2-25
Papillary carcinoma. The neoplastic
cells in papillary carcinoma have
dense cytoplasm with a squamoid
appearance. These cells share simi-
larities and must be distinguished
from cyst-lining cells in adenoma-
toid nodules (Fig. 2-15). Diff-Quik
stain, high power.

A B

FIGURE 2-26
Septate vacuoles. Best seen in Diff-Quik stain, septate vacuoles are small multiple intracytoplasmic vacuoles that resemble soap bubbles. They are
seen in papillary carcinoma, as well as in benign conditions. A, Papillary carcinoma with septate vacuoles. Diff-Quik stain, high power. B, Septate
vacuoles in a histologically confirmed case of papillary hyperplasia. Attention to the nuclear features is paramount in differentiating benign lesions
from papillary carcinomas. Diff-Quik stain, high power.

Ch002-F06731.indd 55 10/26/2006 10:21:28 AM


56 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-27
Papillary carcinoma. Dense, stringy
colloid with a metachromatic ‘bubble-
gum’ appearance is characteristic of
papillary carcinoma. Note the inti-
mate association of colloid with the
neoplastic cells. Diff-Quik stain,
low power.

FIGURE 2-28
Papillary carcinoma. Multinucleated
giant cell with unusual shape, dense
cytoplasm, and numerous nuclei.
Note the lack of cytoplasmic pig-
ments or vacuoles. Finding giant
cells in the absence of cystic degen-
eration or Hashimoto’s thyroiditis
should raise the suspicion for papil-
lary carcinoma. Diff-Quik stain,
high power.

Ch002-F06731.indd 56 10/26/2006 10:21:29 AM


CHAPTER 2 Thyroid 57

FIGURE 2-29
Papillary carcinoma. Psammoma
bodies are refractile concentric cal-
cified laminations. Diff-Quik stain,
high power.

FIGURE 2-30
Cystic papillary carcinoma. Three-
dimensional clusters of ‘histiocyt-
oid’ cells with scalloped borders.
The abundant hypervacuolated cyto-
plasm imparts a histiocytic appear-
ance to these neoplastic cells. Note
the presence of psammoma bodies
within the center of each group.
Liquid-based preparation, Papani-
colaou stain, high power.

Cystic papillary carcinomas comprise 25–30% of pap- be dismissed and misclassified as macrophages. It is also
illary carcinomas. They are diagnostically challenging, important to emphasize that the cyst-lining cells of
and a well-recognized source of false negative results. cystic papillary carcinoma may be difficult to distin-
Aspirates contain numerous macrophages, some pig- guish for those seen in cystic nodular goiter. Recogniz-
mented, some multinucleated, but few or no diagnostic ing the risk of false negative diagnoses in cystic lesions,
neoplastic cells. If the aspirated fluid contains no follicu- a benign diagnosis should be rendered only when
lar epithelial cells, a diagnosis of ‘cystic lesion, not follicular epithelial cell lacking atypia are adequately
further classified’ must be rendered, with the recom- sampled.
mendation to repeat the procedure. In hypocellular Follicular variant of papillary carcinoma also poses a
samples, the presence of three-dimensional clusters of diagnostic challenge. The smears may be misdiagnosed
cells with enlarged nuclei, vacuolated cytoplasm, and as cellular adenomatoid nodules or follicular neoplasms.
scalloped borders, ‘histiocytoid cells’, may be the only The aspirates reveal neoplastic cells arranged in
clue to the diagnosis (Fig. 2-30). These cells can easily monolayers, syncytial clusters, and microfollicular

Ch002-F06731.indd 57 10/26/2006 10:21:30 AM


58 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-31
Papillary hyperplasia. Papillary
tissue fragment with finger-like
projections and branching central
vascular stroma, a finding that may
lead to the erroneous diagnosis of
papillary carcinoma. The clue to the
correct diagnosis rests on the bland
nuclear features. Diff-Quik stain,
low power.

architectural pattern. Nuclear features (elongated nuclei methods is a valuable adjunct in the diagnosis of papil-
with irregular nuclear membranes) are the key in estab- lary carcinoma, but it has also been reported in insular
lishing the correct diagnosis or at least raising suspicion carcinoma, and follicular and Hürthle cell adenoma and
for papillary carcinoma. carcinoma.
The Hürthle cell variant of papillary carcinoma is
composed of neoplastic Hürthle cells with nuclear and
architectural features of papillary carcinoma.
Aggressive variants of papillary carcinoma are rarely
encountered by FNA. Tall cell variant is distinguished DIFFERENTIAL DIAGNOSIS AND PITFALLS
by neoplastic cells that are twice as tall as they are
wide. The cells are characterized by oncocytic cyto-
plasm, distinct cells borders, and ‘soap-bubble-like’ The differential diagnosis of papillary carcinoma varies
intranuclear inclusions. Columnar cell variant is com- with each histologic variant. Cystic papillary carcino-
posed of columnar cells with scant cytoplasm. The mas must be distinguished from cystic adenomatoid
nuclei are elongated, crowded, and stratified. Diffuse nodules. The possibility of papillary carcinomas is
sclerosing variant yields fibrotic stromal fragments, higher in cysts larger than 4.0 cm, hemorrhagic cysts,
psammoma bodies, squamoid cells, and a prominent and cysts that recur rapidly or repeatedly following
FNA. Hypocellular cystic samples should be carefully
lymphoid infiltrate.
examined with particular attention to any cytologic
evidence of papillary carcinoma. Psammoma bodies
and intranuclear pseudoinclusions should be
searched for.
ANCILLARY STUDIES Papillary hyperplasia is a source of pitfall. The pres-
ence of unequivocal papillary elements is not pathogno-
monic of malignancy. Papillary hyperplasia shares many
CK19, HMBE-1, and galectin-3 are helpful in the diag- cytologic features with papillary carcinoma, including
nosis of papillary carcinoma, but lack specificity and high cellularity, papillary fragments with branching
sensitivity to be reliable discriminators. For example, fibrovascular cores, septate vacuoles, and occasional
the low molecular weight CK19, strongly positive in psammoma bodies (Figs 2-26B & 2-31). The clues to the
papillary thyroid carcinoma, is shown to be also posi- correct diagnosis rest on the small, round, uniform
tive in follicular adenoma, follicular carcinoma, and nuclei and finely dispersed chromatin seen in
Hürthle cell carcinoma. In addition, CK19 is strongly hyperplasia.
positive in benign follicular epithelium of Hashimoto’s Follicular variant of papillary carcinoma displaying
thyroiditis. monolayered sheets may be misdiagnosed as adenoma-
RET/PTC gene translocation and BRAF mutations toid nodule, since the flat sheets can be interpreted as
are common genetic alterations in papillary carcinomas. honeycomb arrangements indicative of goiter. Atten-
RET/PTC expression by immunostains or molecular tion to the nuclear features (elongated and irregular

Ch002-F06731.indd 58 10/26/2006 10:21:31 AM


CHAPTER 2 Thyroid 59

FIGURE 2-32
Hyalinizing trabecular adenoma.
The neoplastic follicular cells are
associated with fibrillar metachro-
matic stroma. Diff-Quik stain, high
power.

FIGURE 2-33
Hyalinizing trabecular adenoma.
Loosely cohesive neoplastic cells
with enlarged elongated nuclei and
intranuclear pseudoinclusion. This
neoplasm needs to be distinguished
from papillary and medullary carci-
noma. Diff-Quik stain, high power.

versus round) is essential in distinguishing the two enti- neoplastic cells arranged in loose aggregates, micro-
ties. On the other hand, follicular variant of papillary follicles, or dispersed singly. The neoplastic cells are
carcinoma displaying syncytial arrangements with a typically elongated or spindle-shaped. The nuclei are
microfollicular pattern may be misdiagnosed as a follic- enlarged, ovoid, with pale chromatin, intranuclear cyto-
ular neoplasm. plasmic inclusions, and longitudinal nuclear grooves.
The dense cytoplasm with well-defined borders of Psammoma bodies may be present. A clue to the diag-
papillary carcinoma may be also be confused with the nosis is the presence of metachromatic stromal material,
appearance of Hürthle cells. which is found as deposits between the neoplastic cells
Hyalinizing trabecular adenoma of the thyroid shares (Figs 2-32 & 2-33). It differs from colloid by its distin-
cytologic features with papillary carcinoma. Aspiration guishable finely fibrillar appearance and fringed rather
of these tumors yields cellular aspirates comprised of than sharp edges.

Ch002-F06731.indd 59 10/26/2006 10:21:32 AM


60 FINE NEEDLE ASPIRATION CYTOLOGY

or in association with adrenal pheochromocytoma,


MEDULLARY CARCINOMA mucosal neuromas, gastrointestinal ganglioneuromas,
and skeletal abnormalities (MEN IIB – mucosal
neuroma syndrome, Gorlin syndrome).
CLINICAL FEATURES Patients with the sporadic form present with a soli-
tary, firm, ‘cold’ nodule. In some cases, it is accompa-
nied by intractable diarrhea or Cushing syndrome
Medullary carcinoma is a neuroendocrine malignancy secondary to the paraneoplastic secretions of hormones
arising from calcitonin-secreting C cells. It accounts and prostaglandins. Only rarely this form is clinically
for 5% to 10% of all thyroid malignancies and occurs occult. In contrast, in the familial setting, despite being
sporadically and in familial forms. The sporadic form multifocal and bilateral, medullary carcinoma is usually
accounts for 80% of all cases. The remainder occurs in asymptomatic and is discovered by screening of rela-
the setting of the multiple endocrine neoplasia (MEN) tives of patients with the disease for elevated calcitonin
syndromes MEN IIA and MEN IIB, or as an isolated levels.
familial medullary thyroid carcinoma. The familial Medullary carcinoma is an aggressive tumor with
forms are characterized by an autosomal dominant metastases to cervical lymph nodes and to distant organs,
mode of inheritance and are associated with germline particularly the lungs, liver, and bones. Local recur-
point mutations in the RET proto-oncogene, which is rence occurs in up to 35% of patients. Prognosis is
localized on chromosome 10. Medullary carcinoma related to the stage and clinical presentation. Poor prog-
shows no particular sex predilection and can occur at nostic indicators are extrathyroid extension, age above
any age. The sporadic and isolated familial forms pri- 40 years, and sporadic form. The 5-year survival rate
marily affect middle-aged adults, with a peak incidence ranges from 50% to 80%.
in the forties and fi fties. The MEN II-associated forms
affect younger patients, and may even occur during
childhood. MEN-associated forms occur either in asso-
ciation with adrenal pheochromocytoma and parathy- CYTOPATHOLOGIC FEATURES
roid chief cell hyperplasia (MEN IIA – Sipple syndrome)
The aspirates from medullary carcinoma are highly
cellular. Neoplastic cells are dispersed singly and
arranged in loose syncytial aggregates. Their cytologic
MEDULLARY CARCINOMA – DISEASE FACT SHEET appearance varies. The cells may be oval with eccentric
nuclei (plasmacytoid), large polygonal, spindle, trian-
Definition gular, or small round with scant cytoplasm (Fig. 2-34).
៉ Neuroendocrine malignancy arising from calcitonin-secreting The cytoplasm is abundant and well-preserved. Spindle
C cells cells have a centrally placed elongated nucleus and
indistinct cytoplasm. Nuclear pleomorphism, binucle-
Incidence
៉ 5–10% of thyroid malignancies
៉ Sporadic in 80%
MEDULLARY CARCINOMA – PATHOLOGIC FEATURES
Gender and Age Distribution
៉ Affects both sexes equally Cytopathologic Findings
៉ Occurs in children and adults ៉ Cellular aspirates
៉ Single cells, loose syncytial aggregates
Risk Factors ៉ Plasmacytoid, spindle, small cells
៉ Familial forms: autosomal dominant inheritance; associated with ៉ Coarsely granular, ‘salt and pepper’ chromatin pattern
germline point mutations in the RET proto-oncogene ៉ Intranuclear cytoplasmic inclusions
៉ Inconspicuous nucleoli
Clinical Features ៉ Cytoplasmic granules
៉ Sporadic form: solitary firm, ‘cold’ nodule, paraneoplastic ៉ Amyloid
syndrome
៉ Familial forms: asymptomatic, discovered by screening Ancillary Studies
៉ Aggressive course: cervical lymph node and distant metastases, ៉ Immunoreactive for calcitonin, chromogranin, TTF-1, and
local recurrence monoclonal CEA
៉ Negative for thyroglobulin
Prognosis
៉ Poor prognostic indicators: Differential Diagnosis and Pitfalls
៉ Extrathyroid extension ៉ Hürthle cell neoplasm
៉ Age >40 years ៉ Papillary carcinoma
៉ Sporadic form ៉ Anaplastic carcinoma
៉ Five-year survival is 50–80% ៉ Hyalinizing trabecular adenoma

Ch002-F06731.indd 60 10/26/2006 10:21:33 AM


CHAPTER 2 Thyroid 61

FIGURE 2-34
Medullary carcinoma. Highly cellu-
lar aspirate characterized by
dispersed neoplastic cells with
round–oval nuclei and plasmacytoid
appearance. Note the presence
of intranuclear pseudoinclusions.
Nuclear pleomorphism, binucle-
ation, and multinucleation are
frequently seen in aspirates of med-
ullary carcinoma. Diff-Quik stain,
high power.

FIGURE 2-35
Medullary carcinoma. Cellular smear
composed of spindle cells with the
characteristic ‘salt and pepper’
chromatin. Papanicolaou stain, high
power.

ation, and multinucleation are present. The nuclei have (Fig. 2-37). Congo red stain examined under polarized
a coarsely granular, ‘salt and paper’ chromatin pattern light confirms the diagnosis of amyloid.
and inconspicuous nucleoli (Fig. 2-35). Intranuclear
cytoplasmic inclusions are present in up to one-half of
cases. Intranuclear grooves are infrequently seen. Red
cytoplasmic neurosecretory granules are seen in scat- ANCILLARY STUDIES
tered cells and only appreciated in air-dried smears
(Fig. 2-36).
The presence of amyloid is variable. Amyloid appears Medullary carcinoma is negative for thyroglobulin, and
as amorphous, cotton-like, acellular material that is immunoreactive for calcitonin (Fig. 2-38), monoclonal
metachromatic in Diff-Quik and grayish-green in carcinoembryonic antigen (CEA), TTF-1, low molecu-
Papanicolaou stain. It may be confused with colloid lar weight cytokeratin, and chromogranin.

Ch002-F06731.indd 61 10/26/2006 10:21:33 AM


62 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-36
Medullary carcinoma. Conventional
air-dried smear illustrating red
cytoplasmic neurosecretory gran-
ules. Multinucleation and nuclear
pleomorphism are present. Diff-
Quik stain, high power.

FIGURE 2-37
Medullary carcinoma. Amyloid
appears as amorphous, acellular
metachromatic material. Congo red
and crystal violet stains can be con-
firmatory of its presence. Diff-Quik
stain, high power.

The detection of germline mutations in the RET anaplastic carcinoma, and hyalinizing trabecular
oncogene in the serum is highly specific and sensitive adenoma. In contrast to Hürthle cell neoplasms, macro-
in identifying patients who have or will develop familial nucleoli are not a feature of medullary carcinoma. High
forms of medullary carcinoma. It is replacing calcitonin cellularity, cellular discohesion, and intranuclear inclu-
stimulation testing as a screening method. sions are shared features with papillary carcinoma, but
other architectural, background, and nuclear features
help distinguish between the two. Anaplastic carci-
noma is a consideration when medullary carcinoma is
DIFFERENTIAL DIAGNOSIS AND PITFALLS composed of spindle cells. The spindle cell variant of
medullary carcinoma yields a more uniform population
of neoplastic cells and lacks necrosis. Hyalinizing
The differential diagnosis of medullary carcinoma trabecular adenoma and medullary carcinoma are
includes Hürthle cell neoplasm, papillary carcinoma, both characterized by high cellularity, poor cellular

Ch002-F06731.indd 62 10/26/2006 10:21:35 AM


CHAPTER 2 Thyroid 63

FIGURE 2-38
Medullary carcinoma. Calcitonin
immunocytochemical stain per-
formed on a liquid-based prepara-
tion demonstrates positive staining
in the neoplastic cells. Calcitonin
immunostain, high power.

cohesion, spindle cells, and intranuclear inclusions.


Moreover, the stromal material in hyalinizing trabecu- INSULAR CARCINOMA – DISEASE FACT SHEET
lar adenoma may be misinterpreted as amyloid. A nega-
tive Congo red and calcitonin immunostain should Definition
assist in excluding medullary carcinoma. ៉ Poorly differentiated malignancy of follicular cell origin

Incidence
៉ Very rare in the United States

INSULAR CARCINOMA Gender and Age Distribution


៉ Female predominance
៉ Peak incidence 55 years
CLINICAL FEATURES
Risk Factors
៉ History of papillary or follicular carcinoma
Insular carcinoma is classified as a variant of poorly
differentiated carcinomas. It is a thyroid malignancy of Clinical Features
follicular cell origin with a biologic behavior between ៉ Aggressive malignancy
well-differentiated carcinoma (papillary, follicular car- ៉ Enlarging, ‘cold’ thyroid mass
cinoma) and anaplastic carcinoma. Insular carcinoma ៉ Extrathyroid extension, cervical lymph node and distant
has a distinct histologic appearance with neoplastic metastases
cells arranged in well-formed nests or islands (‘insulae’). ៉ Local recurrences
Its prevalence varies among different geographic ៉ Poor prognosis
regions. The highest prevalence, 5% of all thyroid
malignancies, has been reported from Italy, whereas in
the Unites States it is very low. The disease presents in
middle-aged and elderly patients, with a mean age of 55
years, and is more common in women, with a female The median survival is 3.9 years, and the 5-year sur-
to male ratio of 2 : 1. vival rate is 40–50%.
Insular carcinoma arises de novo or transforms from
well-differentiated thyroid carcinomas. It presents as an
enlarging thyroid mass which appears ‘cold’ on radio-
nuclide scanning. Extrathyroid extension, and cervical CYTOPATHOLOGIC FEATURES
lymph node and distant metastases are commonly
present at the time of diagnosis.
Insular carcinoma is an aggressive malignancy with The aspirates from insular carcinomas are highly
a poor prognosis. Recurrences develop in 60% of cases. cellular, revealing neoplastic cells arranged in loose

Ch002-F06731.indd 63 10/26/2006 10:21:36 AM


64 FINE NEEDLE ASPIRATION CYTOLOGY

DIFFERENTIAL DIAGNOSIS AND PITFALLS


INSULAR CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings The diagnosis of malignancy is easy to establish in


៉ Cellular aspirates FNA of insular carcinoma. The differential diagnosis
៉ Loose clusters, microfollicles, single cells includes poorly differentiated follicular carcinoma,
៉ Hyperchromatic small neoplastic cells
anaplastic carcinoma, medullary carcinoma, and meta-
៉ Increased N/C ratio
៉ Mild to moderate anisokaryosis
static carcinoma. The microfollicular architecture of
៉ Mitoses, necrosis insular carcinoma is a feature shared with follicular
carcinoma. The high nuclear grade, mitotic figures, and
Differential Diagnosis and Pitfalls necrosis may suggest anaplastic carcinoma, but insular
៉ Follicular carcinoma carcinoma lacks the spindle cells, osteoclast-like giant
៉ Medullary carcinoma cells, and bizarre-appearing neoplastic cells of ana-
៉ Anaplastic carcinoma plastic carcinoma. Medullary carcinoma may show
៉ Metastatic carcinoma striking morphologic overlap, but can be distinguished
by immunocytochemical stains (calcitonin-positive/
thyroglobulin-negative). Metastatic carcinoma to the
thyroid should be also considered. The patient’s clini-
cal history and comparison with previous cytologic or
clusters, trabeculae, microfollicles, or dispersed singly. histologic material are recommended.
The cells are relatively small and monomorphic, but
scattered large pleomorphic cells can also be present.
They display an increased nuclear to cytoplasmic
(N/C) ratio, irregular nuclei, hyperchromasia, and ANAPLASTIC CARCINOMA
inconspicuous nucleoli (Figs 2-39 & 2-40). Intranuclear
cytoplasmic pseudoinclusions and intranuclear grooves
are occasionally seen. The cytoplasm is pale and poorly
defined, and occasional intracytoplasmic vacuoles are CLINICAL FEATURES
present. Mitotic figures and necrosis are present.
Anaplastic carcinoma is a highly malignant, undiffer-
entiated thyroid neoplasm that accounts for up to 5%
ANCILLARY STUDIES of all thyroid malignancies. It is a disease of older
adults, with a peak incidence in the sixth and seventh
decades, and is more common in women, with a female
Insular carcinoma is immunoreactive for thyroglobulin to male ratio of 2–3 : 1.
and TTF-1, and negative for calcitonin, chromogranin, Anaplastic carcinoma tends to arise in a background
and monoclonal CEA. of thyroid disease. A history of pre-existing goiter or

FIGURE 2-39
Insular carcinoma. Atypical follicu-
lar cells forming microacini and
showing nuclear crowding and over-
lapping. Note the enlarged nuclei
as compared to adjacent red blood
cells. Diff-Quik stain, high power.

Ch002-F06731.indd 64 10/26/2006 10:21:37 AM


CHAPTER 2 Thyroid 65

FIGURE 2-40
Insular carcinoma. A crowded
cluster of neoplastic cells with
a high N/C ratio and hyperchromatic
nuclei. Papanicolaou stain, high
power.

the majority of patients are inoperable due to extensive


ANAPLASTIC CARCINOMA – DISEASE FACT SHEET local disease. The median survival is 3 to 4 months, and
the 5-year survival rate ranges from 5% to 10%.
Definition
៉ Undifferentiated thyroid neoplasm

Incidence CYTOPATHOLOGIC FEATURES


៉ 5% of thyroid malignancies

Gender and Age Distribution The cytologic features of anaplastic carcinoma vary,
៉ Female predominance
reflecting the histologic diversity of this malignancy.
៉ Occurs in the elderly The smears are variably cellular, and display neoplastic
cells that are unequivocally malignant. The cells are
Risk Factors arranged in large fragments, small clusters, or dispersed
៉ Pre-existing goiter singly. Spindle-shaped cells admixed with polygonal
៉ Well-differentiated thyroid carcinoma and giant cells are typically seen (Fig. 2-41). Some
anaplastic carcinomas contain large numbers of
Clinical Features osteoclast-like giant cells (Fig. 2-42); in others, the
៉ One of the most aggressive malignancies neoplastic cells demonstrate squamous differentiation.
៉ Rapidly enlarging, widely invasive
៉ Compression symptoms due to extrathyroid extension and invasion
of adjacent tissues present at the time of diagnosis
៉ Cervical lymph node and distant visceral metastases
ANAPLASTIC CARCINOMA – PATHOLOGIC FEATURES
៉ Prognosis extremely poor

Cytopathologic Findings
៉ Variably cellular aspirates
៉ Spindle, polygonal cells
well-differentiated (papillary or follicular) thyroid car- ៉ Osteoclast-like giant cells
cinoma is common. The disease presents as a rapidly ៉ Squamous differentiation
enlarging thyroid mass which appears ‘cold’ on radio- ៉ Marked nuclear pleomorphism, mitotic figures
៉ Background necrosis and acute infl ammation
active iodine scan. The carcinoma spreads beyond the
thyroid capsule into the adjacent neck structures,
Differential Diagnosis and Pitfalls
causing compression and invasion symptoms such as
៉ Granulomatous infl ammation
dyspnea, dysphagia, hoarseness, and cough. Metastases
៉ Medullary carcinoma
to neck lymph nodes and distant organs, particularly the
៉ Metastases
lungs and liver, are common. ៉ Sarcoma
Anaplastic carcinoma is a fatal disease. It does not
respond well to either radiation or chemotherapy, and

Ch002-F06731.indd 65 10/26/2006 10:21:38 AM


66 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 2-41
Anaplastic carcinoma. Highly atypi-
cal spindle and polygonal cells with
bizarre nuclei and prominent nucle-
oli. Diff-Quik stain, high power.

FIGURE 2-42
Anaplastic carcinoma. Osteoclast-
like giant cells can be numerous
in anaplastic carcinoma. Diff-Quik
stain, high power.

The nuclei are very large, have irregular membranes, of a rapidly enlarged thyroid in an older patient reveals
and display coarsely granular chromatin and macro- rare pleomorphic cells in a necroinflammatory back-
nucleoli. The cytoplasm varies in appearance and it ground, the diagnosis of anaplastic carcinoma must be
can be dense, granular, or vacuolated. Mitotic figures considered.
including bizarre forms are readily identified. The
background reveals necrosis and acute inflammation
(Fig. 2-43).
The cytologic diagnosis is not always straightfor- ANCILLARY STUDIES
ward. Extensive desmoplasia yields hypocellular samples
with marked crush artifact which are therefore not diag-
nostic. Necrosis and marked inflammation may also Cytokeratin positivity is observed in some anaplastic
obscure the neoplastic cells, making the interpretation carcinomas, and the vast majority are negative for
of the aspirate difficult. Nevertheless, when an aspirate TTF-1 and thyroglobulin.

Ch002-F06731.indd 66 10/26/2006 10:21:39 AM


CHAPTER 2 Thyroid 67

FIGURE 2-43
Anaplastic carcinoma. Necrotic
background with no viable tumor
cells. Diff-Quik stain, low power.

FIGURE 2-44
Metastatic colonic adenocarcinoma.
The tumor cells are columnar, and
display nuclear palisading and
stratification. Glandular arrange-
ment is prominent. Colloid is absent.
Diff-Quik stain, high power.

DIFFERENTIAL DIAGNOSIS AND PITFALLS immunoreactivity for calcitonin, chromogranin, and


monoclonal CEA support the diagnosis of medullary
carcinoma. Metastases to the thyroid are rarely encoun-
The smears are usually diagnostic of malignancy; tered by FNA. Malignant melanoma, lung, breast,
however, the presence of giant cells associated with kidney, pancreas, and colon are the most common
necrosis may be misinterpreted as granulomatous primary sites. Fig. 2-44 illustrates a case of metastatic
inflammation. The differential diagnosis of anaplastic colonic carcinoma. The differential diagnosis rests on
carcinoma includes medullary carcinoma, metastatic the patient’s clinical history, comparison with any
carcinoma, and sarcoma. Medullary carcinoma available previous cytologic or histologic material,
displays a lesser degree of cellular pleomorphism; and performing the appropriate immunocytochemical
moreover, the readily identifiable mitoses and necro- panel. Sarcoma, although rare in the thyroid, should be
inflammatory background seen in anaplastic carcinoma considered, and can be excluded by the demonstration
are absent. In addition, the presence of amyloid and of cytokeratin positivity.

Ch002-F06731.indd 67 10/26/2006 10:21:40 AM


68 FINE NEEDLE ASPIRATION CYTOLOGY

with early stage tumors confined to the gland have a


LYMPHOMA good prognosis, with a 5-year survival rate approaching
80%, whereas patient whose tumors either are of high
stage or have a DLBL component have a significantly
CLINICAL FEATURES poorer prognosis.

Lymphoma may involve the thyroid as part of systemic


lymphoma (secondary thyroid lymphoma) or may arise CYTOPATHOLOGIC FEATURES
primarily in the gland (primary thyroid lymphoma).
Primary lymphomas of the thyroid are rare, and account The aspirates are highly cellular and comprised of
for up to 2% of thyroid malignancies, whereas second- atypical lymphoid cells (Fig. 2-45). The cytologic fea-
ary involvement of the thyroid occurs more frequently. tures depend on the type of lymphoma. DLBL yield
More than 90% of primary thyroid lymphomas are cellular aspirates comprised of numerous monomor-
non-Hodgkin B-cell lymphomas. Diffuse large B-cell phic, large, atypical lymphoid cells with large nuclei,
lymphoma (DLBL), extranodal marginal zone B-cell either cleaved or non-cleaved, displaying a vesicular
lymphoma of mucosa-associated lymphoid tissue chromatin pattern and conspicuous marginated nucle-
(MZBL), and mixed MZBL/DLBL are the most common oli. Large, abnormal plasmacytoid lymphocytes with
variants. Primary thyroid T-cell lymphomas are rare. single macronucleoli may be seen. Extensive karyor-
Hodgkin lymphoma involving the thyroid is almost rhexis is present in the background. MZBL aspirates
always by direct extension from a lymph node or are comprised of a polymorphous lymphocytic popula-
thymus. tion which includes atypical small and intermediate
Primary thyroid lymphoma is a disease of older size lymphocytes, monocytoid B cells, immunoblasts,
adults, with a mean age of presentation of 65 years, and and plasma cells. Mixed DLBL/MZBL lymphomas
is more common in women, with a female to male ratio show features of both.
of 3–7 : 1. Its major risk factor is Hashimoto’s thyroid-
itis. The disease classically presents as a noticeably
enlarging thyroid mass which appears ‘cold’ on radio-
active iodine scan. Compressive symptoms such as ANCILLARY STUDIES
dyspnea, dysphagia, hoarseness, and cough due to extra-
thyroid extension are present in about one-third of
patients at the time of diagnosis. Because of the strong Immunocytochemical staining for leukocyte common
association with Hashimoto’s thyroiditis, patients may antigen (CD45) and CD20, as well as flow cytometry
present with hypothyroidism. and molecular studies are recommended to confirm the
The prognosis primarily depends on the stage and diagnosis.
histologic type of the disease. Patients with MZBL and

DIFFERENTIAL DIAGNOSIS AND PITFALLS


LYMPHOMA – DISEASE FACT SHEET

Definition The main differential diagnosis is Hashimoto’s


៉ Primary lymphoid thyroid malignancy thyroiditis. DLBL is usually not a diagnostic challenge
៉ >90% are non-Hodgkin lymphoma

Incidence
LYMPHOMA – PATHOLOGIC FEATURES
៉ 2% of malignant thyroid neoplasms

Cytopathologic Findings
Gender and Age Distribution
៉ DLBL: monotonous population of large lymphoid cells
៉ Female predominance ៉ MZBL: centrocytes, monocytoid B cells, immunoblasts, plasma
៉ Mean onset: 65 years cells
៉ Mixed DLBL/MZBL: features of DLBL and MZBL
Risk Factor
៉ Hashimoto’s thyroiditis Ancillary Studies
៉ Flow cytometry
Clinical Features ៉ Molecular studies
៉ Rapidly enlarging thyroid mass ៉ Immunocytochemistry: LCA, CD20+
៉ Compression symptoms due to extrathyroid extension
Differential Diagnosis and Pitfalls
Prognosis ៉ Hashimoto’s thyroiditis
៉ Depends on stage and histologic type ៉ Metastatic small cell carcinoma

Ch002-F06731.indd 68 10/26/2006 10:21:41 AM


CHAPTER 2 Thyroid 69

FIGURE 2-45
Diffuse large B-cell lymphoma. Cel-
lular aspirate illustrating a monoto-
nous population of atypical large
lymphoid cells. Tingible-body mac-
rophages are present. Diff-Quik
stain, high power.

and can be easily distinguished. However, distinguishing Powers CN, Frable WJ. Thyroid and parathyroid. In: Fine Needle Aspiration
MZBL from Hashimoto’s thyroiditis can be difficult on Biopsy of the Head and Neck, 1st ed. Boston: Butterworth-Heinemann,
1996.
cytologic grounds alone. Flow cytometric analysis, im-
munocytochemistry, and molecular diagnostic methods
Hashimoto’s Thyroiditis
are very helpful in establishing the diagnosis.
Metastatic small cell carcinoma of the lung should Holm LE, Blomgren H, Lowhagen T. Cancer risks in patients with chronic
also be considered as a differential diagnosis. Small cell lymphocytic thyroiditis. N Engl J Med 1985;312:601–604.
Guarda LA, Baskin HJ. Inflammatory and lymphoid lesions of the thyroid
carcinoma of the lung yields aspirates composed of gland. Cytopathology by fine-needle aspiration. Am J Clin Pathol
loosely cohesive clusters of small to intermediate-sized 1987;87:14–22.
cells with little or no cytoplasm, and nuclei displaying McDonald L, Yazdi HM. Fine needle aspiration biopsy of Hashimoto’s
hyperchromasia, inconspicuous nucleoli, and promi- thyroiditis. Sources of diagnostic error. Acta Cytol 1999;43:400–406.
nent molding. Lymphoglandular bodies are absent from Nguyen GK, Ginsberg J, Crockford PM, Villanueva RR. Hashimoto’s thyroid-
itis: cytodiagnostic accuracy and pitfalls. Diagn Cytopathol 1997;16:
the background. 531–536.

Nodular Goiter
SUGGESTED READINGS Abbas G, Heller KS, Khoynezhad A, Dubner S, Sznyter LA. The incidence
Introduction of carcinoma in cytologically benign thyroid cysts. Surgery 2001;
130:1035–1038.
Cibas ES. Thyroid. In: Cibas ES, Ducatman BS, eds. Cytology: Diagnostic Caraway NP, Sneige N, Samaan NA. Diagnostic pitfall in thyroid fine needle
Principles and Clinical Correlates, 2nd ed. Philadelphia: WB Saunders, aspiration: a review of 394 cases. Diagn Cytopathol 1993;9:345–350.
2003. Dwarakanathan AA, Staren ED, D’Amore MJ, Kluskens LF, Martirano M,
DeMay RM. Thyroid. In: The Art and Science of Cytopathology. Chicago: Economou EG. Importance of repeat fine-needle biopsy in the manage-
ASCP Press, 1996. ment of thyroid nodules. Am J Surg 1993;166:350–352.
Frost AR, Sidawy MK. Thyroid aspiration cytopathology. In: Atkinson BF, Faquin WC, Cibas ES, Renshaw AA. ‘Atypical’ cells in fine-needle aspiration
Silverman JF, eds. Atlas of Difficult Diagnoses in Cytopathology, 1st ed. biopsy specimens of benign thyroid cysts. Cancer (Cancer Cytopathol)
Philadelphia: WB Saunders, 1998. 2005;105:71–79.
Frost AR, Sidawy MK, Ferfelli M, Tabbara SO, Bronner N, Sherman ME. Frost AR, Sidawy MK. The effect of suppressive therapy on thyroid fine
Utility of thin-layer preparations in thyroid fine-needle aspiration: needle aspiration cytology. Acta Cytol 1995;39:402–408.
diagnostic accuracy, cytomorphology, and optimal sample preparation. Greaves TS, Olvera M, Florentine BD, et al. Follicular lesions of the thyroid:
Cancer (Cancer Cytopathol) 1998;84:17–25. a 5-year fine-needle aspiration experience. Cancer (Cancer Cytopathol)
Kini SR. Thyroid. In: Kline TS, ed. Guides to Clinical Aspiration Biopsy, 2000;90:335–341.
2nd edn. New York: Igaku-Shoin, 1996. Harach HR, Zusman SB, Saravia Day E. Nodular goiter: a histo-cytological
Merino MJ, Sidawy MK. The thyroid gland. In: Silverberg SG, DeLelllis RA, study with some emphasis on pitfalls of fine-needle aspiration cytology.
Frable WJ, eds. Principles and Practices of Surgical and Cytopathology, Diagn Cytopathol 1992;8:409–419.
3rd ed. New York: Churchill Livingstone, 1997. Nassar A, Gupta P, LiVolsi VA, Baloch Z. Histiocytic aggregates in benign
Miller JM, Hamburger JI, Kini SR. The impact of needle biopsy on the nodular goiter mimicking cytologic features of papillary thyroid carci-
preoperative diagnosis of thyroid nodules. Henry Ford Hosp Med J noma. Diagn Cytopathol 2003;29:243–245.
1980;28:145–148. Sidawy MK, Costa M. The significance of paravacuolar granules of the
The Papanicolaou Society of Cytopathology Task Force on Standards of thyroid. A histologic, cytologic and ultrastructural study. Acta Cytol
Practice. Guidelines of the Papanicolaou Society of Cytopathology for 1989;33:929–933.
the examination of fine-needle aspiration specimens from thyroid Sidawy MK, Del Vecchio DM, Knoll SM. Fine needle aspiration of
nodules. Mod Pathol 1996;9:710–715. thyroid nodules: correlation between cytology and histology and

Ch002-F06731.indd 69 10/26/2006 10:21:41 AM


70 FINE NEEDLE ASPIRATION CYTOLOGY

evaluation of discrepant cases. Cancer (Cancer Cytopathol) 1997;81: thyroid carcinoma in cytologic specimens. Diagn Cytopathol 2002;27:
253–259. 143–148.
Tabbara SO, Acoury N, Sidawy MK. The origin of multinucleated foreign-
body-type giant cells in thyroid neoplasms: a cytologic, histologic and
Follicular Neoplasms immunohistochemical study. Acta Cytol 1996;40:1184–1188.
Baloch ZW, LiVolsi VA. Follicular-patterned lesions of the thyroid: the bane
of the pathologist. Am J Clin Pathol 2002;117:143–150.
Medullary Carcinoma
Baloch ZW, LiVolsi VA. The quest for a magic tumor marker. Continuing
saga in the diagnosis of the follicular lesions of the thyroid (Editorial). Bhattacharyya N. A population-based analysis of survival factors in dif-
Am J Clin Pathol 2002;118:165–166. ferentiated and medullary thyroid carcinoma. Otolaryngol Head Neck
Cerilli LA, Mills SE, Rumpel CA, Dudley TH, Moskaluk CA. Interpretation of Surg 2003;28:115–123.
RET immunostaining in follicular lesions of the thyroid. Am J Clin Pathol Bose S, Kapila K, Verma K. Medullary carcinoma of the thyroid: a cytologi-
2002;118:186–193. cal, immunocytochemical, and ultra-structural study. Diagn Cytopathol
DeMay RM. Follicular lesions of the thyroid. W(h)ither follicular carci- 1992;8:28–32.
noma? Am J Clin Pathol 2000;114:681–683. Collins BT, Cramer JM, Tabatowski K, Hearn S, Raminhos A, Lampe H. Fine
Kroll TG. Molecular rearrangements and morphology in thyroid cancer. Am needle aspiration of medullary carcinoma of the thyroid. Cytomorphol-
J Pathol 2002;160:1941–1944. ogy, immunocytochemistry and electron microscopy. Acta Cytol
LiVolsi VA, Asa SL. The demise of follicular carcinoma of the thyroid gland. 1995;39:920–930.
Thyroid 1994;4:233–236. Forrest CH, Frost FA, de Boer WB, Spagnolo DV, Whitaker D, Sterrett BF.
Segev DL, Clark DP, Zieger M, Umbricht C. Beyond the suspicious thyroid Medullary carcinoma of the thyroid: accuracy of diagnosis of fine-needle
fine needle aspirate. A review. Acta Cytol 2003;47:709–722. aspiration cytology. Cancer 1998;84:295–302.
Shaha AR, Loree TR, Shah JP. Prognostic factors and risk group analysis Kini SR, Miller JM, Hamburger JI, Smith MJ. Cytopathologic features of
in follicular carcinoma of the thyroid. Surgery 1995;118:1131–1136. medullary carcinoma of the thyroid. Arch Pathol Lab Med 1984;108:
Umbricht CB, Conrad GT, Clark DP, et al. Human telomerase reverse tran- 156–159.
scriptase gene expression and the surgical management of suspicious
thyroid tumors. Clin Cancer Res 2004;10:5762–5768.
Insular Carcinoma
Hürthle Cell Neoplasms Guiter GE, Auger M, Ali SZ, Allen EA, Zakowski MF. Cytopathology of insular
carcinoma of the thyroid. Cancer 1999;87:196–202.
Chen KTK. Fine-needle aspiration cytology of papillary Hurthle-cell tumors Layfield LJ, Gopez EV. Insular carcinoma of the thyroid: report of a case
of thyroid: a report of three cases. Diagn Cytopathol 1991;7:53–56. with intact insulae and microfollicular structures. Diagn Cytopathol
Gonzalez JL, Wang HH, Ducatman BS. Fine-needle aspiration of Hurthle 2000;23:409–413.
cell lesions: a cytomorphologic approach to diagnosis. Am J Clin Pathol Pietribiasi F, Sapino A, Papotti M, Bussolati G. Cytologic features of poorly
1993;100:231–235. differentiated ‘insular’ carcinoma of the thyroid, as revealed by fine-
Lopez-Penabad L, Chiu AC, Hoff AO, et al. Prognostic factors in patients needle aspiration biopsy. Am J Clin Pathol 1990;94:687–692.
with Hurthle cell neoplasms of the thyroid. Cancer 2003;97:1186– Pilotti S, Collini P, Mariani L et al. Insular carcinoma: a distinct de novo
1194. entity among follicular carcinomas of the thyroid gland. Am J Surg
McIvor NP, Freeman JL, Rosen I, Bedard YC. Value of fine-needle aspiration Pathol 1997;21:1466–1473.
in the diagnosis of Hurthle cell neoplasm. Head Neck 1993;15: Sironi M, Collini P, Cantaboni A. Fine needle aspiration cytology of insular
335–341. thyroid carcinoma. A report of four cases. Acta Cytol 1992;36:435–439.
Stojadinovic A, Ghossein RA, Hoos A, et al. Hurthle cell carcinoma: a
critical histopathologic appraisal. J Clin Oncol 2001;19:2616–2625.
Stojadinovic A, Hoos A, Ghossein RA, et al. Hurthle cell carcinoma: a Anaplastic Carcinoma
60-year experience. Ann Surg Oncol 2002;9:197–203.
Berry B, MacFarlane J, Chan N. Osteoclastomalike anaplastic carcinoma of
the thyroid. Diagnosis by fine needle aspiration cytology. Acta Cytol
Papillary Carcinoma 1990;34:248–250.
Brooke PK, Hameed M, Zakowski M. Fine-needle aspiration of anaplastic
Baloch ZW, Abraham S, Roberts S, et al. Differential expression of cyto- thyroid carcinoma with varied cytologic and histologic patterns: a case
keratins in follicular variants of papillary carcinoma: an immunohisto- report. Diagn Cytopathol 1994;11:60–63.
chemical study and its diagnostic utility. Hum Pathol 1999;30: Hundahl SA, Fleming ID, Fremgen AM, Menck HR. A National Cancer Data
1166–1171. Base report on 53,856 cases of thyroid carcinoma treated in the U.S.,
Chan JKC. Strict criteria should be applied in the diagnosis of encapsu- 1985–1995. Cancer 1998;83:2638–2648.
lated follicular variant of papillary thyroid carcinoma. Am J Clin Pathol Peterson CE, Hall W, Baskin HJ. Anaplastic thyroid carcinoma: cytomor-
2002;117:16–18. phology and clinical implications of fine-needle aspiration. Diagn
Goellner JR, Carney JA. Cytologic features of fine-needle aspirates of Cytopathol 1991;7:63–67.
hyalinizing trabecular adenoma of the thyroid. Am J Clin Pathol 1989; Venkatesh YS, Ordonez NG, Schultz PN, et al. Anaplastic carcinoma of the
91:115–119. thyroid. A clinicopathologic study of 121 cases. Cancer 1990;66:
Goodell WM, Saboorian MH, Ashfaq R. Fine-needle aspiration diagnosis of 321–330.
the follicular variant of papillary carcinoma. Cancer 1998;84:349–354.
Lin JS, Komisar A, Opher E, Blaugrund SM. Follicular variant of papillary
carcinoma: the diagnostic limitations of preoperative fine-needle Lymphoma
aspiration and intra-operative frozen section evaluation. Laryngoscope
2000;110:1431–1436. Das DK, Gupta SK, Francis IM, Ahmed MS. Fine-needle aspiration cytology
LiVolsi VA. Unusual variants of papillary thyroid carcinoma. Adv Endocrinol diagnosis of non-Hodgkin lymphoma of thyroid: a report of four cases.
Metab 1995;6:39–54. Diagn Cytopathol 1993;9:639–645.
Renshaw AA. ‘Histiocytoid’ cells in fine-needle aspirations of papillary Derringer GA, Thompson LD, Frommelt RA, Bijwaard KE, Heffess CS, Abbon-
carcinoma of the thyroid: frequency and significance of an under- danzo SL. Malignant lymphoma of the thyroid gland: a clinicopathologic
recognized cytologic pattern. Cancer (Cancer Cytopathol) 2002;96:240– study of 108 cases. Am J Surg Pathol 2000;24:623–639.
243. Detweiler RE, Katz RL, Alapat C, El-Naggar A, Ordonez N. Malignant lym-
Shaha AR, Shah JP, Loree TR. Risk group stratification and prognostic phoma of the thyroid: a report of two cases diagnosed by fine-needle
factors in papillary carcinoma of thyroid. Ann Surg Oncol 1996;3: aspiration. Diagn Cytopathol 1991;7:163–171.
534–538. Skacel M, Ross CW, Hsi ED. A reassessment of primary thyroid lymphoma:
Solomon A, Gupta PK, LiVolsi VA, Baloch ZW. Distinguishing tall cell high-grade MALT-type lymphoma as a distinct subtype of diffuse large
variant of papillary thyroid carcinoma from usual variant of papillary B-cell lymphoma. Histopathology 2000;37:10–18.

Ch002-F06731.indd 70 10/26/2006 10:21:43 AM


3 Lymph Nodes
Paul E Wakely Jr

Lymph nodes (or masses suspected to represent a lymph manifests as a clinically enlarged lymph node that may
node) rival the thyroid gland and the breast as the most or may not be painful upon palpation. It may be due to
frequent sites for application of fine needle aspiration a variety of antigenic stimuli including organisms,
(FNA) biopsy. FNA often unequivocally represents the foreign material (drugs, environmental pollutants,
most direct and cost-effective diagnostic path to explain altered tissue components), altered immune status, and
an enlarged lymph node. Principal indications are to enlargement in the drainage pathway of a neoplasm
exclude or confirm a benign, malignant (primary or without actually harboring neoplastic cells. In the vast
metastatic), or infectious process when the reason is majority of instances, a specific etiology remains
not clinically apparent to the physician. unknown.
Prognosis of reactive lymphoid hyperplasia is excel-
lent, with most examples resolving on their own or after
a course of antibiotic therapy. The utility of FNA is
REACTIVE LYMPHOID HYPERPLASIA particularly noticeable with persistent lymphadenopa-
thy, since the patient, parents, and clinician begin to
consider a more ominous cause if an enlarged node is
present for more than 3 weeks.
CLINICAL FEATURES

Non-specific reactive lymphoid hyperplasia is a common CYTOPATHOLOGIC FEATURES


cause of lymphadenopathy in children and young
adults, but much less so in older individuals, being very
infrequent beyond the fourth decade. Males and females Two consistent findings in cytologic smears of
are equally affected. Reactive lymphoid hyperplasia lymphoid tissue regardless of a benign or malignant
nature are: a) dispersion of cells predominantly as

REACTIVE LYMPHOID HYPERPLASIA – DISEASE FACT SHEET


REACTIVE LYMPHOID HYPERPLASIA – PATHOLOGIC FEATURES
Definition
៉ A benign proliferation of cells in the lymph node cortex, medulla, Cytopathologic Findings
and/or paracortex caused by a variety of antigenic stimuli ៉ A spectrum of lymphocytes ranging from small round forms
(7–8 μm) to transformed centrocytes and centroblasts to larger
Incidence immunoblasts
៉ Very common cause of lymphadenopathy in children and young ៉ Dendritic/lymphocytic aggregates
adults; much less so in adults ៉ Follicular center fragments consisting of a syncytium of
៉ Males = females dendritic cells populated by a range of lymphocytes, tingible-
body macrophages, and short capillary segments
Clinical Features
៉ Usually solitary node enlargement; less commonly produces Ancillary Studies
multiple enlarged nodes ៉ Immunophenotyping shows a polyclonal population of B and T
៉ Soft to palpation, non-tender, not fixed to soft tissue lymphocytes
៉ Typically involves cervical, axillary, or inguinal nodes; rarely
supraclavicular nodes Differential Diagnosis and Pitfalls
៉ Any ‘small cell’ non-Hodgkin lymphoma
Prognosis and Treatment ៉ Small cell neuroendocrine carcinoma
៉ Completely benign and reversible ៉ Certain benign causes of lymphadenopathy, e.g. progressive
៉ Self-limited; surgical excision often occurs if node is >1 cm in transformation of germinal centers, Castleman disease,
dimension and does not regress after a certain duration toxoplasma lymphadenitis, HIV lymphadenopathy

71

Ch003-F06731.indd 71 10/26/2006 10:22:48 AM


72 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 3-1
Reactive lymphoid hyperplasia. A, A polymorphous population of lympho-
cytes is present in a dissociated (single cell) pattern. Small lymphocytes
with rounded nuclei are most numerous, but large and intermediate-size
lymphocytes are also present. Note the blue globular lymphoglandular
bodies scattered between cells. Romanowsky stain, high power. B, A
centrally placed immunoblast looms over the remaining smaller lympho-
cytes in this field. Note the single nucleolus and the zone of deeply
basophilic staining at the immunoblast periphery. Romanowsky stain,
high power. C, Three tingible-body macrophages are easily recognized
because of their large size, and the presence of phagocytosed cellular
debris. A heterogeneous cell mixture is seen in the background. Papani-
colaou stain, high power.

non-clustered, individual forms – the so-called single phases), small round lymphocytes predominate with
cell/dissociated cell pattern; and b) presence of variation in the percentage of other lymphocytes. It is
individual small globular or flake-like fragments of important to remember that the spatial relationships
cytoplasm that have been termed lymphoglandular visible in tissue sections of lymph node are lost in aspi-
bodies. Lymphoglandular bodies are best seen with rates. Thus, follicular, diffuse, sinusoidal, or marginal
a Romanowsky-type stain (Giemsa, Wright-Giemsa, zone architecture cannot be recognized with FNA.
May-Grünwald-Giemsa, or Diff-Quik), where they Since the reactive node undergoes expansion of corti-
stain a pale blue or blue–gray, are about the size of a cal, medullary, and/or paracortical zones, the hallmark
red cell, and may contain small vacuoles; their size can of reactive lymphoid hyperplasia is the structural poly-
vary somewhat. Their presence, particularly in large morphism/heterogeneity typical of sampling the normal
numbers, is assurance that lymphoid tissue has been cellular elements from various parts of a normal but
aspirated, but it does not guarantee that the lesion expanded node. These constituents include small
on the slide is lymphoid. It merely states that a lym- lymphocytes, centrocytes, centroblasts, dendritic cells,
phoid population exists on the smear (e.g. metastatic tingible-body macrophages, immunoblasts, plasmacytoid
seminoma and undifferentiated nasopharyngeal carci- monocytes, plasma cells, and, less often, capillaries,
noma typically contain lymphocytes as part of the eosinophils, and endothelial cells. Small round lympho-
neoplasm). cytes prevail in aspirate smears, followed by centrocytes
Because little stroma exists in reactive lymphoid and centroblasts in examples of follicular reactive
hyperplasia to retain lymphoid cells during the back- lymphoid hyperplasia (Fig. 3-1). Centrocytes are similar
and-forth cutting action of an aspirating needle, rela- in size to or slightly larger than small lymphocytes,
tively large numbers of cells can be obtained, resulting having slight nuclear contour irregularity. Centroblasts
in moderately to highly cellular smears. Degree of cel- are non-cleaved lymphocytes (almost twice the diameter
lularity has no correlation with benignancy or malig- of mature lymphocytes) with coarser chromatin, slightly
nancy. Even if reactive lymphoid hyperplasia is more cytoplasm, and may contain visible nucleoli.
subdivided into various stages (early, mid, and late Plasmacytoid lymphocytes/monocytes are small to

Ch003-F06731.indd 72 10/26/2006 10:22:49 AM


CHAPTER 3 Lymph Nodes 73

intermediate in size with eccentrically placed nuclei, This can be accomplished either by rinsing the aspirat-
smudged chromatin, a variable perinuclear clear zone, ing needle in the media after cells are expressed onto
and a small amount of slightly basophilic cytoplasm. glass slides or by directly placing cells into such a
Immunoblasts are large cells (20–25 μm) with open fine medium from one of the FNA passes. The most impor-
chromatin, one or more distinct nucleoli, and moderate tant ancillary test for reactive lymphoid hyperplasia is
to abundant pale to basophilic cytoplasm. A perinuclear immunophenotyping. Immunophenotyping should be
clear zone may be seen. Tingible-body macrophages are performed on all lymph node aspirates that contain
large cells, with rounded nuclei, finely granular chroma- only a lymphoid population, to help assure that a
tin, a small distinct nucleolus, and a copious debris- non-Hodgkin lymphoma is excluded. Those aspirates
laden cytoplasm. Dendritic cells have oval hypochro-
matic nuclei, indistinct or absent nucleoli, and long
cytoplasmic processes; they are commonly binucleated
(Fig. 3-2). Cell processes are paradoxically better seen in
Papanicolaou-stained smears. Thus, it is not so critical in
reactive lymphoid hyperplasia that one tries to identify
each cell on the smear, rather to recognize that a range
of different lymphoid tissue cellular elements exists.
An exception to the single cell pattern in a normal
but reactive node is the presence of dendritic/lympho-
cytic aggregates and/or follicular center cell fragments.
Dendritic/lymphocytic aggregates are loose collections
(50–120 μm) of small lymphocytes and larger dendritic
cells devoid of tingible-body macrophages or capillaries
(Fig. 3-3A). The cytoplasmic processes of dendritic cells
are best seen in these aggregates. Follicular center cell
fragments contain the additional embellishments of
branching capillaries and tingible-body macrophages,
and are most often observed in examples of florid fol-
licular reactive lymphoid hyperplasia (Fig. 3-3B).

FIGURE 3-2
ANCILLARY STUDIES Reactive lymphoid hyperplasia, dendritic cells. A grouping of dendritic
cells displays the web-like cytoplasmic extensions that emanate from
the cell body. Cell borders are indistinct, creating a syncytium. Nuclei
are hypochromatic with smooth nuclear contours and small nucleoli.
All lymph node FNA biopsies should obtain cells in a Note the binucleated dendritic cell at the bottom of this group whereby
liquid medium that preserves cells for ancillary testing. the two nuclei partially overlap. Papanicolaou stain, high power.

A B

FIGURE 3-3
Reactive lymphoid hyperplasia. A, Dendritic/lymphocytic aggregate. A discrete cluster of dendritic cells and heterogeneous lymphocytes is present.
The former are distinguished by their larger nuclear size and abundant amphophilic cytoplasm that creates a syncytium. Note that the large number
of lymphocytes partially obscures these cells. B, Follicular center cell fragment. A branching capillary traverses an aggregate of heterogeneous
lymphocytes and partially hidden dendritic cells. Tingible-body macrophages are embedded within the aggregate or closely associated with it (lower
center). Romanowsky stain, high power.

Ch003-F06731.indd 73 10/26/2006 10:22:49 AM


74 FINE NEEDLE ASPIRATION CYTOLOGY

containing a metastatic cancer do not necessarily The principal diagnostic pitfall is confusing reactive
require immunophenotyping unless the clinical cir- lymphoid hyperplasia with a small cell type of non-
cumstances dictate otherwise. For reactive lymphoid Hodgkin lymphoma (NHL). The major morphologic
hyperplasia and all non-Hodgkin lymphomas, immuno- feature that separates these two categories is the pres-
phenotyping is best accomplished using flow cytometry ence of a range of lymphocytes in the former and its
(FCM). FCM is the current standard for immunologic relative absence in the latter. Most small cell lymphomas
analysis of lymphoid tissue/cells. Its advantages include are composed of monotonous (or nearly monotonous)
the ability to evaluate numerous lymphoid antigenic lymphocytes. Follicular lymphoma, marginal zone lym-
markers on a smaller number of cells, an objective phoma, and some T-cell lymphomas, however, show a
assessment of clonality, the ability to detect small degree of cell heterogeneity that allows them to be con-
monoclonal cell populations, and a rapid turnaround fused with reactive lymphoid hyperplasia. Grade II
time. A more time-consuming and less frequently used follicular lymphoma in particular often has a two-cell
method is immunocytochemistry of cytospin prepared population of small and large lymphocytes, but a true
slides. With immunocytochemistry, an objective mea- spectrum of lymphoid cells is absent. This can be diffi-
surement of clonality is lost, it is more laborious, and cult to appreciate. Thus, immunophenotyping is manda-
fewer markers are utilized. Its major advantage is the tory in all aspirates of presumptive reactive lymphoid
preservation of cell morphology, but this is offset by its hyperplasia.
disadvantages, except in cases of presumptive Hodgkin Small cell neuroendocrine carcinoma may be mis-
lymphoma. taken infrequently with reactive lymphoid hyperplasia.
In reactive lymphoid hyperplasia, FCM demonstrates It is more apt to be confused with small cell forms of
a polyclonal population of T and B lymphocytes. The NHL, and thus is discussed in detail in that section. A
FCM result combined with the appropriate morphology, cytologic diagnosis (regardless of what tissue or organ
and the proper clinical context are diagnostic of reactive is aspirated) of reactive lymphoid hyperplasia must fit
lymphoid hyperplasia. FCM is, however, imperfect. with the overall clinical context of the patient’s problem.
Some lymph node aspirates contain too few cells to If it does not, then the cytologic diagnosis must be vali-
evaluate, some B-cell lymphomas are immunoglobulin- dated using another method, usually surgical biopsy. In
negative, and some lymphomas have such a high cell the example of an individual with a large, fixed node
turnover rate that an insufficient number of viable lym- whose combined aspirate morphology and immunophe-
phocytes exist for immunophenotyping. In these cases, notyping shows reactive lymphoid hyperplasia, a cyto-
molecular studies can be employed to further identify logic diagnosis has been made that does not correlate
the lesion. with the clinical scenario. Partial involvement of a node
Molecular and genetic tests can be applied to sus- by a malignant process is a potential reason why a diag-
pended cells in a liquid medium or directly to cells nosis of reactive lymphoid hyperplasia, although techni-
smeared on a glass slide. One method uses the poly- cally correct, may be invalid in the overall clinical
merase chain reaction (PCR) that amplifies the DNA of circumstance.
lymphocytes, and can determine if the heavy chain
immunoglobulin genes of B cells or the T-cell receptor
genes of a T-cell lymphoma have been rearranged.
Another method is fluorescence in situ hybridization
(FISH), whereby commercially available probes are GRANULOMATOUS LYMPHADENOPATHY
used to label parts of a chromosome to determine
whether a cytogenetically abnormal rearrangement has
occurred. Specific examples are addressed under certain CLINICAL FEATURES
disease states.

Granulomatous lymphadenopathy is very common and


is due to various etiologies. Most examples are second-
DIFFERENTIAL DIAGNOSIS AND PITFALLS ary to infectious disease, but granulomas may appear
as a consequence of placement of foreign material,
immune-related disease, environmental toxins and
Some benign lymphadenopathies have distinctive archi- dust, or neoplasms. Sarcoidosis is a common cause of
tectural features and spatial relationships in tissue sec- granulomatous lymphadenopathy. In the United States,
tions that allow for a specific diagnosis. However, these the incidence is 40 per 100,000 in black individuals,
features are not readily transferable to smears. Thus, which is 8–10 times more common than in whites. All
benign lymphadenopathies such as Castleman disease, ages can be affected, but peak incidence is in the third
toxoplasma lymphadenitis, HIV-associated lymph- to fi fth decade, with a female predominance (2 : 1). In
adenopathy, progressive transformation of germinal addition to peripheral adenopathy, enlargement of
centers, and some examples of dermatopathic lymph- pulmonary hilar nodes is common. Patients may also
adenopathy for the most part are morphologically display signs and symptoms of uveitis, sinusitis, ery-
similar to reactive lymphoid hyperplasia smears, and thema nodosum, and joint lesions, along with elevated
are rarely distinguishable with absolute certainty by angiotensin-converting enzyme, erythrocyte sedimen-
using FNA. tation rate, and serum calcium.

Ch003-F06731.indd 74 10/26/2006 10:22:51 AM


CHAPTER 3 Lymph Nodes 75

CYTOPATHOLOGIC FEATURES
GRANULOMATOUS LYMPHADENOPATHY – DISEASE FACT SHEET

Definition Regardless of cause, the cytopathology of granuloma-


៉ The presence of granulomas with or without inflammation and tous lymphadenopathy is similar with minor differ-
necrosis in a lymph node; etiology may be infectious or ences. Granulomas appear in smears as either loose or
non-infectious
tightly clustered collections of epithelioid histiocytes of
variable number and size. The key feature of an epithe-
Incidence
lioid histiocyte (which allows its distinction from a
៉ Sarcoidosis: 40 per 100,000 in black population; 8–10 times less
macrophage or dendritic cell) is an elongated, almost
common in whites
៉ Infectious causes: common in children and immunosuppressed
elliptical or spindle-shaped nucleus. A slight smooth
patients indentation of the nuclear contour is present on one
side, producing an outline that has been variously
Gender and Age Distribution described as ‘footprint-, boomerang-, or banana-shaped’.
៉ Sarcoidosis: 20–40 years of age; women > men
Not all nuclei in a collection of epithelioid histiocytes
៉ Infectious causes: any age, with equal male/female ratio; will have this shape, but enough will to allow recogni-
dependent on immunologic status tion as a granuloma. These nuclei are hypochromatic
and may have small nucleoli or no visible nucleoli (Fig.
Clinical Features 3-4). Nuclei may project away from a granuloma center
៉ Enlarged node or group of nodes in a ‘starburst’ pattern. Epithelioid histiocytes have a
៉ Usually firm to palpation in non-infectious causes moderate to abundant amount of finely granular, non-
៉ May be tender to palpation with cutaneous erythema in infectious vacuolated cytoplasm. Multinucleated giant cells are
causes variably present in slides of granulomatous lymphade-
nopathy; their presence is not necessary for the
Prognosis and Treatment diagnosis.
៉ Usually excellent prognosis Infectious causes, particularly cat-scratch disease,
៉ Antibiotic, antifungal, antimycobacterial therapy, dependent on atypical mycobacteria, and some examples of tubercu-
etiology lous lymphadenitis, have an added neutrophilic cellular
component on the smear, and may mimic an abscess
(Fig. 3-5). Mycobacterial lymphadenitis has a variety of
appearances, with some cases displaying no granulo-
mas, only necrosis with or without acute inflammation,
or foamy macrophages containing cytoplasmic stria-
tions. The latter represent ingested mycobacterial
Patients with fungal or mycobacterial lymphadenitis organisms (Fig. 3-6).
often contain granulomas within enlarged nodes. These
can occur at any age, and are often associated with a
deficient immune status in the host. Patients may
present with non-specific lymphadenopathy, or may
have the constitutional signs and symptoms of dissemi-
nated infection. Some fungal infections such as Crypto-
coccus and Histoplasma are endemic to certain areas
of the United States. Tuberculous and non-tuberculous GRANULOMATOUS LYMPHADENOPATHY – PATHOLOGIC FEATURES
(M. avium and M. intracellulare) lymphadenitis are
particularly common in AIDS patients and others with Cytopathologic Findings
poor immune function, and in the under-developed ៉ Granulomas: syncytial clusters of epithelioid histiocytes
world. Atypical mycobacteria lymphadenitis is often a characterized by oval and elongated, indented nuclei in loose or
childhood disease with enlarged cervical nodes mimick- tight clusters
ing a bacterial infection. Cat-scratch disease is a lymph- ៉ Variable number of multinucleated giant cells
adenitis secondary to Bartonella henselae. The incidence ៉ Variable necrosis and acute infl ammation; acute infl ammation is
is 9 per 100,000 in the United States, and most patients common in cat-scratch disease and mycobacterial infection;
are less than 20 years of age. Although a high proportion absent in sarcoid
of patients are in contact with a cat, only about 50%
Ancillary Studies
report actually being scratched.
៉ Histochemical stains for fungi and/or acid-fast bacilli
Prognosis of patients with infectious granuloma-
៉ Microbiologic testing
tous lymphadenitis depends on response to antimi-
crobials, and correction of immune status. Children
Differential Diagnosis and Pitfalls
with atypical mycobacteria recover fully in nearly all
៉ Dendritic/lymphocytic aggregates
cases. Cat-scratch disease is self-limited in immuno-
៉ Suppurative lymphadenitis
competent patients. Sarcoidosis remits spontaneously ៉ Spindle cell neoplasm
in a large percentage of cases; many are treated with
steroids.

Ch003-F06731.indd 75 10/26/2006 10:22:51 AM


76 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 3-4 FIGURE 3-6


Granulomatous lymphadenopathy: sarcoidosis. This syncytial cluster of Granulomatous lymphadenopathy: mycobacterial lymphadenitis. This
epithelioid histiocytes forms a granuloma. Some hypochromatic nuclei smear from an immunosuppressed patient contained no granulomas,
are oval, but many have a markedly elongated spindled quality. Indenta- only macrophages and lymphocytes, whose nuclei are seen. The striking
tion of the nuclear contour varies from subtle to dramatic, particularly feature in this image is the randomly arranged and overlapping unstained
so in the bottom half of the image. Nucleoli are indistinct. Papanicolaou lines that exist extracellularly. These represent the acid-fast bacilli
stain, high power. whose lipid coat prevents them from taking up the stain, thus creating
the so-called ‘negative’ image. Romanowsky stain, high power.

FIGURE 3-5 FIGURE 3-7


Granulomatous lymphadenopathy: cat-scratch disease. This well-formed Granulomatous lymphadenopathy: mycobacterial lymphadenitis. Same
granuloma has frayed edges. It is infiltrated and surrounded by numer- case as seen in Fig. 3-6, now stained with an acid-fast stain highlight-
ous polymorphonuclear leukocytes that partly obscure histiocyte nuclei. ing the innumerable red rod-shaped bacilli. Culture proved to be M.
All areas of the smear are covered with neutrophils. Papanicolaou stain, avium-intracellulare. Fite stain, high power.
high power.

most direct path toward organism identification (Fig.


3-7). The diagnosis of sarcoidosis is based on ancillary
ANCILLARY STUDIES serologic tests, and clinicoradiologic examination.

The primary study for any infectious cause should be


microbiologic testing for an organism. Some of the aspi- DIFFERENTIAL DIAGNOSIS AND PITFALLS
rate can be submitted in a culture tube for fungal, bac-
terial, or mycobacterial testing. Direct histochemical
staining of the slide for fungi and mycobacterial bacilli The cytologic morphology of a granuloma in a well-
using conventional silver and acid-fast stains is the made smear is so characteristic that other entities rarely

Ch003-F06731.indd 76 10/26/2006 10:22:51 AM


CHAPTER 3 Lymph Nodes 77

enter into the diagnosis. Follicular dendritic cells or tial complication. Infectious mononucleosis is a self-
dendritic/lymphocytic aggregates could potentially be limited condition lasting from 3 to 4 weeks, and
mistaken for granulomas; however, the round to oval supportive medical treatment is the rule. Less than 1%
nuclear shape that is universal to these cells allows of affected patients develop serious complications from
their distinction from epithelioid histiocytes. A sup- the viral infection resulting in death.
purative bacterial lymphadenitis may mimic cat-scratch
disease because of the neutrophilic background, but
will lack the granulomas that exist in the latter. Rarely,
the spindle cell nature of epithelioid histiocytes is CYTOPATHOLOGIC FEATURES
exaggerated, and mimics a spindle cell proliferation.
However, not all granuloma clusters are affected, and
Aspiration smears typically contain many lymphocytes
one finds more typical granuloma morphology in other
in a dispersed pattern. Although a range of cells is
areas of the smear.
present, somewhat mimicking a reactive lymphoid
hyperplasia smear, some crucial differences exist. A
marked increase in the percentage of immunoblasts (a
consequence of the host response to EBV), centroblasts,
INFECTIOUS MONONUCLEOSIS plasmacytoid monocytes, and plasma cells is noted
along with a diminution or absence of follicular center
cell fragments. This abnormal proliferation of immu-
CLINICAL FEATURES noblasts results in a skewed elevated percentage of large
lymphocytes that can be misinterpreted easily for a
large cell lymphoma (Fig. 3-8). Immunoblasts display
Infectious mononucleosis is a disease caused by the enlarged nuclei, finely granular chromatin, one to three
Epstein-Barr virus (EBV). It is spread through person- nucleoli (that are indistinct in Romanowsky-stained
to-person contact, and occurs most commonly in ado- smears), and a moderate amount of basophilic cyto-
lescents and young adults. The estimated incidence is plasm (Fig. 3-9). EBV also induces infected B cells to
50 per 100,000 individuals in the United States, equally transform into plasmacytoid forms with eccentrically
affecting males and females. The classic clinical triad placed nuclei, basophilic cytoplasm, and a minimal
includes fever, pharyngitis, and peripheral lymphade- perinuclear clear zone.
nopathy that is always bilateral in the cervical chain.
Axillary and inguinal adenopathy may also occur.
Occasionally, unusual features will exist that mislead
the physician, resulting in delayed diagnosis. Lymph ANCILLARY STUDIES
nodes are often tender upon palpation, and movable.
Splenomegaly is common, and splenic rupture a poten-
The primary role of FNA is to suggest, confirm, or
exclude infectious mononucleosis in a patient who has
either not undergone serologic testing or has negative
serologic results. Laboratory findings include periph-
INFECTIOUS MONONUCLEOSIS – DISEASE FACT SHEET eral blood atypical lymphocytosis, and a positive mono-
spot (heterophile) test. IgM heterophile antibodies are
Definition
៉ A lymphadenopathy due to Epstein-Barr viral infection

Incidence INFECTIOUS MONONUCLEOSIS – PATHOLOGIC FEATURES


៉ About 50 per 100,000 in the US
Cytopathologic Findings
Gender and Age Distribution ៉ A range of lymphocytes but with a noticeable increase in the
៉ Most frequent in adolescents and young adults; rare in middle- percentage of immunoblasts, plasmacytoid lymphocytes/
aged or elderly monocytes, and plasma cells
៉ Few dendritic/lymphocytic aggregates and follicular center cell
Clinical Features fragments
៉ Pharyngitis, fever, splenomegaly
៉ Local or generalized lymphadenopathy Ancillary Studies
៉ Peripheral blood lymphocytosis ៉ Flow cytometry is polyclonal with a reversed T4/T8 ratio
៉ Positive heterophile antibodies ៉ Serologic testing for heterophile antibodies

Prognosis and Treatment Differential Diagnosis and Pitfalls


៉ Self-limited; death in <1% of cases owing to complications of ៉ Reactive lymphoid hyperplasia
EBV infection ៉ Large cell non-Hodgkin lymphoma
៉ Treatment is supportive in most instances ៉ Hodgkin lymphoma

Ch003-F06731.indd 77 10/26/2006 10:22:53 AM


78 FINE NEEDLE ASPIRATION CYTOLOGY

population, and a reversed CD4 : CD8 ratio with an


increase in cytotoxic CD8 suppressor T cells.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

Distinguishing infectious mononucleosis from reactive


lymphoid hyperplasia requires recognition of the
increased number of immunoblasts and plasmacytoid
monocytes coupled with the clinical picture. Unlike
reactive lymphoid hyperplasia, tingible-body macro-
phages and follicular center cell fragments are only a
minor component of the infectious mononucleosis
smear, if present at all. However, very early in the
course of infectious mononucleosis, lymphadenopathy
may demonstrate only a florid reactive lymphoid hyper-
plasia type of smear, without the large population of
FIGURE 3-8 immunoblasts and centroblasts, making infectious
Infectious mononucleosis. A nearly monotonous population of immuno- mononucleosis indistinguishable from an early phase
blasts mimicking a large cell lymphoma is present. These cell nuclei are reactive lymph node. Other causes of nodal immuno-
enlarged with slight irregularity of their nuclear outline. Nucleoli are
not particularly obvious with the Romanowsky stain. Note the two much
blast proliferation such as anticonvulsant-induced
smaller plasmacytoid lymphocytes in the center of the field and one at lymphadenopathy, herpes simplex lymphadenitis, and
9 o’clock with focal perinuclear clearing and eccentric nuclear position- drug hypersensitivity are cytologically identical to
ing. Romanowsky stain, high power. infectious mononucleosis. Clinical and serologic data
combined with the cytopathology are therefore neces-
sary to establish a specific diagnosis.
Infectious mononucleosis may be confused for large
cell NHL cytologically and clinically, since a small
number of lymphoma patients may present with infec-
tious mononucleosis-like clinical symptoms. Smears of
large cell lymphoma exhibit greater cell monotony, do
not show the range of lymphocyte types seen in infec-
tious mononucleosis, and uncommonly possess the
prominent plasmacytoid cell population. With FCM,
large B-cell lymphoma will demonstrate monoclonality.
Since a small number of proliferating immunoblasts
in infectious mononucleosis may exhibit binucleation,
Hodgkin lymphoma may enter into the differential diag-
nosis. True Reed-Sternberg cells have pale cytoplasm
rather than the basophilic cytoplasm of B immunoblasts,
and their enlarged nucleoli are much larger than those
seen in the immunoblasts of infectious mononucleosis.

FIGURE 3-9
Infectious mononucleosis. A mixture of immunoblasts, plasmacytoid
HODGKIN LYMPHOMA
lymphocytes, and small lymphocytes is seen. There is marked basophilia
of the immunoblast cytoplasmic periphery. The intermediate-sized
plasmacytoid lymphocytes display small zones of perinuclear clearing.
Romanowsky stain, high power. CLINICAL FEATURES

Hodgkin lymphoma (HL) is considered a B-cell lym-


the principal diagnostic serologic test for infectious phoma defined by its unique pathologic features. The
mononucleosis. These are present in almost 70% of incidence is 2–4 per 100,000 in the United States, with
patients after the first week of symptom onset. Almost a broad age range, peaking in the second to fourth
20% of patients, however, have a negative heterophile decades. A smaller spike occurs in the elderly, but this
test; this percentage is even higher in children less may be artificial. HL comprises up to 35% of all lym-
than 5 years of age. FCM is the principal ancillary phomas in Caucasians, but only 5–10% of all lympho-
study that should be performed from the FNA-captured mas that develop in Orientals. HL is slightly more
cells. This will demonstrate a polyclonal lymphocyte common (1.5 : 1) in males. Clinically, patients present

Ch003-F06731.indd 78 10/26/2006 10:22:54 AM


CHAPTER 3 Lymph Nodes 79

HODGKIN LYMPHOMA – DISEASE FACT SHEET HODGKIN LYMPHOMA – PATHOLOGIC FEATURES

Definition Cytopathologic Findings


៉ A lymphoma composed of cytologically abnormal cells termed ៉ Spectrum of lymphocytes with classic mirror-image Reed-
Reed-Sternberg (R-S) cells and Hodgkin cells residing in a Sternberg (R-S) cells or mononuclear variants
polymorphous mixture of lymphocytes and other inflammatory ៉ R-S cell: enlarged (≥20 μm), binucleated or mononucleated,
cells enlarged nucleus, polylobated nucleus, enlarged misshapen
nucleolus, meager to moderate amount of pale cytoplasm
Incidence ៉ Acute infl ammation and necrosis infrequent
៉ A wide variation globally ៉ Small granulomas possible
៉ 2–4 per 100,000 in the US and western Europe; uncommon in
Asian populations Ancillary Studies
៉ Classical Hodgkin lymphoma: R-S cells positive for CD15, CD30,
Gender and Age Distribution and fascin; negative for CD20
៉ Peak age is 15–35 years with a smaller peak in the 60s; rare ៉ Nodular lymphocyte-predominant Hodgkin lymphoma: R-S cells
before 10 years in the US positive for CD20, CD45, and EMA; negative for CD15 and CD30
៉ Slightly more common in males
Differential Diagnosis and Pitfalls
Clinical Features ៉ Reactive lymphoid hyperplasia
៉ Peripheral lymphadenopathy (cervical nodes most common) with ៉ Large cell non-Hodgkin lymphoma
single or multiple matted nodes above or below the diaphragm ៉ Infectious mononucleosis
and mediastinum; extranodal involvement rare
៉ Node(s) often firm to palpation
៉ Symptoms: fever, night sweats, weight loss, pruritus

Prognosis and Treatment


៉ Curable in >90% with stage I–II disease
៉ Multiagent chemotherapy, radiotherapy, or both
single macronuclei and macronucleoli. Nucleoli are
often comparable to the diameter of red cells in the
same field, and are often misshapen rather than smooth
and round (Fig. 3-11). Various combinations of nuclear
number and appearance can be seen (Fig. 3-12). The
classic R-S cell is binucleated with the same nuclear and
with painless enlargement of cervical or mediastinal cytoplasmic characteristics as the mononuclear form
lymph nodes. A single node or group of nodes may be (Fig. 3-13). In some cases, only bare R-S nuclei are seen,
encountered. About 10% also have constitutional making the diagnosis much more challenging. These
symptoms of fever, weight loss, pruritis, and drenching abnormal cells are randomly scattered in a hetero-
night sweats. No clinical laboratory test is specific for geneous background of lymphocytes. Plasma cells and
HL. Prognosis is related to clinical stage; overall 5-year eosinophils may be increased in these smears, and
survival is >95% for stage I and II disease with chemo- necrosis is also possible. Small, poorly formed clusters
therapy or radiotherapy regimens. of epithelioid histiocytes typical of granulomas are very
infrequent. A very small number of HL cases are asso-
ciated with neutrophilic inflammation. These examples
of so-called suppurative HL contain R-S cells admixed
CYTOPATHOLOGIC FEATURES with acute inflammatory cells.

Aspirates may be hyper- or hypocellular depending on


the degree of sclerosis in the affected node. Smears ANCILLARY STUDIES
mimic those of reactive lymphoid hyperplasia at low-
power examination with a range of various types of
lymphocytes present. Key to this diagnosis is the ability FCM has no role to play in the diagnosis of HL itself,
to identify Reed-Sternberg (R-S) cells. The classic binu- but has if one is also strongly considering large cell
cleated R-S cell is, in most instances, not easily found. lymphoma in the differential diagnosis. One should
Rather, one must be able to appreciate the presence of attempt to collect cells for a paraffin-embedded cell
enlarged mononuclear R-S variants in a heterogeneous block or cytospin preparation from aspirated cells, so
lymphocytic milieu that simulates reactive hyperplasia immunostaining is possible. Classical HL, which com-
(Fig. 3-10). These mononuclear so-called Hodgkin cells prises 90% of HL cases, has a completely different
(typically readily identifiable at medium power) are phenotype than nodular lymphocyte-predominant HL
markedly enlarged (20–30 μm) with rounded smooth (Table 3-1). Without such immunophenotyping, the
or lobated nuclear contours and a moderate amount of ability to separate HL into its two major subtypes using
pale, i.e. non-basophilic, cytoplasm. Hodgkin cells have cytology alone is compromised.

Ch003-F06731.indd 79 10/26/2006 10:22:55 AM


80 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 3-10
Hodgkin lymphoma, mononuclear Reed-Sternberg (R-S) cells. A, Two mononuclear R-S variants (center) are nearly hidden in this polymorphous lym-
phocytic focus. At this power, their recognition depends primarily on their size in relation to the lymphocytes surrounding them. Nuclear streaking
is most likely secondary to the sclerosis that was present in this case of classical Hodgkin lymphoma, nodular sclerosis type. Romanowsky stain,
medium power. B, This image is almost identical to (A), except for the stain. Note how much more obvious the macronucleoli appear, even though
the cells are partially obscured by the smeared nuclear chromatin of small lymphocytes. Papanicolaou stain, medium power.

TABLE 3-1
Immunophenotyping of Classical and Nodular
Lymphocyte-Predominant Hodgkin Lymphoma

R-S cells Classical HL NLP HL

CD45 Negative Positive


CD15 Positive/negative Negative
CD30 Positive (may be weak) Negative/positive
EMA Negative Positive/negative
Fascin Positive Negative
CD20 Negative/positive Positive

R-S, Reed-Sternberg; HL, Hodgkin lymphoma; NLP, nodular


lymphocyte-predominant; EMA, epithelial membrane
antigen.

FIGURE 3-11
Hodgkin lymphoma, mononuclear Reed-Sternberg (R-S) cell. A mononu-
clear R-S cell is just to the right of center. The macronucleus is very
faintly lobated, and contains an enlarged rounded nucleolus. Note the principal sources of error. Since most examples of HL
pale cytoplasm that lacks vacuoles. Small lymphocytes predominate in
this field, but an eosinophil lies directly to the left of the R-S cell, and
belong to the nodular sclerosis subtype, FNA of scle-
a vacuolated histiocyte is at the lower left. Romanowsky stain, high rotic nodes may lead to an absence or insufficient
power. number of R-S cells. A way to avoid this is to aspirate
not just the firm large node, but also a smaller node in
the same field. The latter often will have much less
sclerosis, and therefore enough R-S cells are extracted
DIFFERENTIAL DIAGNOSIS AND PITFALLS for identification.
Some cases of HL will have R-S cells in clusters rather
than as isolated randomly dispersed structures, thus
The differential diagnosis of HL includes large cell lym- invoking a possible diagnosis of large cell NHL. Large
phoma, reactive lymphoid hyperplasia, and infectious cell lymphoma aspirates do not exhibit the reactive lym-
mononucleosis. Failure to capture R-S cells in the aspi- phoid hyperplasia-type of accompanying cell types seen
rate, and secondarily the inability to identify them in HL. Rather, large cells most often populate smears,
in a polymorphous lymphocyte background are the sometimes with a minor population of small lympho-

Ch003-F06731.indd 80 10/26/2006 10:22:55 AM


CHAPTER 3 Lymph Nodes 81

A B

FIGURE 3-12
Hodgkin lymphoma, Reed-Sternberg (R-S) variants. A, This enlarged cell with voluminous pale chromatin contains a dual set of mirror-image macro-
nuclei and macronucleoli. Romanowsky stain, high power. B, Enlarged reddish nucleoli in this trinucleated R-S variant display an asymmetric rect-
angular shape, unlike the rounded edges in the prior image. Note the marked size difference between this R-S cell and the background lymphocytes.
Papanicolaou stain, high power.

immunoblasts, dendritic cells, plasmablasts, malignant


melanocytes, and, less often, the cells of a sarcoma or
large cell carcinoma (Fig. 3-14).

NON-HODGKIN LYMPHOMA,
SMALL CELL TYPES

CLINICAL FEATURES

The major small cell types of NHL include follicular


lymphoma, small lymphocytic lymphoma, mantle cell
lymphoma, and marginal zone lymphoma (Table 3-2).
Nearly all types appear in middle-age or older individu-
als, and are rare in children. Gender preference is
FIGURE 3-13 dependent on NHL subtype. Men are affected more
Hodgkin lymphoma, classic Reed-Sternberg (R-S) cell. Of the three R-S often (5 : 1) with mantle cell lymphoma, almost equal
cells in this field, only the one on the far right shows the classic ‘owl’s incidence occurs with follicular lymphoma and small
eye’ binucleation with round macronucleoli. The one in the middle is a
typical mononuclear so-called ‘Hodgkin cell’, and the cell at the far left
lymphocytic lymphoma, and a slight female predomi-
is multilobated with macronucleoli that are out of the focal plane. nance exists with marginal zone lymphoma. With the
Neutrophils are common in this field. Papanicolaou stain, high power. exception of mantle cell lymphoma, these lymphomas
are biologically indolent, but persistent. About 50,000
newly diagnosed patients (includes all types of NHL)
cytes. The malignant cells in large cell NHL also do not in all age groups present yearly in the United States
typically display the marked macronucleoli typical of (16 per 100,000). Of these pathologic subtypes, follicu-
R-S cells. FCM can be used to demonstrate light chain lar lymphoma is commonest (20–25% of cases), with
restriction in large B-cell NHL or an aberrant T-cell mantle cell lymphoma (6%), small lymphocytic lym-
phenotype in T-cell NHL. phoma (6%), and marginal zone lymphoma (7%) occur-
A binucleated cell(s) with mirror-image nucleo- and ring less often. Most patients present with asymptomatic
nucleolomegaly is not diagnostic of an R-S cell. This cell peripheral lymphadenopathy. Localized (stage I) disease
must exist in the proper milieu of a polymorphous lym- is uncommon for this group, with the exception of
phocyte background to be considered an R-S cell. Binu- marginal zone lymphoma, which is most commonly
cleated cells that can be confused as R-S cells include an extranodal disease (lymphoma of mucosa-associated

Ch003-F06731.indd 81 10/26/2006 10:22:56 AM


82 FINE NEEDLE ASPIRATION CYTOLOGY

A B

C D

FIGURE 3-14
Reed-Sternberg (R-S) cell look-alikes. A, Plasma cell myeloma. A large binucleated cell with nuclear sameness and equally sized nucleoli is sur-
rounded by a single cell pattern of smaller cells. These smaller forms are the key to the diagnosis. They are relatively monotonous (unlike the milieu
of Hodgkin lymphoma) with cytologic features of plasma cells. Papanicolaou stain, high power. B, Non-small cell carcinoma, metastatic. A binucle-
ated cell with nucleo- and nucleolomegaly has the typical morphologic features of an R-S cell. Yet, the surrounding cells are not lymphocytes, but
large epithelial cells in a single cell pattern. Papanicolaou stain, high power. C, Malignant melanoma, metastatic. An imitation R-S cell exists in
the setting of monotonous cells with plasmacytoid features typical of melanoma. Note the enlarged and misshapen nucleoli in the R-S look-alike.
Papanicolaou stain, high power. D, Small lymphocytic lymphoma. A binucleated immunoblast in the center of this image is surrounded by monoto-
nous rather than polymorphic lymphocytes. Note the absence of marked nucleolomegaly in this immunoblast. Romanowsky stain, high power.

NON-HODGKIN LYMPHOMA, SMALL CELL TYPE – Clinical Features


DISEASE FACT SHEET ៉ Localized or diffuse lymphadenopathy
៉ Peripheral blood and/or bone marrow involvement possible
Definition ៉ Extranodal presentation (parotid, periorbital, thyroid) typically
៉ A lymphoma composed of relatively monotonous ‘small MALT lymphoma
lymphocytes’; vast majority are B-cell ៉ Node(s) rubbery or firm

Incidence Prognosis and Treatment


៉ 16 per 100,000 in the US and Australia; much less in Asia, Africa, ៉ Aggressive to indolent, depending on subtype and International
and South America Prognostic Index
៉ Follicular lymphoma, marginal zone lymphoma, small lymphocytic ៉ Multiagent chemotherapy
lymphoma, and mantle cell lymphoma types are the most
common

Gender and Age Distribution


៉ Median age is 50–70 years
៉ Slight male predominance overall, but varies with subtype

Ch003-F06731.indd 82 10/26/2006 10:22:57 AM


CHAPTER 3 Lymph Nodes 83

NON-HODGKIN LYMPHOMA, SMALL CELL TYPE –


TABLE 3-2
PATHOLOGIC FEATURES
WHO Classification of Lymphoid Tissue,
B-Cell Neoplasms* Cytopathologic Findings
៉ Relatively monotonous small lymphocyte population lacking
• Precursor B-cell lymphoblastic leukemia/lymphoma cellular heterogeneity
• Mature B-cell neoplasms ៉ Evenly dispersed nucleoplasm with small or indistinct nucleoli,
B-cell CLL/small lymphocytic lymphoma smooth or irregular nuclear contour, sparse cytoplasm in small
B-cell prolymphocytic leukemia lymphocytes
Lymphoplasmacytic lymphoma ៉ Minor population of slightly larger ‘transformed’ lymphocytes
Mantle cell lymphoma depending on subtype
Follicular lymphoma ៉ Dendritic/lymphocytic aggregates most common in follicular
Marginal zone B-cell lymphoma of mucosa-associated lymphoma; uncommon in other subtypes
lymphoid tissue (MALT) type ៉ Tingible-body macrophages are uncommon
Nodal marginal zone lymphoma with or without
monocytoid B-cells Ancillary Studies
Splenic marginal zone B-cell lymphoma ៉ Light chain restriction; surface Ig+
Hairy cell leukemia ៉ CD5+ = small lymphocytic lymphoma (CD23+) or mantle cell
Diffuse large B-cell lymphoma lymphoma (CD23−)
Subtypes: mediastinal (thymic), intravascular, primary ៉ CD5− = follicular lymphoma (CD10 ±, bcl-6+) or marginal zone
effusion lymphoma (CD10−)
Burkitt lymphoma
Plasmacytoma Differential Diagnosis and Pitfalls
Plasma cell myeloma
៉ Distinction among various subtypes of small cell non-Hodgkin
lymphoma requires ancillary studies
*More common entities are in bold font. ៉ Reactive lymphoid hyperplasia.
៉ Small cell neuroendocrine carcinoma
៉ Myeloid (granulocytic) sarcoma

lymphoid tissue, i.e. MALT lymphoma). The Inter-


national Prognostic Index, which factors in patient age,
performance status, serum LDH level, number of extra- typically lacking tingible-body macrophages. Dendritic
nodal sites, and clinical stage, is commonly used to cells can be particularly prominent. Some examples of
predict survival. follicular lymphoma show pronounced plasmacytoid
differentiation. Attempts to grade follicular lymphoma
by counting the percentage of large centrocytic/centro-
blastic cells have been made.
CYTOPATHOLOGIC FEATURES
M ANTLE CELL LYMPHOMA

Except for marginal zone lymphoma, a morphologic These are highly cellular smears with almost a pure
feature common to this group is highly cellular smears population of small to intermediate-sized rounded lym-
with an almost monotonous population of small lym- phocytes. Nuclear notches and grooves are uncommon,
phocytes, that is, cells with small rounded or irregular or at least much less exaggerated than seen in follicular
nuclei, no visible nucleoli, fine or coarse nuclear chro- lymphoma, but some cases have nuclear contour irregu-
matin, and meager, attenuated cytoplasm. A dispersed larity (Fig. 3-16). Plasmacytoid monocytes, tingible-
single cell pattern is the rule (with the exception of body macrophages, dendritic/lymphocytic aggregates,
dendritic/lymphocytic aggregates), and lymphoglandu- follicular center cell fragments, and larger lymphocytes
lar bodies are numerous. Differences are listed for each such as centroblasts are often not seen. A blastoid
subtype. variant mimics the cytologic morphology of lympho-
blastic lymphoma (see below).
FOLLICULAR LYMPHOMA
SMALL LYMPHOCYTIC LYMPHOMA
Follicular lymphoma smears are composed of monot-
onous small lymphocytes, or a mixture of small lympho- Small lymphocytic lymphoma is the nodal counter-
cytes and larger transformed centrocytes and centroblasts. part of chronic lymphocytic leukemia (CLL). The cyto-
Indentations, grooves, and small projections are markers pathology overlaps with follicular lymphoma and mantle
of nuclear irregularity which is common to follicular cell lymphoma, due to a predominance of small lympho-
lymphoma; some grooves/clefts appear to bisect the cytes and the near exclusion of other types of lympho-
nucleus (Fig. 3-15). Dendritic/lymphocytic aggregates cytes. The nuclei of small lymphocytic lymphoma have
are common also in follicular lymphoma smears, as a clumped chromatin pattern, and are smooth or mini-
are follicular center cell fragments, but the latter are mally irregular (Fig. 3-17). A small population of large

Ch003-F06731.indd 83 10/26/2006 10:22:59 AM


84 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 3-15
Follicular lymphoma. A, The focus of these matching small lymphocytes is the irregular shape of their nuclear membrane. In some there is only a
minimal indentation (almost reniform), while others show deep clefts in the nucleus. Nucleoplasm is evenly dispersed without visible nucleoli, and
only a meager amount of cytoplasm is seen. Romanowsky stain, high power. B, A two-cell population of very darkly stained small lymphocytes, and
larger lymphocytes with small to intermediate amounts of cytoplasm is present. However, a true range of lymphocytes is absent. This dual popula-
tion of small–intermediate-size lymphocytes is common in grades I and II follicular lymphoma. A dendritic/lymphocytic aggregate is noticeable at
the lower right corner. Romanowsky stain, high power.

FIGURE 3-16 FIGURE 3-17


Mantle cell lymphoma. These small, almost identical lymphocytes have Small lymphocytic lymphoma. Small morphologically equivalent lympho-
such a high N/C ratio that they appear to be bare nuclei, but an cytes show some nuclear molding, and focal perinuclear clearing sug-
extremely attenuated rim of cytoplasm is present. Lymphoglandular gesting plasmacytoid differentiation. Note the abundant lymphoglandular
bodies are not well seen with this stain. Note the tingible-body bodies. Romanowsky stain, high power.
macrophage in the upper left corner. Papanicolaou stain, high power.

M ARGINAL ZONE LYMPHOMA

transformed cells corresponding to pseudofollicular Aspirates, particularly those of extranodal tissue,


growth centers is present. These large lymphocytes show variable cytomorphology. In some cases, a more
represent prolymphocytes and paraimmunoblasts. In or less monomorphous lymphocyte population is seen,
smears, it is difficult to distinguish them from each while in others the composition is one of lymphocytic
other; however, it is not necessary to do so. Both are heterogeneity that imitates a reactive condition. Mixed
two to three times the diameter of small lymphocytes, small and large lymphocytes having round, smooth or
and have a visible nucleolus and a moderate amount of irregular nuclei can be seen. Follicular dendritic cells,
pale or basophilic cytoplasm. plasmacytoid monocytes, follicular center cell fragments,

Ch003-F06731.indd 84 10/26/2006 10:22:59 AM


CHAPTER 3 Lymph Nodes 85

Unfortunately, lymphomas do not exhibit strict immu-


nologic fidelity, nor are lymphocyte markers present
and absent in 100% of cases. Molecular methods have
been introduced recently to overcome this handicap.
Mantle cell lymphoma has a t(11;14)(q13;q32) trans-
location in almost 75% of patients that produces an
upregulation of the bcl-1(PRAD1) gene, which in turn
leads to overexpression of a protein termed cyclin D1.
Follicular lymphoma is characterized by a t(14;18)
translocation with rearrangement of the bcl-2 gene in
over 75% of cases. These cytogenetic abnormalities are
currently being exploited by interphase FISH using
commercially available probes to detect abnormal
nuclear fusion signals from FNA specimens, thus
improving diagnostic specificity.

FIGURE 3-18 DIFFERENTIAL DIAGNOSIS AND PITFALLS


Marginal zone lymphoma, nodal. These monocytoid lymphocytes contain
slightly more visible cytoplasm, but their cellular sameness mimics that
of other small cell forms of lymphoma. The extranodal form of marginal Without doubt, the most important differential deci-
zone lymphoma is typically more heterogeneous. Romanowsky stain, sion is to accurately separate any small cell lymphoma
high power. from reactive lymphoid hyperplasia. In most cases, this
is readily accomplished by light microscopy alone, due
to the lymphocyte monotony of the former and hetero-
geneity of the latter. However, since small cell lym-
TABLE 3-3 phoma populations are not pure, those having any
Immunophenotyping of Small Cell B-Cell Lymphomas variation in cell size and type can cause confusion with
reactive lymphoid hyperplasia. This problem is most
Marker SLL MtCL FL MZL acute with follicular lymphoma, where a mixture of
small and large lymphocytes in nearly equal numbers
CD5 Pos Pos Neg Neg is possible, and not uncommon. If one looks carefully
CD10 Neg Neg/pos Pos/neg Neg at each of these forms of small cell lymphoma (except
CD20 Pos/neg Pos Pos Pos for marginal zone lymphoma), it is rare indeed for them
CD23 Pos Neg Neg/pos Neg to show a true range of lymphocyte forms (immuno-
FMC-7 Neg Pos Pos Pos blasts, plasmacytoid monocytes, and tingible-body mac-
CD43 Pos Pos Neg Neg/pos rophages) as occurs in a reactive lymphoid hyperplasia
Tdt Neg Neg Neg Neg smear. That said, these different cell types can be
present in small cell lymphoma smears, but only as a
SLL, small lymphocytic lymphoma; MtCL, mantle cell minor population, and even more importantly it is rare
lymphoma; FL, follicular lymphoma; MZL, marginal zone for all the various cell elements to exist in similar per-
lymphoma; pos/neg, majority of cases are positive, small centages as they do in reactive lymphoid hyperplasia.
percentage negative; neg/pos, majority of cases are
negative, small percentage positive.
Thus, immunophenotyping is currently the best pro-
tection the cytopathologist has in not confusing a small
cell type of NHL for reactive lymphoid hyperplasia.
Small cell neuroendocrine carcinoma consists of cells
that are about three times the size of small lymphocytes,
immunoblasts, tingible-body macrophages, and plasma
and show no range in cell types. Instead, because of
cells are all possible. Monocytoid lymphocytes, those
their high cell turnover, a two-cell population of viable
having smooth rounded nuclei and a moderate amount
intact cells and apoptotic cells characterized by pyk-
of pale cytoplasm, are variable in number (Fig. 3-18).
notic, karyorrhectic nuclei exists (Fig. 3-19). This
invariably creates smears having abundant background
necrosis. Necrosis can be seen in small cell lymphomas,
ANCILLARY STUDIES but is distinctly unusual. Smearing or streaking of nuclei
is non-specific, since it is present in both lymphoma and
carcinoma. Although small cell neuroendocrine carci-
Because a large degree of morphologic overlap exists noma cells have a high nuclear to cytoplasmic (N/C)
among this group of NHL, it is imperative to phenotype ratio and may superficially resemble small lymphocytes,
these aspirates for specific subclassification. This is they display clustering, crowding, and ‘molding’ of their
best accomplished using FCM whereby the application nuclei into one another. This is not to say that
of various surface markers is employed (Table 3-3). nuclear molding is completely absent in all small cell

Ch003-F06731.indd 85 10/26/2006 10:23:01 AM


86 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 3-19
Small cell neuroendocrine carcinoma, metastatic. A, Although a few dissociated cells are found, most are arranged in these small compressed
aggregates leading to angulated distortion of nuclear shapes. Romanowsky stain, high power. B, Markedly hyperchromatic nuclei cling together
loosely. Note that some of these have an angulated or spindle shape. The abundant necrosis creates numerous lymphoglandular body look-alikes,
particularly with this stain. Papanicolaou stain, high power.

lymphomas, but when present is less florid than that


seen in carcinoma. A search for lymphoglandular bodies NON-HODGKIN LYMPHOMA, LARGE CELL TYPES –
in small cell neuroendocrine carcinoma will uncover DISEASE FACT SHEET
cell debris fragments, but not the globular, vacuolated
structures that are common to lymphoma. Definition
Myeloid (granulocytic) sarcoma is a rare tumor of ៉ A lymphoma composed of relatively monotonous ‘large
myeloid precursors that can arise in extramedullary lymphocytes’; majority are B-cell, 10–12% are T-cell
sites prior to (rare) or concurrent with acute myeloid
leukemia. Skin and soft tissue are preferred sites. The Incidence
blastic immature types have features of small or ៉ 30–40% of all adult lymphomas
៉ T-cell lymphomas are more common in Asians
intermediate-size lymphoma with a single cell pattern,
high N/C ratio, and minimal cytoplasm. Lymphoglandu-
Gender and Age Distribution
lar bodies can be present. The more differentiated forms
of myeloid sarcoma show a spectrum of myeloid differ- ៉ Broad age range, from children to elderly; peak age in 7th decade
in adults
entiation with granulated promyelocytes, metamyelo-
៉ Slightly more common in men
cytes, or band forms seen. Immunophenotyping shows
expression of myeloid markers CD13, CD33, CD43, and Clinical Features
myeloperoxidase.
៉ Localized or diffuse adenopathy, extranodal disease
៉ Symptoms: fever, night sweats, weight loss, pruritus
៉ Node(s) often firm to palpation

NON-HODGKIN LYMPHOMA, Prognosis and Treatment


LARGE CELL TYPES ៉ Aggressive, potentially curable
៉ Multiagent chemotherapy

CLINICAL FEATURES

Large cell lymphomas encompass malignancies derived tries and in immunosuppressed patients. In the T-cell
from B, T, or NK cells. Of these, diffuse large B-cell group, anaplastic large cell lymphoma and peripheral
lymphoma is most common, constituting about 35– T-cell lymphoma are most common (Table 3-4). T-cell
40% of all adult lymphomas in Western Europe and lymphomas comprise about 10% of all NHL in North
North America (about 25,000 new cases annually) and America, and about twice that percentage in Asia. The
about one-third of pediatric lymphomas. A higher inci- median age in adults of large cell lymphomas is 70
dence of large cell lymphoma exists in developing coun- years, but there is a broad age range. Anaplastic large

Ch003-F06731.indd 86 10/26/2006 10:23:01 AM


CHAPTER 3 Lymph Nodes 87

CYTOPATHOLOGIC FEATURES
TABLE 3-4
WHO Classification of Lymphoid Tissue, Common to all forms of large cell lymphoma is an aspi-
T-Cell Neoplasms*
rate smear containing a predominance of large lympho-
cytes. These range from two to five times the diameter
• Precursor T-cell lymphoblastic leukemia/lymphoma of small mature lymphocytes. In some examples, large
• Mature T-cell and NK-cell neoplasms lymphocytes are the only cell type found, but in most
T-cell prolymphocytic leukemia
T-cell large granular lymphocytic leukemia
cases there is a minor population of small lymphocytes.
NK-cell leukemia In diffuse large B-cell lymphoma, centroblastic-type
Extranodal NK-/T-cell lymphoma, nasal type cells are most commonly seen, with a smaller number
Mycosis fungoides of cases having immunoblastic features. Nuclei vary
Sézary syndrome from smooth rounded structures to those with irregu-
Angioimmunoblastic T-cell lymphoma lar contours, finely dispersed or coarse chromatin, and
Peripheral T-cell lymphoma variable presence of nucleoli; single macronucleoli are
Adult T-cell leukemia/lymphoma (HTLV1) characteristic of the immunoblastic variant of diffuse
Systemic anaplastic large cell lymphoma (T- and large B-cell lymphoma (Fig. 3-20). Binucleated, multi-
null-cell types) nucleated, multilobated, and reniform nuclei can be
Primary cutaneous anaplastic large cell lymphoma
Subcutaneous panniculitis-like T-cell lymphoma
seen, particularly with anaplastic large cell lymphoma
Enteropathy-type intestinal T-cell lymphoma (Fig. 3-21). A greater amount of cytoplasm is present
Hepatosplenic γ/δ T-cell lymphoma when compared with the small cell lymphoma group.
It is often deeply basophilic in Romanowsky-stained
*More common entities are in bold font.
smears, and small cytoplasmic vacuoles are not uncom-
mon. Tingible-body macrophages and individual cell
necrosis are also more frequent in large cell lymphomas
than in the small cell category; there is a paucity of
follicular center cell fragments. As in all examples of
lymph node aspirates, nuclear streaking and smearing
are not uncommon; this is particularly evident in those
large cell lymphomas associated with sclerosis.
cell lymphoma and diffuse large B-cell lymphoma, for Although peripheral T-cell lymphoma aspirates are
example, are the two most common forms of childhood/ discussed in this section of large cell lymphoma, their
young adult large cell lymphoma. Men are affected
slightly more than women in diffuse large B-cell lym-
phoma, but there is a marked male predominance (6 : 1)
for anaplastic large cell lymphoma. A rapidly enlarging
mass, either nodal or extranodal, is the usual present-
ing complaint. Extranodal sites include the gastrointes- NON-HODGKIN LYMPHOMA, LARGE CELL TYPES –
tinal tract (most common), skin, mediastinum, soft PATHOLOGIC FEATURES
tissue, bone, central nervous system, and salivary
Cytopathologic Findings
gland. Disseminated disease at the time of diagnosis is
៉ Relatively monotonous large lymphocytes
common. The International Prognostic Index is predic-
៉ Nuclear size equal to or exceeds that of macrophage; 2–5 times
tive of survival. Only about 40% of patients with
larger than mature lymphocyte
diffuse large B-cell lymphoma are cured with polyche- ៉ Nucleoli common, but not universal
motherapy. Gene expression profi ling has recently been ៉ Nuclear pleomorphism with lobulated, indented nuclei and
introduced to develop molecularly distinct portraits of multiple nuclei common in anaplastic large cell lymphoma
diffuse large B-cell lymphoma (and, parenthetically, ៉ Cytoplasm moderate to abundant; can be vacuolated
follicular lymphoma also) to predict survival and thus ៉ Variable tingible-body macrophages and necrosis
target patients for various types of chemotherapy
regimens. Ancillary Studies
In anaplastic large cell lymphoma, a relatively homo- ៉ Immunophenotyping best accomplished using flow cytometry
geneous group defined by a specific cytogenetic abnor- ៉ B-cell: light chain restriction; positive for CD20 and CD19;
mality has emerged. These patients develop systemic variable CD10, surface Ig, aberrant CD43 expression
disease in the first three decades of life, and their lym- ៉ T-cell: aberrant phenotype with loss of CD7, CD5, or CD2;
phomas are associated with a specific translocation, positive for CD3, CD43, and CD30
៉ PCR: clonally rearranged T-cell receptor genes
t(2;5)(p23;q35), which fuses the ALK gene with the
NPM gene, thereby producing a protein (anaplastic
Differential Diagnosis and Pitfalls
large cell lymphoma kinase) to which antibodies have
៉ Infectious mononucleosis
been developed. These patients have a much improved
៉ Hodgkin lymphoma
prognosis (70% 5-year survival) than their anaplastic ៉ Non-lymphoid large cell malignancies
large cell lymphoma kinase-negative counterparts (30%
5-year survival).

Ch003-F06731.indd 87 10/26/2006 10:23:02 AM


88 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 3-20 FIGURE 3-22


Diffuse large B-cell lymphoma. Large lymphocytes contain one or more Peripheral T-cell lymphoma, unspecified. A mixture of lymphoid cells
nucleoli in hypochromatic vesicular nuclei. A second population of is present, including an immunoblast (right of center), several large
darkly stained small lymphocytes is not uncommon in large cell lym- lymphocytes harboring visible nucleoli, plasmacytoid lymphocytes, and
phoma. Papanicolaou stain, high power. even an eosinophil (far left). This cytomorphology is not specific, but
is typical of this lymphoma. Romanowsky stain, high power.

their absence could be a clue that one is not dealing with


a normal population of lymphocytes.

ANCILLARY STUDIES

Immunophenotyping using FCM is the most efficacious


method for determining light chain restriction, and
thus B-cell monoclonality. Nearly all are positive for
surface Ig and CD19, with variable expression of CD10,
CD22, CD23, and FMC-7.
For T-cell lymphomas, immunophenotyping will
show an absence of B-cell markers and a variable panel
of T-cell marker (CD1a, CD2, CD3, CD4, CD5, CD7,
CD8) positivity. Typically, a loss of one or more of these
markers, particularly CD7, or combined absence of
CD4/CD8 is seen. The term anaplastic large cell lym-
FIGURE 3-21
phoma is currently restricted to CD30-positive T- and
Anaplastic large cell lymphoma. All lymphocytes in this field are large,
with some more anaplastic than others. Several nuclei have reniform
null-cell lymphomas. CD30 is typically added to an FCM
configurations which are typical, but not pathognomonic, of this lym- panel if a large lymphocyte population is observed. PCR
phoma. Romanowsky stain, high power. analysis of T-cell receptor (TCR) beta gene rearrange-
ment is the most specific test to determine clonality of
a T-cell lymphoma. This can be accomplished using
PCR fragment size analysis of the FNA material, for
cytomorphology is much more heterogeneous than that
which instruments are commercially available.
of diffuse large B-cell lymphoma or anaplastic large cell
lymphoma. These smears may be composed of an over-
whelmingly monomorphous population of large cells
similar to any large cell NHL. However, many examples DIFFERENTIAL DIAGNOSIS AND PITFALLS
exhibit a mixture of small, intermediate-sized, and large
lymphocytes reminiscent of reactive lymphoid hyper-
plasia (Fig. 3-22). A variable number of plasma cells, The high percentage of immunoblasts makes infectious
eosinophils, and histiocytes can be present. Follicular mononucleosis a mimicker of large cell lymphoma.
center cell fragments and tingible-body macrophages are Close examination of the infectious mononucleosis
uncommon to rare in peripheral T-cell lymphoma, and smear will show a subpopulation of plasmacytoid

Ch003-F06731.indd 88 10/26/2006 10:23:02 AM


CHAPTER 3 Lymph Nodes 89

FIGURE 3-23 FIGURE 3-24


Nasopharyngeal carcinoma, metastatic. A discrete tight cluster of epi- Seminoma, metastatic. A mixture of large germ cells and smaller lym-
thelial cells to the right of center is surrounded and infiltrated by small phocytes are set in a distinctive background of reticulated cytoplasm
lymphocytes. Note that epithelial cell nuclei lack discrete nucleoli, but (so-called ‘tigroid background’). Some of the germ cells contain one
are about three times larger than the lymphocyte population. Papani- or more visible nucleoli and a moderate amount of cytoplasm. Bare
colaou stain, high power. germ cell nuclei are also present (lower right). Romanowsky stain, high
power.

lymphocytes, and some plasma cells. Immunopheno- young adult male. A second population of lymphocytes
typing will demonstrate the polyclonality typical of accompanies the large germ cells. Thus, smears will
infectious mononucleosis. HL has occasionally been contain lymphoglandular bodies. Germ cells having
mistaken as a large cell lymphoma also. Cells having large rounded nuclei with enlarged nucleoli are distrib-
the morphologic features of R-S cells have been seen in uted singly or in clusters, bare nuclei are common, and
all forms of large cell lymphoma. The key to avoiding cell cytoplasm is smeared in a reticular pattern, or as
this trap of mistaking such cells as true R-S cells lies short strips, giving the appellation ‘tigroid’ to this pattern
in examination of all components that exist in the (Fig. 3-24). It is important to remember that this pattern
smear. With HL, the smear background simulates that is not universally present, nor is it seen well in
of reactive lymphoid hyperplasia, whereas in large cell Papanicolaou- or hematoxylin and eosin (H&E)-stained
lymphomas it lacks this lymphocytic heterogeneity. smears.
The two principal non-small cell carcinomas meta- Malignant melanoma is probably the most frequent
static to lymph nodes, metastatic squamous cell carci- mimic of lymphoma since it commonly involves lymph
noma and adenocarcinoma, are rarely mistaken as nodes, displays a monomorphic population of cells, has
large cell lymphoma, since they typically lack lympho- a single cell pattern, is habitually amelanotic, and may
glandular bodies and nearly always display some cell present in a patient without a known primary malig-
aggregation. Metastatic nasopharyngeal carcinoma, nancy. Smears lack lymphoglandular bodies. Character-
undifferentiated type, however, can be misdiagnosed as istically, cells are often binucleated with mirror-image
lymphoma for several reasons. First, patients frequently nuclei, have intranuclear cytoplasmic inclusions, and
present with an enlarged cervical node like lymphoma, cell nuclei eccentrically displaced toward the cyto-
bypassing the usual clinical scenario where an individ- plasmic edge, giving them a plasma cell-like appearance
ual is known to have cancer and only then develops (Fig. 3-25).
lymphadenopathy. Also, lymphocytes are a normal
constituent of this tumor; therefore, lymphoglandular
bodies are present. A range of lymphocytes often com-
mingles with and can sometimes obscure the clusters of
malignant epithelial cells (which may be few), and epi- PEDIATRIC NON-HODGKIN LYMPHOMAS
thelial cells can be dispersed in loose aggregates. Epithe-
lial cells have large nuclei, may or may not have visible
nucleoli, and have a small amount of cytoplasm that CLINICAL FEATURES
lacks keratinization due to their undifferentiated state
(Fig. 3-23). Cytokeratin staining of the smear may be
necessary to fully characterize these cells. The principal lymphomas of childhood include precur-
Metastatic seminoma should also be considered if the sor T- and B-cell lymphoblastic, Burkitt, and large cell
clinical setting is suggestive, such as mediastinal/hilar lymphoma, comprising 30–40%, 40–50%, and 15–25%
adenopathy or retroperitoneal lymphadenopathy in a of cases, respectively. Burkitt lymphoma is rare in the

Ch003-F06731.indd 89 10/26/2006 10:23:04 AM


90 FINE NEEDLE ASPIRATION CYTOLOGY

United States, with only 100–150 new cases annually,


compared with in equatorial Africa, where 0.01% of
children develop it. Other lymphoma subtypes are
exceedingly rare in this age group. Large cell lympho-
mas can be B-cell, T-cell, or null-cell types; this group
is discussed in the section above.
In lymphoblastic lymphoma, the male to female
ratio is about 2 : 1, with the majority occurring in the
older child–adolescent years. Lymphoblastic lymphoma
almost always presents as supra-diaphragmatic lymph-
adenopathy. An anterior mediastinal mass is found in
up to 80% of patients. This mass may induce symptoms
of bronchial asthma, persistent cough, cyanosis, syncope,
or superior vena cava syndrome, and tracheal compres-
sion. Burkitt lymphoma is also a neoplasm of adoles-
cence and young adulthood, with a male predominance
(2–3 : 1). In developed countries, Burkitt lymphoma
presents primarily as an intra-abdominal mass. Some
FIGURE 3-25
individuals present with signs and symptoms of acute
appendicitis. In children, precursor T-cell lymphoblastic
Malignant melanoma, metastatic. A polymorphous population of epi-
thelioid cells is seen. Note that two of these cells have mirror-image lymphoma has a good prognosis, with 70–90% 5-year
nuclei, and three have distinct intranuclear cytoplasmic inclusions. disease-free survival. The prognosis is worse in adults,
Eccentric nuclear placement in some gives them a plasmacytoid look. with a 5-year disease-free survival of 45–55%. Burkitt
Compare the size of these to the small lymphocytes and neutrophils in lymphoma patients with localized disease or with com-
the background. Papanicolaou stain, high power.
pletely resected abdominal disease can have a long-term
survival rate of 90% or more.

PEDIATRIC NON-HODGKIN LYMPHOMAS – DISEASE FACT SHEET CYTOPATHOLOGIC FEATURES


Definition
៉ Lymphoblastic: lymphoma of precursor lymphocytes, 85–90% T-cell Smears of lymphoblastic lymphoma are hypercellular
៉ Burkitt: lymphoma of medium-size B cells with translocation with uniform lymphoid blasts about twice the size of
involving c-myc gene small lymphocytes, having round to convoluted nuclei,
finely granular so-called ‘dusty’ chromatin, inconspicu-
Incidence ous or small nucleoli, and extremely meager, rarely
៉ Lymphoblastic lymphoma and Burkitt lymphoma each account for vacuolated cytoplasm. Tingible-body macrophages may
30–40% of pediatric lymphomas be present (Fig. 3-26). Aspirates of Burkitt lymphoma
៉ Burkitt: endemic in equatorial Africa, sporadic in North America/
Europe

Gender and Age Distribution PEDIATRIC NON-HODGKIN LYMPHOMAS – PATHOLOGIC FEATURES


៉ Male to female ratio about 2 : 1
៉ Peak in 2nd and 3rd decades of life for both Cytopathologic Findings
៉ Lymphoblastic: monomorphic intermediate-size cells, round or
Clinical Features convoluted nuclei, finely granular chromatin, indistinct nucleoli,
៉ Lymphoblastic: cervical, supraclavicular, or axillary infrequently vacuolated sparse cytoplasm
lymphadenopathy, 50–80% with anterior mediastinal mass, pleural ៉ Burkitt: monomorphic intermediate-size cells, coarse chromatin,
effusion, dyspnea, wheezing 2–5 nucleoli, commonly vacuolated sparse cytoplasm
៉ Burkitt: abdominal visceral disease most common in North ៉ Numerous tingible-body macrophages
America/Europe, head and neck masses less common; associated
with EBV infection, AIDS Ancillary Studies
៉ Lymphoblastic: Tdt-positive in both B- and T-cell types; 85–90%
Prognosis and Treatment express T-cell antigens
៉ 90% survival for stage I and II disease, about 70–80% for stages ៉ Burkitt: B-cell, Tdt-negative, monoclonal light chain; positive
III and IV for surface Ig, CD10, CD19, and CD20; FISH: c-myc translocation
៉ Multiagent chemotherapy for both types
Differential Diagnosis and Pitfalls
៉ Blastoid variant of mantle cell lymphoma
៉ Myeloid (granulocytic) sarcoma
៉ Malignant small round cell tumors of childhood

Ch003-F06731.indd 90 10/26/2006 10:23:05 AM


CHAPTER 3 Lymph Nodes 91

mas may be either. Lymphoblastic lymphoma is defined


by the near universal presence of Tdt (terminal deoxy-
nucleotidyl transferase), which exists in both T- and
B-cell types, and can be analyzed using FCM. A con-
sistent non-random translocation, t(8;14), that involves
the c-myc gene occurs with Burkitt lymphoma.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

In pediatric patients, the principal diagnostic problem


for either lymphoblastic lymphoma or Burkitt lym-
phoma is to exclude other entities that exist in the
category of ‘malignant small round cell tumor’. These
include rhabdomyosarcoma, Ewing sarcoma, desmo-
plastic small round cell tumor, and, less often, neuro-
blastoma or Wilms tumor. This is often accomplished
FIGURE 3-26 merely by clinical and radiologic evaluation. The single
Precursor T-cell lymphoblastic lymphoma. An identical population of cell pattern combined with background lymphoglandu-
lymphocytic blasts display delicate, evenly dispersed chromatin without
discrete nucleoli, and minimal cytoplasm. Some nuclei show slight
lar bodies will usually suffice to allow categorization
unevenness. Smaller, more darkly stained nuclei represent apoptotic of the aspirate as a lymphoma since these two features
cells. Romanowsky stain, high power. are absent in the other lesions in this category. A poten-
tial pitfall exists if an anterior mediastinal mass is
aspirated. Since benign thymocytes are Tdt-positive,
one must ensure that cells have a blast morphology.
The blastoid variant of mantle cell lymphoma can
mimic lymphoblastic lymphoma. Clinically, these
patients are not in the same age group as those with
lymphoblastic lymphoma. The morphology, however,
can be very similar with intermediate to large cells;
immunotyping is required. All mantle cell lymphomas
are B-cell and lack Tdt.
Myeloid sarcoma may occur in children prior to a
leukemic phase. The undifferentiated forms can mimic
a blastic morphology. FCM shows lack of lymphocytic
markers and expression of myeloid markers, as dis-
cussed above.

SUGGESTED READINGS
Reactive Lymphoid Hyperplasia
FIGURE 3-27 Flanders E, Kornstein MJ, Wakely PE Jr, Kardos TF, Frable WJ. Lympho-
Burkitt lymphoma. The prominent feature in these blasts is the coarse glandular bodies in fine needle aspiration cytology. Am J Clin Pathol
chromatin with one to several small nucleoli. Cytoplasmic vacuoles are 1993;99:566–569.
barely visible in this image (center cell). Mitotic figures are an expected Geisinger KR, Stanley MW, Raab SS, Silverman JS, Abati A. Lymph nodes
finding in Burkitt lymphoma smears. Romanowsky stain, high power. and spleen. In: Modern Cytopathology. Philadelphia: Churchill-
Livingstone, 2004:643–687.
Glant MD. Cytopathology of lymph nodes in nonspecific reactive hyper-
plasia: prognostication and differential diagnosis. Am J Clin Pathol
show nuclei with coarse chromatin, small nucleoli, and 1997;108:S31–S55.
variable cytoplasmic vacuoles. Tingible-body macro- Wakely PE Jr. Aspiration and touch preparation of lymph nodes. In:
phages are more frequent in Burkitt lymphoma smears, Atkinson BF, ed. Atlas of Diagnostic Cytopathology, 2nd ed. Philadelphia:
Saunders, 2004:411–458.
but this too is variable (Fig. 3-27). Wakely PE Jr, Cibas ES. Lymph nodes. In: Cibas ES, Ducatman BS, eds.
Cytology: Diagnostic Principles and Clinical Correlates, 2nd ed. Edinburgh:
Elsevier, 2003:307–344.

ANCILLARY STUDIES
Granulomatous Lymphadenopathy
Das DK, Bhambhani S, Pant JN, et al. Superficial and deep-seated tuber-
Most lymphoblastic lymphomas are T-cell (85–90%), culous lesions: fine-needle aspiration cytology diagnosis of 574 cases.
all Burkitt lymphomas are B-cell, and large cell lympho- Diagn Cytopathol 1992;8:211–215.

Ch003-F06731.indd 91 10/26/2006 10:23:06 AM


92 FINE NEEDLE ASPIRATION CYTOLOGY

DeMay RM. Lymph node. In: The Art and Science of Cytopathology. Meda BA, Buss DH, Woodruff RD, et al. Diagnosis and subclassification of
Chicago: ASCP Press, 1996:779–846. primary and recurrent lymphomas. The usefulness and limitations of
Ellison E, Lapuerta P, Martin SE. Fine needle aspiration diagnosis of combined fine needle aspiration cytomorphology and flow cytometry.
mycobacterial lymphadenitis. Sensitivity and predictive value in the Am J Clin Pathol 2000;113:688–699.
United States. Acta Cytol 1999;43:153–157. Rassidakis GZ, Tani E, Svedmyr E, Porwit A, Skoog L. Diagnosis and sub-
Stanley MW, Horwitz CA, Burton LG, Weisser JA. Negative images of bacilli classification of follicle center and mantle cell lymphomas on fine-
and mycobacterial infection: a study of fine-needle aspiration smears needle aspirates. A cytology and immunocytochemical approach based
from lymph nodes in patients with AIDS. Diagn Cytopathol 1990;6: on the revised European-American lymphoma (REAL) classification.
118–121. Cancer (Cancer Cytopathol) 1999;87:216–223.
Stastny JF, Wakely PE Jr, Frable WJ. Cytologic features of necrotizing Suh YK, Shin HJ. Fine-needle aspiration biopsy of granulocytic sarcoma:
granulomatous inflammation consistent with cat scratch disease. Diagn a clinicopathologic study of 27 cases. Cancer (Cancer Cytopathol) 2000;
Cytopathol 1996;15:108–115. 90:364–372.
Young NA, Al-Saleem T. Diagnosis of lymphoma by fine-needle aspiration
cytology using the revised European-American classification of lym-
Infectious Mononucleosis phoid neoplasms. Cancer (Cancer Cytopathol) 1999;87:325–345.
Zeppa P, Marino G, Troncone G, et al. Fine-needle cytology and flow
Kardos TF, Kornstein MJ, Frable WJ. Cytology and immunocytology of cytometry immunophenotyping and subclassification of non-Hodgkin
infectious mononucleosis in fine needle aspirates of lymph nodes. Acta lymphoma: a critical review of 307 cases with technical suggestions.
Cytol 1988;32:722–726. Cancer (Cancer Cytopathol) 2004;102:55–65.
Stanley MW, Steeper TA, Horwitz CA, Burton LG, Strickler JG, Borken S.
Fine needle aspiration of lymph nodes in patients with acute infectious
mononucleosis. Diagn Cytopathol 1990;6:323–329. Non-Hodgkin Lymphoma, Large Cell Types
Wakely PE Jr. Aspiration and touch preparation of lymph nodes. In:
Atkinson BF, ed. Atlas of Diagnostic Cytopathology, 2nd ed. Philadel- Al Shanqeety O, Mourad WA. Diagnosis of peripheral T-cell lymphoma by
phia: Saunders, 2004:411–458. fine-needle aspiration biopsy: a cytomorphologic and immunopheno-
typic approach. Diagn Cytopathol 2000;23:375–379.
Dong HY, Harris NL, Preffer FI, Pitman MB. Fine-needle aspiration biopsy
Hodgkin Lymphoma in the diagnosis and classification of primary and recurrent lymphoma:
a retrospective analysis of the utility of cytomorphology and flow
Chhieng DC, Cangiarella JF, Symmans WF, Cohen J-M. Fine-needle aspira- cytometry. Mod Pathol 2001;14:472–481.
tion cytology of Hodgkin disease. A study of 89 cases with emphasis Ng W-K, Ip P, Choy C, Collins RJ. Cytologic and immunocytochemical
on the false-negative cases. Cancer (Cancer Cytopathol) 2001;93:52– findings of anaplastic large cell lymphoma. Analysis of ten fine-needle
59. aspiration specimens over a 9-year period. Cancer (Cancer Cytopathol)
Fulciniti F, Vetrani A, Zeppa P, et al. Hodgkin’s disease: diagnostic accu- 2003;99:33–43.
racy of fine needle aspiration; a report based on 62 consecutive cases. van Heerde P. Lymph nodes. In: Orell SR, Sterrett GF, Walters MN-I,
Cytopathology 1994;5:226–233. Whitaker D, eds. Manual and Atlas of Fine Needle Aspiration Biopsy.
Jaffe ES, Harris NL, Stein H, Vardiman JW, eds. World Health Organization London: Churchill-Livingstone, 1999:74–108.
Classification of Tumours. Pathology and Genetics of Tumours of Hae- Yao JL, Cangiarella JF, Cohen J-M, Chhieng DC. Fine-needle aspiration
matopoietic and Lymphoid Tissues. Lyon, France: IARC Press, 2001. biopsy of peripheral T-cell lymphomas. A cytologic and immunopheno-
Kardos TF, Vinson JH, Behm FG, Frable WJ, O’Dowd GJ. Hodgkin’s disease: typic study of 33 cases. Cancer (Cancer Cytopathol) 2001;93:151–
diagnosis by fine needle aspiration biopsy. Analysis of cytologic criteria 159.
from a selected series. Am J Clin Pathol 1986;86:286–291.
Tani E, Ersoz C, Svedmyr E, et al. Fine needle aspiration cytology and
immunocytochemistry of Hodgkin’s disease – suppurative type. Diagn Pediatric Non-Hodgkin Lymphomas
Cytopathol 1998;18:437–440.
Geisinger KR, Silverman JF, Wakely PE Jr. Pediatric Cytopathology. Chicago:
ASCP Press, 1994:201–228.
Non-Hodgkin Lymphoma, Small Cell Types Jacobs JC, Katz RL, Shabb N, et al. Fine needle aspiration of lymphoblastic
lymphoma. A multiparameter diagnostic approach. Acta Cytol 1992;36:
Caraway NP, Gu J, Lin P, et al. The utility of interphase fluorescence in 887–894.
situ hybridization for the detection of the translocation t(11;14)(q13;32) Stastny JF, Almeida MM, Wakely PE Jr, Kornstein MJ, Frable WJ. Fine
in the diagnosis of mantle cell lymphoma on fine-needle aspiration needle aspiration biopsy and imprint cytology of small non-cleaved cell
specimens. Cancer (Cancer Cytopathol) 2005;105:110–118. (Burkitt’s) lymphoma. Diagn Cytopathol 1995;12:201–207.
Chhieng DC, Cohen J-M, Cangiarella JF. Cytology and immunophenotyping Tani E, Maeda S, Frostad B, et al. Aspiration cytology with phenotyping
of low- and intermediate-grade B-cell non-Hodgkin’s lymphomas with and clinical presentation of childhood lymphomas. Diagn Oncol
a predominant small-cell component: a study of 56 cases. Diagn Cyto- 1993;3:294–301.
pathol 2001;24:90–97. Wakely PE Jr, Kornstein MJ. Aspiration cytopathology of lymphoblastic
Crapanzano JP, Lin O. Cytologic findings of marginal zone lymphoma. A lymphoma and leukemia: the MCV experience. Pediatr Pathol Lab Med
study of 14 specimens. Cancer (Cancer Cytopathol) 2003;99:301–309. 1996;16:243–252.

Ch003-F06731.indd 92 10/26/2006 10:23:07 AM


4 Soft Tissue
Marlo M Nicolas Ritu Nayar
Fadi W Abdul-Karim
• •

when compared to the primary tumor. Radiation and


INTRODUCTION chemotherapy can induce changes that mimic recurrent
disease. In addition, many non-neoplastic, rapidly pro-
liferating or exuberant reactive lesions can have cyto-
Fine needle aspiration biopsy (FNAB) is a safe and logic atypia that approximates that seen in higher-grade
reliable method for the assessment of soft tissue tumors sarcomas. Awareness of these changes helps avoid
and tumor-like lesions. Many of these entities have overdiagnosis.
distinctive clinical attributes such as specific age ranges This chapter focuses on the cytologic features of soft
and predilection for certain body sites, so that a limited tissue tumors and tumor-like lesions that are most com-
list of differential diagnoses or a definitive diagnosis monly encountered in the practice of cytopathology.
can be rendered after cytologic evaluation and clinical
correlation. The line of communication between the
cytopathologist, radiologist, and clinician should always
be open to assure an optimal interpretation of the avail- ADIPOSE TISSUE TUMORS
able material.
Familiarity with the various spectra of the histo-
pathologic features of soft tissue lesions is important for LIPOMA
interpretation of the FNAB specimens. The cytomor-
phology of these lesions often reflects the histologic
features by which most of these entities are defined. An CLINICAL FEATURES
accompanying cell block (CB) may prove useful in some
instances due to the approximation to the native histo-
logic architecture of the lesion. A needle core biopsy Lipomas are composed of mature adipose tissue and
(NCB) can establish the histologic specific subtype of are the most common benign soft tissue tumors. They
a lesion, especially when a diagnosis of sarcoma, not usually occur in older adults and have a male predilec-
otherwise specified is rendered from FNAB materials. tion. Lipomas present as a painless mass in the head
Nevertheless, surgeons are often satisfied with the and neck region, shoulder, back, and extremities. Com-
diagnosis of a low- or high-grade sarcoma, and further plete excision is usually curative.
subclassification may not be necessary in the immediate The admixture of other mesenchymal elements
management. NCBs are most effective in establishing results in several variants: spindle cells (spindle cell
the diagnosis of soft tissue lesions when employed in lipoma), capillaries (angiomyolipoma), smooth muscle
instances where the FNAB is unsatisfactory. In the (myolipoma), and giant ‘floret’ cells (pleomorphic
setting of a non-diagnostic NCB or FNAB, an open or lipoma). In addition to subcutaneous tissue, lipomas
excisional biopsy should be obtained for further may arise in skeletal muscle (muscular lipoma) and
evaluation. synovial connective tissue (lipoma arborescens).
Performance of the FNAB by an experienced pathol-
ogist with immediate assessment of adequacy will reduce
the number of unsatisfactory specimens and results in
better material for interpretation. The pathologist can LIPOMAS – DISEASE FACT SHEET
also, following rapid assessment, triage the specimens
Incidence and Location
for ancillary studies such as DNA ploidy, cytogenetics,
៉ Most common soft tissue tumor
and electron microscopy (EM). In selected cases, the
៉ Trunk, proximal extremities, and head and neck
judicious application of immunohistochemical stains
done on the CB or NCB will aid in narrowing down the
Gender, Race and Age Distribution
differential diagnosis.
៉ Male predominance
FNAB is most useful for documenting recurrence
៉ No racial predilection
or metastases in a patient previously diagnosed with ៉ Adult (more than 40–60 years)
sarcoma. Recurrent and metastatic sarcomas, however,
may show significant variations in cell morphology
93
94 FINE NEEDLE ASPIRATION CYTOLOGY

PATHOLOGIC FEATURES

Histologically, lipomas are typically composed of


mature adipose tissue indistinguishable from non-
neoplastic fatty tissue.
FNAB of lipomas consists of mature adipocytes,
usually as tissue fragments and rarely as singly dis-
persed cells (Fig. 4-1). The adipocytes are round or
ovoid and have a dominant cytoplasmic vacuole that
does not indent the eccentrically located nucleus. Capil-
laries may be seen separating clusters of adipocytes. In
addition to the mature adipose tissue, spindle cells and
large multinucleated tumor cells are seen in aspirates
of spindle cell and pleomorphic lipomas, respectively.
FNAB of intramuscular lipomas and angiolipomas can
FIGURE 4-1
yield other mesenchymal elements, such as skeletal
Lipoma: FNAB. The aspirated adipocytes are cohesively clustered. The
muscle and excessive amounts of capillaries, cells have abundant cytoplasm and peripherally located small nuclei.
respectively. Papanicolaou stain, high power.

ANCILLARY STUDIES DIFFERENTIAL DIAGNOSIS AND PITFALLS

The tumor cells of lipomas are positive for vimentin The cytologic features of lipomas may be indistinguish-
and S-100 protein. Up to 75% of lipomas can have able from those of atypical lipomatous tumor (ALT)/
an abnormal karyotype, the most common of which well-differentiated liposarcoma (WDLS), which may
involves the long arm of chromosome 12. consist predominantly of mature adipose tissue. Lipo-
blasts and atypical adipocytes are expectedly rare in
these aspirates and are often difficult to recognize.
Cytogenetic studies may assist in recognizing these
LIPOMAS – PATHOLOGIC FEATURES tumors.
The cells of lipomas are identical to those of non-
Cytopathologic Findings neoplastic subcutaneous fat. The possibility of sampling
៉ Aspirated material consists of fatty droplets when extruded on to the subcutaneous fat should be entertained when the
the slide; fairly cellular to hypocellular smears; mature adipose aspirates yield mature adipocytes, especially if detailed
cells have a dominant cytoplasmic vacuole, nuclei are small and clinical information is not provided or the FNAB is
eccentrically placed performed by health professionals other than the cyto-
៉ Spindle cell lipomas: spindle cells admixed with mature adipose pathologist interpreting the case.
cells
៉ Pleomorphic lipomas: multinucleated cells and mature adipose
cells
៉ Other variants may consist of other types of mesenchymal tissue LIPOSARCOMA
admixed with mature adipose cells: skeletal muscle (intramuscular
lipoma), chondroid matrix (chondrolipoma), bone marrow elements
(myelolipoma), and smooth muscle (myolipoma) CLINICAL FEATURES
Immunohistochemistry
៉ Adipocytes are positive for vimentin and S-100 protein; spindle Liposarcomas are malignant mesenchymal tumors with
cells of spindle cell lipomas are positive for CD34 fatty differentiation.
៉ Tumor cells are negative for epithelial, smooth muscle, skeletal Four histologic types are recognized: atypical lipoma-
muscle, and neuroendocrine markers tous tumor (ALT)/well-differentiated liposarcoma
(WDLS), dedifferentiated liposarcoma (DDLS), myxoid
Genetics
liposarcoma (MLS)/round cell liposarcoma (RCLS), and
៉ Approximately 55–75% have an abnormal karyotype; 66% involve
pleomorphic liposarcoma (PLS).
12q13–15
Liposarcomas are tumors of adults and infrequently
occur in children. More than 90% of the liposarcomas
Differential Diagnosis and Pitfalls
are ALTs/WDLSs and MLSs/RCLSs. PLSs and DDLSs
៉ Atypical lipomatous tumor/well-differentiated liposarcoma
account for less than 10% of liposarcomas. PLSs are
៉ Normal subcutaneous adipose tissue (adjacent normal adipose
tissue is sampled and needle missed the lesion)
quite rare.
ALTs and WDLSs are composed of mature adipose
tissue with scattered atypical cells. Superficial tumors of
CHAPTER 4 Soft Tissue 95

LIPOSARCOMA – DISEASE FACT SHEET LIPOSARCOMA – PATHOLOGIC FEATURES

Incidence Cytopathologic Findings


៉ Atypical lipomatous tumor(ALT)/well-differentiated liposarcoma ៉ Atypical lipomatous tumor(ALT)/well-differentiated liposarcoma
(WDLS): 50% of liposarcomas (WDLS): variably cellular smears of mature adipose tissue
៉ Myxoid liposarcoma (MLS)/round cell liposarcoma (RCLS): 30–40% indistinguishable from non-neoplastic fat; scattered atypical
of liposarcomas cells; lipoblasts are scarce
៉ Dedifferentiated liposarcoma (DDLS): 5% of liposarcomas ៉ Myxoid liposarcoma (MLS)/round cell liposarcoma (RCLS): uniform
៉ Pleomorphic liposarcoma (PLS): <5% of liposarcomas round or oval cells; myxoid matrix appears as purple or magenta
in Romanowsky-stained smears; branching capillary network;
Location signet-ring lipoblasts
៉ ALT: superficial trunk and extremities ៉ Dedifferentiated liposarcoma (DDLS): clusters of spindle or
៉ WDLS: deep soft tissue of thigh, retroperitoneum, paratesticular pleomorphic cells without intervening mature adipose tissue;
area, and mediastinum no lipoblasts; may have cells similar to ALT/WDLS
៉ MLS/RCLS: lower extremities (70% in thigh) ៉ Pleomorphic liposarcoma (PLS): highly cellular aspirate with
៉ DDLS: Retroperitoneum, mediastinum, and paratesticular region pleomorphic or bizarre-appearing tumor cells; pleomorphic
៉ PLS: lower extremities, less often retroperitoneum and (mulberry) lipoblasts
mediastinum
Immunohistochemistry
Gender, Race, and Age Distribution ៉ ALT/WDLS: atypical cells are positive for S-100 protein
៉ No sex or race predilection ៉ MLS/RCLS: tumor cells are positive for vimentin and S-100
៉ ALT/WDLS: middle-aged adults (peak – 6th decade) protein
៉ MLS/RCLS: young and middle-aged adults (peak – 4th decade) ៉ DDLS: dedifferentiated areas usually express antibodies
៉ DDLS: middle-aged adults expected of the morphology; residual ALT/WDLS is positive for
៉ PLS: elderly patients (>50 years) S-100 protein
៉ PLS: tumor cells are positive for vimentin and S-100 protein;
epithelioid variant may be positive for cytokeratin

Genetics
the trunk or extremities are designated as ALTs, while
៉ ALT/WDLS: supernumerary ring and giant marker chromosomes;
WDLSs apply to similar tumors in deep-seated locations
amplified sequences of 12q14–15 and, less frequently, 12q21–22
such as the retroperitoneum, paratesticular region, and
and 1q21–25
mediastinum. A liposarcoma is categorized as a dedif- ៉ MLS/RCLS: t(12;16)(q13;p11) in 90% of cases; t(12;22)(q13;q12)
ferentiated type when an area of non-lipogenic sarcoma in 10% of cases
is present in a newly diagnosed ALT/WDLS (de novo), ៉ DDLS: supernumerary ring and giant marker chromosomes;
in a local recurrence of an ALT/WDLS, or in a distant amplified sequences of 12q14–15 and, less frequently, 12q21–22
metastasis following or with a concurrent diagnosis and 1q21–25
of ALT/WDL. DDLSs have a significant potential for ៉ PLS: complex cytogenetic aberrations
metastasis.
MLSs and RCLSs have identical cytogenetic abnor- Differential Diagnosis and Pitfalls
malities. Histopathologically, RCLSs are considered a ៉ ALT/WDLS: lipoma, fat necrosis, and non-neoplastic fat
less differentiated type of MLSs. The presence of an ៉ MLS/RCLS: extraskeletal myxoid chondrosarcoma and low-grade
RCLS component imparts a more aggressive behavior. fibromyxoid sarcoma; small round cell tumors in pure or
PLSs are highly malignant tumors characterized by an predominantly RCLS
៉ DDLS: may be indistinguishable from other sarcomas including
early onset of metastasis and a high mortality rate.
malignant fibrous histiocytoma, malignant peripheral nerve
sheath tumor, and leiomyosarcoma if ALT/WDLS areas are not
sampled
៉ PLS: malignant fibrous histiocytoma, other pleomorphic
PATHOLOGIC FEATURES sarcomas and carcinomas

Histologically, ALTs/WDLSs are composed mainly of


mature adipose tissue with fibrous septae containing
atypical cells. Lipoblasts may be present. The foci of lipoblasts are rarely present. The diagnosis of ALT/
DDLSs are usually high grade and are indistinguish- WDLS usually necessitates a biopsy. DDLSs are recog-
able from other sarcomas. MLSs/RCLSs have charac- nized by the presence of a high-grade non-lipomatous
teristic branching capillaries and uniform tumor cells. component in a background of ALT/WDL (Fig. 4-2).
RCLSs are considerably more cellular than MLSs. PLSs MLSs/RCLSs consist mainly of uniform round or ovoid
are characterized by extremely atypical cells and lipo- tumor cells with a high nuclear to cytoplasmic (N/C)
blasts of the pleomorphic (mulberry) type. ratio and occasionally signet-ring lipoblasts in a back-
Smears of ALTs/WDLSs consist predominantly ground of abundant myxoid matrix (Figs 4-3 & 4-4).
of mature adipose tissue indistinguishable from non- Branching capillaries are conspicuously present. Bizarre
neoplastic fatty tissue and lipoma. The atypical cells may tumor cells and a variable number of pleomorphic
be sparse and difficult to recognize. Multivacuolated lipoblasts typify PLSs (Fig. 4-5).
96 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 4-2 FIGURE 4-4


Dedifferentiated liposarcoma: touch preparation. The dedifferentiated Myxoid liposarcoma: needle core biopsy. Signet-ring lipoblasts are
foci are cellular and usually densely aggregated. These tumor cells are randomly scattered. Note the relative uniformity of the lesional cells
indistinguishable from other high-grade sarcomas. Demonstration of without unusually large or bizarre tumor cells. H&E stain, medium
such cells in a patient with a previous or concurrent diagnosis of atypi- power.
cal lipomatous tumor/well-differentiated liposarcoma suggests trans-
formation of the tumor to dedifferentiated liposarcoma. Diff-Quik stain,
high-power.
ALTs/WDLSs and DDLSs have similar genetic
abnormalities. Both tumors have supernumerary ring
and giant cell marker chromosomes mostly derived from
the 12q14–15 karyotypic abnormality. The majority of
MLSs/RCLSs have t(12;16)(q13;p11) and about 10% of
these tumors show t(12;22)(q13;q12). PLSs do not show
a specific genetic aberration, and the cytogenetic make-
up of these tumors is complex.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

ALTs/WDLSs may consist predominantly of mature


adipose tissue and can be indistinguishable from
normal fat or lipoma. Aspirates of mature-appearing
adipose tissue in deep-seated locations should be
examined carefully for any atypical fibroblast-like cells.
In general, a biopsy is necessary to establish this diag-
FIGURE 4-3 nosis. Necrotic non-neoplastic fat may harbor histio-
Myxoid liposarcoma: FNAB. The aspirate is moderately cellular. The cytes with large nuclei which simulate the atypical cells
tumor cells are round or oval and of uniform size. The stromal back- of ALTs/WDLSs. DDLS components are indistinguish-
ground is myxoid. Prominent branching capillaries are present in the
middle of the tumor clusters. Diff-Quik stain, medium power. able from those of high-grade sarcomas such as malig-
nant fibrous histiocytoma, malignant peripheral nerve
sheath tumor, and leiomyosarcoma. The diagnosis of
DDLS can only be established when foci of ALT/WDLS
ANCILLARY STUDIES are demonstrated in the current specimen or a previous
history of ALT/WDLS is elicited.
The presence of vascular channels and limited atypia
The mature adipocytes and lipoblasts are positive for of tumor cells, in a myxoid background is not unique
vimentin and S-100 protein. With the exception of for MLS/RCLS and may be observed in other neoplasms
some epithelioid variants of PLSs, all types of liposar- such as low-grade fibromyxoid sarcoma and extra-
comas are negative for cytokeratins (CK). Dedifferen- skeletal chondromyxoid sarcoma. A biopsy is necessary
tiated foci other than malignant fibrous histiocytoma to discriminate these lesions.
are reactive to antibodies expected for the morphology, In the absence of mulberry-type lipoblasts, PLSs
e.g. rhabdomyosarcomatous areas are MyoD1- and are indistinguishable from other poorly differentiated
myogenin-positive. mesenchymal and epithelial malignancies.
CHAPTER 4 Soft Tissue 97

A B

FIGURE 4-5
Pleomorphic liposarcoma: touch preparation. Pleomorphic cells and lipoblasts are shown here. The cytoplasmic vacuoles indent the peripherally
displaced nuclei. Due to variable numbers of lipoblasts, these may not be demonstrated in FNAB. Diff-Quik stain, high power.

FIBROBLASTIC AND FIBROHISTIOCYTIC NODULAR FASCIITIS – PATHOLOGIC FEATURES


TUMORS
Cytopathologic Findings
៉ Cellular aspirate; the mildly atypical tumor cells are spindle or
NODULAR FASCIITIS plump cells; stellate or ganglion-like cells may be present; small
but sometimes prominent nucleoli; infl ammatory infiltrates and
mitotic figures are variably present
CLINICAL FEATURES ៉ Myxoid or collagenous stroma
៉ Depending on the site of lesion, degenerated tissue such as fat
necrosis or skeletal muscle may accompany the lesional cells
Nodular fasciitis (NDF) usually arises in the subcuta-
neous tissue of the extremities, head and neck, or trunk Immunocytochemistry
regions. The mass lesion often abuts the underlying ៉ Positive for vimentin, smooth muscle actin, and muscle-specific
adjacent fascia. NDF may develop within blood vessels actin, and rarely for desmin
(intravascular NDF), skeletal muscle (intramuscular ៉ Negative for S-100 protein, CD34, bcl-2, and cytokeratin
NDF), skull (cranial NDF), and, rarely, in visceral
organs. The typical presentation is that of a rapidly Genetics
growing painful mass in a young adult. A history of ៉ Diploid by DNA ploidy analysis
៉ t(15;15)(q13;q22 or q25) was described in three cases
antecedent trauma is often reported.
Differential Diagnosis and Pitfalls
PATHOLOGIC FEATURES ៉ Benign and malignant spindle cell and myxoid tumors including
solitary fibrous tumor, low-grade fibromyxoid sarcoma,
fibrosarcoma, malignant peripheral nerve sheath tumor, and
Histologically, NDF is composed of spindly and wavy leiomyosarcoma
៉ Spindle cell carcinoma
cells haphazardly arranged or in short fascicles (tissue

NODULAR FASCIITIS – DISEASE FACT SHEET culture pattern). The stromal background is characteri-
stically myxoid and patchy in distribution. Mitotic
Incidence and Location
figures can be readily found. Older lesions are less
៉ Not uncommon mitotically active with more fibrocollagenous stroma.
៉ Upper and lower extremities, head and neck, and trunk; rarely
The FNAB smears of NDF are cellular and composed
within skeletal muscle, vessels, and skull/scalp
of tightly clustered and singly scattered spindle, poly-
Gender, Race, and Age Distribution
gonal, or stellate cells in a myxoid background (Fig.
4-6). The tumor cells are relatively uniform round or
៉ No sex or race predilection
៉ Peak incidence between 2nd and 5th decades
spindled with a smooth outline. They have cytoplasmic
processes, and centrally located nucleoli reminiscent of
a ganglion cell are sometimes identified (proliferative
98 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 4-6
Nodular fasciitis: FNAB. The aspirated materials are often cellular. The lesional cells are spindle or stellate with a moderate N/C ratio, inconspicu-
ous nucleoli, and abundant cytoplasm. Myxoid stroma often predominates. A few fragments of skeletal muscle are also present. Papanicolaou stain,
medium power.

fasciitis). A variable number of inflammatory cells


including neutrophils, eosinophils, and mononuclear FIBROMATOSIS – DISEASE FACT SHEET
cells may be seen. Mitotic figures are usually present.
Incidence
៉ Superficial: common among those of northern and eastern
European descent; 20% of 65 years or older
ANCILLARY STUDIES ៉ Desmoid-type (aggressive): 2–4 per million population per year

Location
The tumor cells are usually positive for vimentin,
៉ Superficial: palmar and plantar regions
smooth muscle actin (SMA), and muscle-specific actin
៉ Desmoid-type (aggressive): abdominal wall, retroperitoneum, trunk,
(MSA). Desmin may also be focally expressed. There
shoulder and proximal extremities
is no immunoreactivity with S-100 protein, melanoma
markers (HMB-45 and Melan A), CK, CD34, or bcl-2. Gender, Race, and Age Distribution
The tumor cells are diploid by flow cytometry. Cytoge-
៉ Superficial:
netic analysis performed on a few reported cases ៉ Palmar fibromatosis: 3–4 times more common in men,
showed t(15;15)(q13;q22 or q25). particularly those of northern or eastern European descent;
rare in non-Caucasians
៉ Plantar fibromatosis: slight male predilection; 35% of cases
DIFFERENTIAL DIAGNOSIS AND PITFALLS are 35 years or younger
៉ Desmoid-type (aggressive):
៉ Children: no sex or race predilection; extra-abdominal
NDFs are perhaps the most common benign tumors to be location is more common
៉ Child-bearing age: female predilection; abdominal location is
mistaken for malignancy on FNAB. An important clue
more common
to their diagnosis is the clinical information of a rapidly ៉ Older age: no sex predilection; equal distribution (abdominal
growing mass. Cytologically, several benign and malig- and extra-abdominal locations)
nant spindle cell neoplasms can have similar features to
those of the cells of NDFs. Oftentimes, a definitive diag-
nosis can not be made on smear preparations, and biopsy
or excision is necessary for definitive diagnosis.
fibroblastic differentiation. They are usually divided
based on their location into superficial (palmar or
FIBROMATOSIS
Dupuytren FM, and plantar or Ledderhose disease)
and desmoid types (aggressive FM).
CLINICAL FEATURES Palmar FMs often present as subcutaneous nodules
with flexion contractures of the fingers. The overlying
skin of the palm and fingers may be dimpled. In most
Fibromatoses (FMs) are locally infi ltrative tumors instances, the subcutaneous nodules of plantar FMs
composed of spindle cells with fibroblastic and myo- are asymptomatic. Pain on pressure from standing or
CHAPTER 4 Soft Tissue 99

A B

FIGURE 4-7
Fibromatosis: FNAB. (A) The aspirates are usually paucicellular in tumors with collagenous stroma. The tumor cells are spindle-shaped. Groups of
tumor cells (B) are held by fibrocollagenous stroma. Singly scattered nuclei are often devoid of their cytoplasm. Papanicolaou stain, medium
power.

walking may be the initial presentation. These lesions


are usually clinically diagnosed and not subject to FIBROMATOSIS – PATHOLOGIC FEATURES
FNAB.
Desmoid-type FMs are further subdivided into Cytopathologic Findings
abdominal, intra-abdominal, and extra-abdominal types. ៉ Cellularity depends on the stroma: myxoid lesions tend to
These tumors have a tendency for local recurrence, aspirate better than collagenous lesions
either from multicentricity or residual tumor. Although ៉ Clusters or singly scattered cells; tumor cells have ovoid or
they do not have the capacity for distant spread, signifi- round nuclei with a smooth nuclear membrane and fine
chromatin material; elongated cytoplasm
cant morbidity through local destructive invasion may
result.
Immunohistochemistry
Abdominal FMs often present as asymptomatic
៉ Positive for vimentin, smooth muscle actin, muscle-specific
abdominal wall mass lesions following previous surgery
actin, and β-catenin; may be positive for desmin
such as caesarian section. Owing to their location, ៉ Negative for S-100 protein, CD34, and cytokeratin
intra-abdominal FMs are symptomatic if vital structures
are compromised. Extra-abdominal FMs present as Genetics
palpable masses in the neck, shoulder, trunk, and ៉ No specific karyotypic abnormality
pelvic girdle. ៉ No specific genetic abnormality
៉ Patients with familial polyposis and, rarely, sporadic
fibromatosis may have APC gene inactivation

Differential Diagnosis and Pitfalls


PATHOLOGIC FEATURES ៉ Benign and malignant spindle cell tumors such as
myofibroblastoma, neurofibroma, solitary fibrous tumor,
leiomyoma, benign fibrous histiocytoma, dermatofibrosarcoma
Histologically, the bland-appearing spindle cells are protuberans, and low-grade fibromyxoid sarcoma
arranged in longitudinal fascicles with a collagenous or
myxoid background.
The superficial FMs have a typical clinical presenta-
tion and are excised without preoperative cytologic
evaluation. Desmoid-type FMs are clinically indistin-
guishable from other soft tissue tumors, and FNAB may
be performed as part of the initial evaluation. The col- ANCILLARY STUDIES
lagenous stroma may lead to paucicellular aspirates with
only singly scattered spindle cells often stripped of their
cytoplasm (Fig. 4-7). Clusters of these bland-appearing The tumor cells are immunoreactive for vimentin,
spindle cells are commonly seen. The tumor cells have SMA, MSA, and β-catenin. Rarely, desmin is also
oval and uniform nuclei with a smooth outline, and a expressed. The tumor cells are usually negative for
moderate amount of cytoplasm. If present, the myxocol- S-100, CD34, and CK. There are no known genetic
lagenous stroma is characteristically metachromatic on aberrations in FMs, although patients with familial
Romanowsky-based stains. polyposis (FP) may have APC gene inactivation.
100 FINE NEEDLE ASPIRATION CYTOLOGY

DIFFERENTIAL DIAGNOSIS AND PITFALLS


DERMATOFIBROSARCOMA PROTUBERANS –
PATHOLOGIC FEATURES
On FNAB, the spindle cells of FMs are morphologically
Cytopathologic Findings
indistinguishable from many spindle cell lesions includ-
ing myofibroblastoma, neurofibroma, solitary fibrous ៉ Fairly cellular aspirates composed of tightly clustered spindle
cells and scattered single cells devoid of cytoplasm; tumor cells
tumor, benign fibrous histiocytoma, dermatofibrosar-
are spindly with ovoid, smooth-outlined nuclei, inconspicuous
coma protuberans, and low-grade fibromyxoid sarcoma. nucleoli, and bipolar cytoplasmic processes
More aggressive tumors such as desmoplastic mela- ៉ Myxocollagenous stroma
noma and spindle cell carcinoma may also have a
similar appearance. Immunohistochemical work-up Immunohistochemistry
can assist in the differential. The diagnosis of FM is ៉ Positive for vimentin and CD34
usually strongly suspected on clinical and radiologic ៉ Negative for S-100 protein, actins, desmin, cytokeratin, and
grounds. NCB or excisional biopsy may be necessary epithelial membrane antigen
for a definitive diagnosis.
Genetics
៉ Ring chromosomes from chromosome 17 and 22
៉ t(17;22)(q22;q13) resulting in COL1A–PDGFB fusion transcript
DERMATOFIBROSARCOMA PROTUBERANS
Differential Diagnosis and Pitfalls
៉ Benign and malignant spindle cell tumors such as
CLINICAL FEATURES myofibroblastoma, neurofibroma, benign fibrous histiocytoma,
fibromatosis, low-grade fibromyxoid sarcoma, and fibrosarcoma

Dermatofibrosarcoma protuberans (DFSP) presents as


a slow-growing, pink or erythematous skin plaque or
as a subcutaneous mass. The tumors occur most fre-
quently in the trunk and proximal extremities in aggregates with ill-defined boundaries (Fig. 4-8). The
middle-aged patients without sex predilection. DFSPs spindled tumor cells have elongated or ovoid nuclei
are locally aggressive tumors with a high recurrence with fine chromatin and inconspicuous nucleoli. Their
rate following excision. Transformation to fibrosar- cytoplasm may demonstrate bipolar cytoplasmic pro-
coma is rare but well-documented. Rare cases of meta- cesses. The background stroma may be myxoid, collag-
static DFSPs have been reported, typically following enous, or fibrillary, and exhibits metachromasia with
multiple recurrence and prolonged follow-up. Romanowsky-based stains. Mast cells and Touton-type
giant cells may be seen. Aspirated fragments of DFSPs
may display the characteristic storiform pattern. Trans-
formation to a higher-grade sarcoma (e.g. fibrosarcoma)
PATHOLOGIC FEATURES may not be possible to establish on FNAB specimens
due to lack of distinctive cytologic features unless
frankly atypical tumor cells are present.
Histologically, DFSPs are composed of spindle cells
arranged in short interlacing fascicles forming a stori-
form or cartwheel pattern. The neoplastic cells are
relatively uniform and show minimal pleomorphism.
Infi ltrative tumor cells in the deep subcutaneous tissue
often surround individual or groups of fat cells.
The aspirated material is usually cellular. The tumor
cells are either individually dispersed or in small

DERMATOFIBROSARCOMA PROTUBERANS – DISEASE FACT SHEET

Incidence and Location


៉ 1.8% of soft tissue tumors
៉ Trunk, proximal portion of the extremities, and head and neck

Gender, Race, and Age Distribution


៉ No sex or race predilection FIGURE 4-8
៉ Peak incidence: young and middle-aged adults Dermatofibrosarcoma protuberans: touch preparation. The tumor cells
have fine chromatin and are devoid of nucleoli. Significant pleomor-
phism of tumor cells is not demonstrated. Diff-Quik stain, high power.
CHAPTER 4 Soft Tissue 101

ANCILLARY STUDIES
FIBROSARCOMA – PATHOLOGIC FEATURES

The tumor cells of DFSPs are positive for vimentin and Cytopathologic Findings
CD34, and negative for S-100 protein, SMA, MSA, ៉ Cellular aspirate; uniform spindle tumor cells; considerable
desmin, CK, and epithelial membrane antigen (EMA). cellular atypia in poorly differentiated tumors
៉ Myxocollagenous stroma
The main reported cytogenetic abnormality in DFSPs
is a ring chromosome derived from chromosomes 17
Immunohistochemistry
and 22. This translocation, t(17;22)(q22;q13), results
៉ Positive for vimentin; may be focally positive for smooth
in a chimeric transcript, COL1A-PDGFB (collagen
muscle actin; fibrosarcoma arising from dermatofibrosarcoma
type 1–platelet-derived growth factor beta), which can protuberans is positive for CD34
be detected by reverse transcriptase-polymerase chain ៉ Negative for epithelial markers (cytokeratin and epithelial
reaction (RT-PCR) or fluorescence in situ hybridization membrane antigen), muscle markers (MyoD1, myogenin, and
(FISH). The same genetic abnormality is seen in giant calponin), and neural/melanoma markers (S-100, HMB-45,
cell fibroblastoma, a closely related entity. Melan A)

Genetics
DIFFERENTIAL DIAGNOSIS AND PITFALLS ៉ Complex karyotype

Differential Diagnosis and Pitfalls


The spindle cell lesions listed under fibromatosis are ៉ Benign and malignant spindle cell tumors such as
included in the differential diagnosis of DFSP. The myofibroblastoma, neurofibroma, benign fibrous histiocytoma,
typical immunophenotype of DFSP, however, is not fibromatosis, low-grade fibromyxoid sarcoma, and
observed in these entities. The presence of areas that leiomyosarcoma
are negative for CD34 together with the appropriate ៉ Pleomorphic areas of fibrosarcoma are indistinguishable from
cellular morphology suggests transformation to FS. other high-grade sarcomas
When available, molecular studies can assist in the
diagnosis of DFSPs.

fascicles that pull towards opposite directions (herring-


FIBROSARCOMA bone pattern). A collagenous background is variably
present. Fibrosarcomas have become a diagnosis
of exclusion after entities (e.g. synovial sarcoma) of
CLINICAL FEATURES overlapping histology are classified under separate
categories.
The aspirated material is often cellular. Tissue frag-
Fibrosarcomas most often arise in the upper trunk,
ments often show fibrocollagenous stroma in which
proximal extremities, and head and neck region of
tumor cells are embedded (Fig. 4-9). Individually
adults. They tend to be slow growing, and may be asso-
scattered neoplastic cells are spindle-shaped and often
ciated with pain or paresthesia secondary to mass
devoid of cytoplasm. When intact, the cytoplasmic pro-
effect. A preceding diagnosis of DFSP may be extracted
cesses are elongated at both ends of the cell. Consider-
from the history.
able pleomorphism may be appreciated in tumors with
less differentiated components.
PATHOLOGIC FEATURES

The characteristic feature of fibrosarcomas is the pres- ANCILLARY STUDIES


ence of spindly tumor cells forming long sweeping
FSs express vimentin but none of the more specific
antibodies. Epithelial, neural, and melanoma markers
FIBROSARCOMA – DISEASE FACT SHEET are negative. At present, a specific genetic abnormality
has not been described.
Incidence and Location
៉ Rare tumors (other histologically similar tumors should be ruled
out)
៉ Deep soft tissue of lower extremities, trunk, head and neck DIFFERENTIAL DIAGNOSIS AND PITFALLS

Gender, Race, and Age Distribution


The entities in the differential diagnosis of fibrosarco-
៉ No sex or race predilection
៉ Middle-aged and older adults
mas are similar to those listed under fibromatoses. A
more pleomorphic fibrosarcoma is difficult to distin-
guish from high-grade sarcomas. A more definitive
102 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 4-9
Fibrosarcoma: FNAB. FNAB often yields cellular material. Tumors cells are spindled and show variable, moderate pleomorphism. The stroma may
be prominent or entirely non-existent. Component cells are indistinguishable from those in other spindle cell tumors such as synovial sarcomas.
Papanicolaou stain, high power.

diagnosis requires NCB or open biopsy. Expression of function are secondary to mass effect or tumor infi ltra-
an antibody or set of antibodies known to more or tion. Metastasis to the lungs may be present at the time
less define an entity should exclude fibrosarcoma from of diagnosis. MMFHs are slow-growing subcutaneous
consideration. tumors that frequently arise from the extremities;
ulceration of the overlying skin from tumor infi ltration
may be present. Lower-grade MMFHs may have an
indolent behavior.
MALIGNANT FIBROUS HISTIOCYTOMA

CLINICAL FEATURES PATHOLOGIC FEATURES

Two types of malignant fibrous histiocytomas (MFHs) Malignant fibrous histiocytomas (MFHs) are malig-
are considered here: pleomorphic MFH (PMFH), and nant soft tissue tumors composed of tumor cells
myxoid MFH (MMFH) or myxofibrosarcoma. PMFHs without definitive morphologic, immunohistochemical,
usually present as a rapidly growing deep-seated mass. and ultrastructural evidence of specific tissue differen-
The common locations are the proximal extremities, tiation. The tumor cells are closely related to fibro-
trunk. and retroperitoneum. Pain, swelling, and loss of blasts/myofibroblasts.
Histologically, PMFHs are hypercellular with mark-
edly atypical tumor cells and brisk mitotic activity. An
admixture of spindle and polygonal cells is common and
MALIGNANT FIBROUS HISTIOCYTOMA – DISEASE FACT SHEET a storiform pattern is typically present. MMFHs have
variable cellularity composed of spindle cells with few
Incidence scattered pleomorphic cells in a myxoid background.
៉ Pleomorphic MFH: 1–2 cases per 100,000 per year Relatively thick-walled vessels associated with fibrosis
៉ Myxoid MFH: common sarcoma in elderly patients are usually recognized.
FNAB of PMFHs consists of a pleomorphic popula-
Location tion of spindle, polygonal, and multinucleated cells (Fig.
៉ Pleomorphic MFH: lower extremity and trunk (deep or cutaneous) 4-10). The tumor cells may cling to vascular structures,
៉ Myxoid MFH: lower and upper extremities (proximal part) giving a pseudopapillary appearance. Erythrophagocy-
tosis can be identified. The neoplastic cells do not show
Gender, Race, and Age Distribution cytomorphologic evidence of specific tissue differentia-
៉ Slight male predilection tion. Mitoses are easily recognized. FNAB of MMFHs
៉ No race predilection
shows hypocellular or moderately cellular aspirates
៉ Pleomorphic MSH: most common sarcoma in persons aged 40 years
composed of a relatively less pleomorphic population
or older
៉ Myxoid MSH: most common in elderly patients
of tumor cells (usually spindly) with randomly inter-
spersed bizarre cells (Fig. 4-11). A copious myxoid
stroma is characteristic.
CHAPTER 4 Soft Tissue 103

A B

FIGURE 4-10
Malignant fibrous histiocytoma, pleomorphic type: FNAB. The hypercellular aspirates are composed of pleomorphic tumor cells without specific
cytomorphologic differentiation. Mitotic figures are easy to find. Diff-Quik stain, high power.

MALIGNANT FIBROUS HISTIOCYTOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Pleomorphic MFH: cellular aspirates, bizarre tumor cells, mitotic
figures abound; fibrocollagenous or myxoid stroma
៉ Myxoid MFH: moderately cellular aspirates, spindle and polygonal
tumor cells, nuclear to cytoplasmic ratio is lower than in
pleomorphic MFH; prominent myxoid stroma

Immunohistochemistry
៉ Pleomorphic MFH: positive for vimentin; rare cells positive for
epithelial (cytokeratin and epithelial membrane antigen),
muscle (actin, desmin, MyoD1, and myogenin), and other
markers of specific tissue differentiation
៉ Myxoid MFH: positive for vimentin; rare cells positive for
epithelial (cytokeratin and epithelial membrane antigen),
muscle (actin, desmin, MyoD1, and myogenin), and other
markers of specific tissue differentiation
FIGURE 4-11
Malignant fibrous histiocytoma, myxoid type: FNAB. The tumor cells are
Genetics
spindle, stellate, or polygonal cells and less pleomorphic compared with
៉ Complex karyotype the pleomorphic type of MFH. The tumor cells are usually drawn toward
៉ No specific genetic abnormality the immediate periphery of the vascular channels. Note the prominent
myxoid stroma. Diff-Quik stain, high power.
Differential Diagnosis and Pitfalls
៉ Pleomorphic MFH: pleomorphic variants of other sarcomas
including liposarcoma, rhabdomyosarcoma, and leiomyosarcoma
៉ Myxoid MFH: malignant myxoid tumors (low-grade fibromyxoid
sarcoma, myxoid malignant peripheral nerve sheath tumor, and DIFFERENTIAL DIAGNOSIS AND PITFALLS
myxoid liposarcoma)

FNAB of high-grade sarcomas including liposarcoma,


rhabdomyosarcoma, and leiomyosarcoma, and poorly
differentiated carcinomas can be morphologically
ANCILLARY STUDIES indistinguishable from PMFHs. Similarly, aspirates of
MMFHs may resemble myxoid malignant peripheral
nerve sheath tumor and myxoid liposarcoma. Diligent
MFH does not exhibit a specific immunophenotype. search for isolated cells or clusters of cells that demon-
Rare scattered tumor cells may express epithelial and strate specific differentiation (lipoblasts, rhabdomyo-
muscle markers, and S-100 protein. The cytogenetic blasts, keratinization, and mucin production) can
abnormality is complex. exclude the diagnosis of MFH.
104 FINE NEEDLE ASPIRATION CYTOLOGY

NEURAL TUMORS NEURAL TUMORS – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Neurofibroma: hypocellular to fairly cellular aspirates; spindle
NEUROFIBROMA AND SCHWANNOMA tumor cells; fibrocollagenous or myxoid stroma
(NEURILEMMONA) ៉ Schwannoma: fairly cellular aspirates; spindle tumor cells, tight
clusters sometimes predominate; atypical cells representing
degenerative changes may be prominent; fibrocollagenous or
CLINICAL FEATURES myxoid stroma

Immunohistochemistry
NEUROFIBROMA
៉ Neurofibroma: positive for S-100 (less staining intensity
Peripheral neurofibromas often present as visible and compared with schwannoma); negative for cytokeratin and
palpable subcutaneous nodules. Neurofibromas of the epithelial membrane antigen
៉ Schwannoma: positive for S-100 (strong and diffuse); negative
spinal roots may be incidentally discovered or manifest
for cytokeratin and epithelial membrane antigen
symptoms from neural impingement. Neurofibromas
occur in two settings, either as a sporadic case or in a
Electron Microscopy
background of neurofibromatosis type 1 (NF1). Patients
៉ Neurofibroma: admixture of Schwann cells and fibroblasts
with NF1 usually have other stigmata of the syndrome,
៉ Schwannoma: Luse bodies (compact fibrous long spacing
including café-au-lait spots, skin freckling (usually in collagen), long cytoplasmic processes and basal lamina
the axillary and inguinal areas), iris hamartomas (Lisch surrounding Schwann cells
nodules), and other neoplasms (somatostatinoma,
pheochromocytoma). An increased incidence of trans- Genetics
formation to malignant peripheral nerve sheath tumor ៉ Neurofibroma: neurofibromatosis type 1 (NF1) syndrome; loss of
(MPNST) is seen in the setting of NF1. wild-type NF1 allele
៉ Schwannoma: loss of chromosome 22; loss of wild-type NF2
SCHWANNOMA allele

The distribution of schwannomas is similar to that Differential Diagnosis and Pitfalls


of neurofibromas, with few exceptions, including intra- ៉ Benign and malignant spindle cell tumors such as
cranial locations and the propensity to arise from the myofibroblastoma, neurofibroma, benign fibrous histiocytoma,
8th cranial nerve. The tumors are asymptomatic unless fibromatosis, low-grade fibromyxoid sarcoma, and
there is impingement on a nerve, and compression of dermatofibrosarcoma protuberans
the spinal cord or other vital structures.

nous stroma. The histology of NF1-associated neuro-


PATHOLOGIC FEATURES fibromas is similar to that of sporadic cases. Plexiform
neurofibromas, seen almost exclusively in NF1 patients,
consist of thickened nerve trunks and branches.
Histologically, neurofibromas show spindle tumor cells Schwannomas are encapsulated tumors. Alternating
with buckled nuclei in a variably myxoid and collage- cellular (Antoni A) and hypocellular (Antoni B) foci are
usually identified. The nuclei of schwannoma cells may
line up, forming a palisading pattern referred to as
NEURAL TUMORS – DISEASE FACT SHEET Verocay bodies. The intratumoral blood vessels typi-
cally have thick and hyalinized walls.
Incidence In both neurofibromas and schwannomas, a sharp
៉ Neurofibroma: common tumors pain or a tingling sensation extending to the immediate
៉ Schwannoma: common tumors; may be multiple in distribution of the involved nerve may be felt by the
neurofibromatosis type 2 patient during the FNAB procedure.
The cytomorphologic features of neurofibromas and
Location schwannomas are generally similar (Figs 4-12–4-14).
៉ Neurofibroma: skin and subcutaneous tissue, spinal and peripheral Aspirates from both tumors are variably cellular. The
nerves but not cranial nerves tumor cells are arranged in clusters and rarely are indi-
៉ Schwannoma: peripheral nerves, spinal and cranial nerves,
vidually scattered. The tumor cells are spindle-shaped
particularly 8th cranial nerve
or, rarely, epithelioid. The nuclei are uniformly wavy
and devoid of nucleoli. When tissue fragments are
Gender, Race, and Age Distribution
obtained, tumor cells of schwannomas lie in a homoge-
៉ No sex or racial predilection
neous or fibrillary myxocollagenous stroma. Occasion-
៉ Neurofibroma: affects all ages
៉ Schwannoma: affects all ages (peak – 4th to 6th decades)
ally, palisades of cellular nuclei (Verocay bodies) may
be appreciated in schwannomas. The patchy areas of
hypercellularity and rarity of single cells also suggest a
CHAPTER 4 Soft Tissue 105

FIGURE 4-12
Neurofibroma: FNAB. FNAB yield is variably cellular. The spindle tumor
cells are often held together by fibromyxoid stroma. Diff-Quik stain, high
power.

A B

FIGURE 4-13
Neurofibroma: needle core biopsy. The tumor cells display tapered and buckled nuclei and ample cytoplasm with abundant intervening fibrocollage-
nous or fibromyxoid stroma. The tumor cells express S-100 protein (B). A, H&E stain, high-power. B, S-100 protein, high power.

A
B

FIGURE 4-14
Schwannoma: FNAB. The aspirates are variably cellular. Tumors with a thick fibrocollagenous background may yield only a few cell clusters. Tumor
cells have a plump or epithelioid appearance. A, Diff-Quik stain, high power. B, Papanicolaou stain, high power.
106 FINE NEEDLE ASPIRATION CYTOLOGY

diagnosis of a schwannoma rather than a neurofibroma. trunk, breast, and aerodigestive tract. The clinical
Ancient schwannomas may have a few large atypical manifestations of GCTs are site-dependent. In the skin
cells which represent degenerative atypia and do not and aerodigestive tract, the tumor is often discovered
necessarily signify transformation to malignancy. These as an asymptomatic subcutaneous or submucosal slow-
atypical cells are not mitotically active. growing nodule, usually mistaken for a more serious
lesion. GCTs can be multicentric.

ANCILLARY STUDIES
PATHOLOGIC FEATURES
Immunohistochemically, anti-S-100 protein stains
neurofibromas in a patchy distribution and schwan- Histologically, the tumor cells are polygonal or spindle-
nomas in a diffuse and relatively strong manner. shaped with abundant granular cytoplasm and small
round nuclei. The cell borders are usually distinct.
Infi ltration into adjacent tissue is common and not
indicative of a more aggressive behavior. Typically, S-
DIFFERENTIAL DIAGNOSIS AND PITFALLS 100 protein and CD68 are positive, and smooth muscle
markers are negative.
FNAB of GCTs is usually cellular and composed of
Neurofibromas and schwannomas may be mistaken for
clusters and dispersed tumor cells with abundant granu-
other benign spindle cell tumors and low-grade spindle
lar cytoplasm (Figs 4-15 & 4-16). Fragmentation of the
cell sarcomas, particularly in small samples. S-100
granular cytoplasm may impart a hazy background on
protein expression is very helpful in confirming a diag-
low magnification. The cells have centrally located
nosis of neurofibromas and schwannomas. Immuno-
round nuclei. Naked round or oval nuclei with a smooth
histochemical staining for CD34 and smooth muscle
outline and devoid of prominent nucleoli may be
markers can assist in excluding solitary fibrous tumor
observed. Pseudonuclear inclusions may be present.
and smooth muscle and fibroblast/myofibroblast-derived
Cytologic atypia including hyperchromasia, pleomor-
neoplasms such as fibromatosis, low-grade fibromyxoid
phism and prominent nucleoli, necrosis, and mitotic
sarcoma, and dermatofibrosarcoma protuberans. A
activity are not identified in clinically benign GCTs.
recent increase in the size of a pre-existing nerve sheath
tumor warrants a careful assessment for the presence
of atypical cells that may signify transformation to
MPNST.

GRANULAR CELL TUMOR – PATHOLOGIC FEATURES

GRANULAR CELL TUMOR Cytopathologic Findings


៉ Aspirates are usually cellular; tumor cells have abundant and
granular cytoplasm
CLINICAL FEATURES ៉ Disintegrated cytoplasm may impart a necrotic background;
closer examination will show fragments of granular cytoplasm
៉ Naked nuclei are round and uniform with small but sometimes
Granular cell tumors (GCTs) arise commonly in the
prominent nucleoli
tongue, subcutaneous tissue of the extremities and
Immunohistochemistry
៉ Positive for vimentin, S-100 protein, CD68, laminin, and myelin
proteins
៉ Negative for cytokeratin, smooth muscle actin, neurofilament,
GRANULAR CELL TUMOR – DISEASE FACT SHEET and glial fibrillary acidic protein

Incidence and Location Electron Microscopy


៉ Relatively common ៉ Numerous lysosomes
៉ Usual location is the subcutis of the extremities, breast and
trunk, and submucosa of the aerodigestive tract; rarely, the Genetics
lesion(s) may be multiple ៉ No specific karyotypic abnormality
៉ No specific genetic abnormality
Gender, Race, and Age Distribution
៉ Slight female predilection Differential Diagnosis and Pitfalls
៉ More common in Blacks ៉ Oncocytic neoplasms and paraganglioma
៉ Any age bracket; rare in children <5 years; peak – 4th to 6th ៉ Alveolar soft part sarcoma, renal cell carcinoma, and
decades rhabdomyosarcoma
CHAPTER 4 Soft Tissue 107

A B

FIGURE 4-15
Granular cell tumor: FNAB. The aspirates are moderately cellular. The tumor cells are spindle or polygonal with round nuclei and ample, granular
cytoplasm. A, Papanicolaou stain, medium power. B, Diff-Quik stain, high power.

A B

FIGURE 4-16
Granular cell tumor: cell block. The tumor cells typically have granular cytoplasm and a low N/C ratio. The tumor cells are strongly positive for
S-100 protein (B). Note the absence of staining in gastric epithelial cells. A, H&E stain, low power. B, S-100 protein, low power.

chemistry readily differentiates GCT from these neo-


ANCILLARY STUDIES plasms, GCT being positive for S-100 protein and CD68,
and negative for epithelial markers. Ultrastructural
studies demonstrate the abundance of lysosomes in
GCTs stain with S-100 protein (Fig. 4-16B), CD68, and
GCTs and mitochondria in oncocytic neoplasms.
myelin proteins. GCTs do not stain for CK, SMA,
Paragangliomas may resemble GCTs. The absence of
neural fi lament, and glial fibrillary acidic protein
cytoplasmic granularity and expression of CK and neu-
(GFAP). Similar to neurofibroma and schwannoma,
roendocrine markers are more in keeping with para-
GCTs can also express laminin. The cytoplasm is fi lled
ganglioma. In rare instances, alveolar soft part sarcoma,
with numerous lysosomes on EM.
renal cell carcinoma (RCC), and rhabdomyosarcoma
(RMS) may be difficult to separate from GCTs, particu-
larly in small biopsies. The immunophenotypes of
DIFFERENTIAL DIAGNOSIS AND PITFALLS GCTs, RCCs, and RMSs should be relied upon in such
instances.
Gingival GCTs of the newborn, although morpho-
GCTs may be mistaken for oncocytic neoplasms that logically similar to GCTs of other sites, do not express
are known to arise in several organs, including the S-100 protein and the immunohistochemical profile sug-
salivary gland, thyroid, and kidney. Immunohisto- gests a smooth muscle differentiation.
108 FINE NEEDLE ASPIRATION CYTOLOGY

MALIGNANT PERIPHERAL NERVE


SHEATH TUMORS MALIGNANT PERIPHERAL NERVE SHEATH TUMORS –
PATHOLOGIC FEATURES

CLINICAL FEATURES Cytopathologic Findings


៉ Cellular aspirates; tumor cells are spindle and polygonal cells;
less frequently, epithelioid cells or pleomorphic cells may be
Malignant peripheral nerve sheath tumors (MPNSTs) present
present as an enlarging mass with pain or with symp- ៉ Cellular atypia and mitotic figures are readily appreciated:
toms from neural impingement. MPNSTs are most hyperchromasia, coarse chromatin, pleomorphism, and
commonly located in the proximal extremities and prominence of nucleoli
៉ Myxocollagenous stroma
paraspinal region. Approximately 50% of MPNSTs
arise in NF1 patients. Progressive enlargement of an
Immunohistochemistry
existing neurofibroma may herald transformation to
៉ Positive for vimentin; foci of rhabdomyosarcomatous
malignancy.
differentiation are positive for muscle markers
៉ Epithelioid MPNST and epithelial components are positive for
cytokeratin
៉ 50–70% are positive for S-100 protein (faint/patchy)
PATHOLOGIC FEATURES
៉ Negative for CD34, HMB-45, and tyrosinase

Histologically, MPNSTs are hypercellular. Most tumors Genetics


are composed of spindle cells with buckled nuclei. A ៉ Complex karyotype
transition to a rounded or epithelioid tumor cell popu- ៉ May have deletion of the INKA/CDK2A gene (9p)

lation is noted in a minority of tumors. Malignant glan-


Differential Diagnosis and Pitfalls
dular (glandular MPNST) and rhabdomyosarcomatous
(malignant Triton tumor) elements may be present. ៉ Spindle cell MPNSTs: fibrosarcoma, synovial sarcoma,
gastrointestinal stromal tumor, and desmoplastic melanoma
The aspirates are usually very cellular with predomi-
៉ Glandular MPNSTs: biphasic synovial sarcoma and carcinosarcoma
nance of spindle cells in tight clusters and myxocollage- ៉ Epithelioid MPNSTs: carcinoma, epithelioid sarcoma, and
nous stroma (Figs 4-17 & 4-18). The tumor cells may melanoma
also be epithelioid, glandular, or rhabdoid forms. Cellular
evidence of malignancy including hyperchromasia,
pleomorphism, nucleolar prominence, and brisk mitotic
activity are usually evident. Sampling of MPNSTs arising
in a background of neurofibromas might yield benign- and schwannomas, which are strongly reactive.
appearing spindle tumor cells typical of neurofibromas. MPNSTs with epithelioid morphology may be positive
for epithelial markers. The glandular epithelial compo-
nents (glandular MPNST) are expectedly CK-positive.
ANCILLARY STUDIES Foci of rhabdomyosarcomatous differentiation (Triton
tumors) are immunoreactive to skeletal muscle markers
(MyoD1 and myogenin).
MPNSTs typically express S-100 protein in a patchy
and faint distribution, in contrast to neurofibromas
DIFFERENTIAL DIAGNOSIS AND PITFALLS
MALIGNANT PERIPHERAL NERVE SHEATH TUMORS –
DISEASE FACT SHEET Spindle cell MPNSTs may be indistinguishable from
other spindle cell tumors. Remnants of neurofibroma
Incidence or schwannoma in the sampled material may indicate
៉ 5% of soft tissue tumors origin of the malignancy. The stromal cells of synovial
៉ 66% arise from neurofibromas of NF1 patients and less frequently sarcoma (SS) are spindle and uniform, in contrast to
in sporadic neurofibromas the more pleomorphic mesenchymal components of
MPNSTs. The glandular MPNSTs may be mistaken for
Location
biphasic SS. The epithelial components of glandular
៉ Large nerve trunk of the proximal extremities and spinal nerve
MPNSTs are bland-appearing compared to their
roots
counterpart in biphasic SSs. The malignant epithelial
components of carcinosarcoma are considerably more
Gender, Race, and Age Distribution
pleomorphic than the glandular elements of MPNSTs.
៉ Slight predilection for females
Epithelioid MPNSTs may have components that are
៉ No race predilection
៉ Peak at 3rd to 6th decades
morphologically similar with carcinoma, melanoma,
and other sarcomas with epithelioid/epithelial compo-
nents, including epithelioid sarcoma.
CHAPTER 4 Soft Tissue 109

A B

FIGURE 4-17
Malignant peripheral nerve sheath tumor: touch preparation. The tumor is hypercellular and composed of spindle and epithelioid cells. Considerable
cytologic atypia is usually demonstrated. Note the prominent nucleoli. Diff-Quik stain, high power.

RHABDOMYOSARCOMA – DISEASE FACT SHEET

Incidence
៉ Embryonal rhabdomyosarcoma (ERMS): most common sarcoma in
children
៉ Alveolar rhabdomyosarcoma (ARMS): less common than ERMS
៉ Pleomorphic rhabdomyosarcoma (PRMS): rare

Location
៉ ERMS: most common sites are the head and neck, and
genitourinary tract; spindle cell variant arises in scrotal area, and
head and neck; botryoid variant arises in the urinary bladder,
biliary tract, and pharynx
៉ ARMS: most common in the extremities, perineum, paraspinal
area, and paranasal sinuses
៉ PRMS: most common in the lower extremities
FIGURE 4-18
Malignant peripheral nerve sheath tumor: FNAB. The tumor cells are Gender, Race, and Age Distribution
predominantly spindled with elongated cytoplasmic processes. Note the
presence of necrotic debris in the background. Papanicolaou stain, high ៉ ERMS: slight male predilection; incidence is higher in Caucasians;
power. most common variant of rhabdomyosarcoma in children less than
15 years of age (45% occur in those less than 5 years of age)
៉ ARMS: no sex predilection; no racial predilection; more common
in adolescent and young adults
SKELETAL MUSCLE TUMORS ៉ PRMS: male predilection; no race predilection; adult population
(6th decade); very rare in children

RHABDOMYOSARCOMA
extremities (10%), and less commonly in other sites
CLINICAL FEATURES (10%). Head and neck ERMSs usually present as pro-
ptosis, diplopia, or hearing loss. Botryoid ERMSs repre-
sent a distinct subtype arising in a submucosal location,
Rhabdomyosarcomas (RMSs) are generally divided mostly from the urinary bladder, vagina, and bile ducts.
into three types: embryonal (ERMS), alveolar (ARMS), In these organs, the patients may present with hematu-
and pleomorphic (PRMS). RMSs are rapidly growing ria, obstructive uropathy, or jaundice.
tumors and present with symptoms secondary to mass ARMSs occur more frequently in the extremities or
effect. paranasal sinuses of adolescents and young adults and
ERMSs are the most common RMS in children. They may have similar manifestation as ERMSs. On occasion,
occur predominantly in the head and neck (50%), widespread dissemination of the tumor is noted at the
followed by the genitourinary tract (30%) and the time of diagnosis.
110 FINE NEEDLE ASPIRATION CYTOLOGY

The rare PRMSs arise from the lower extremities of


adults. RHABDOMYOSARCOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Embryonal rhabdomyosarcoma (ERMS): admixture of round cells
PATHOLOGIC FEATURES and rhabdomyoblasts (spider cells, tennis-racket cells, or strap
cells) with demonstrable striations; elongated cells are common
in the spindle cell variant
Histologically, rhabdomyoblasts range from round to ៉ Alveolar rhabdomyosarcoma (ARMS): relatively monomorphic
stellate and are usually identified in either paucicellular population of round cells with eccentrically located nuclei;
myxoid foci or densely populated aggregates of primi- cytoplasmic vacuoles may be present
tive cells. Botryoid ERMSs are characterized by the ៉ Pleomorphic rhabdomyosarcoma (PRMS): bizarre tumor cells with
presence of a cambium layer which is a cellular band demonstrable striations in some cells; mitotic figures are easy
to find
of neoplastic cells beneath the mucosal surface.
ARMSs are composed of relatively small round cells
Immunohistochemistry
devoid of prominent nucleoli, aggregating as nests sepa-
៉ ERMS: positive for vimentin, MyoD1, myogenin, desmin, actin,
rated by thin vascularized septae. The cellular nests may
and myoglobin; rare expression of cytokeratin, S-100, Leu7,
undergo degenerative changes and fixation artifact neuron-specific enolase, neurofilament, CD99, and B-cell markers
resulting in the formation of a central ‘cavity’ filled with (CD20)
detached tumor cells. The solid variants of ARMSs have ៉ ARMS: same immunophenotype; may be negative for myogenin
a nested pattern of tumor cells separated by fibrocollag- ៉ PRMS: same immunophenotype
enous septa. PRMSs are high-grade pleomorphic sarco-
mas composed of bizarre tumor cells showing morphologic Genetics
or immunohistochemical evidence of skeletal muscle ៉ ERMS: complex karyotype; allelic loss in chromosomal region
differentiation. 11p15
FNAB of RMSs is usually markedly cellular, consist- ៉ ARMS: about 90% of cases, t(2;13)(q35;q14); less common,
ing of tissue fragments and dissociated cells. ERMSs t(1;13)(p36;q14); PAX3/FKHR and PAX7/FKHR fusion transcripts
(Fig. 4-19) tend to have an admixture of primitive- ៉ PRMS: complex karyotype
appearing round cells and variable forms of rhabdomyo-
blasts including ‘spider’ cells or ‘strap’/‘tadpole’ cells Differential Diagnosis and Pitfalls
with occasional demonstrable striations. The latter cells ៉ ERMS: carcinoma, melanoma, ARMS, and epithelioid sarcoma
៉ Spindle ERMS: spindle cell tumors including fibrosarcoma,
are also seen in PRMSs and, to a lesser extent, in
synovial sarcoma, leiomyosarcoma, malignant peripheral nerve
ARMSs. A myxoid and collagenous stroma may be
sheath tumor, and melanoma
exceptionally abundant in any of the RMS subtypes. ៉ ARMS: alveolar soft part sarcoma, renal cell carcinoma,
Uniform round cells with pink cytoplasm and eccentri- epithelioid sarcoma, and melanoma
cally located nuclei are the typical findings in ARMSs ៉ PRMS: pleomorphic sarcomas, and carcinomas
(Fig. 4-20). The presence of such cells helps to distin-
guish ARMSs from other differential diagnostic consid-
erations of small round cell tumors. Spindle ERMSs
consist principally of elongated slender cells, some with

A B

FIGURE 4-19
Rhabdomyosarcoma, embryonal type: FNAB. Aspirates are usually cellular. The tumor cells are predominantly round cells with a high N/C ratio.
Tumor cells may have eccentrically placed nuclei and a scant eosinophilic cytoplasm. Papanicolaou stain, high power.
CHAPTER 4 Soft Tissue 111

A B

FIGURE 4-20
Rhabdomyosarcoma, alveolar type: FNAB. The tumor cells are round and uniform in size, indistinguishable from other entities under the ‘small round
blue cell tumor’ category. Cells with more abundant cytoplasm (B, arrow) are rare, but, when present, they suggest a skeletal muscle differentia-
tion. Papanicolaou stain, high power.

demonstrable striations. The tumor cells of PRMSs are Ewing/peripheral nerve sheath tumor (PNET), neuro-
expectedly large with great variation in size and blastoma, and desmoplastic small round cell tumor.
shape. Pulmonary small cell carcinoma, poorly differentiated
squamous cell carcinoma, and Merkel cell carcinoma
are also included in the cytologic differentials; however,
these neoplasms are only encountered in adults. When
ANCILLARY STUDIES an alveolar pattern is prominent, alveolar soft part
sarcoma may be difficult to discriminate from ARMSs
All variants of RMSs share a similar immunopheno- in small tissue biopsies; desmin may be positive in both
type. MyoD1 and myogenin (nuclear stain) are highly tumors, further compounding the dilemma. Additional
specific for skeletal muscle differentiation, and about tumors with a prominent alveolar pattern such as
90% of RMSs express these antibodies. Myoglobin is renal cell carcinoma, epithelioid sarcoma, and mela-
not commonly used due to high background staining noma may resemble ARMSs.
and may not stain less differentiated RMSs. RMSs Other spindle cell tumors such as synovial sarcoma,
also stain with desmin and actins (SMA and MSA). rhabdomyoma, and even reactive conditions (postopera-
Ultrastructural studies may reveal diagnostic features tive spindle cell nodules) should be differentiated from
including thick and thin fi laments and rudimentary ERMSs in the urinary bladder and scrotal area. Poorly
sarcomeres. differentiated variants of other sarcomas and carcino-
Of the three histologic types, ARMSs have the most mas are indistinguishable from PRMSs except for the
consistent cytogenetic abnormality: t(2;13)(q35;q14) and presence of scattered rhabdomyoblasts.
t(1;13)(p36;q14). Usually, these translocations are part In all three types of RMSs, demonstration of rhabdo-
of a more complex karyotype. The fusion transcripts myoblasts is supportive of the diagnosis. Antibodies
(PAX3/FKHR and PAX7/FKHR) act as transcriptional against skeletal muscle differentiation (MyoD1 and
activators and can be demonstrated by RT-PCR. It has myogenin) are quite specific and often essential for
been reported that allelic loss in chromosomal region diagnosis. Triton tumors have true rhabdomyoblastic
11p15 is common among ERMSs. The karyotype of both differentiation and have to be taken into consideration
ERMSs and PRMSs is often complex. in the evaluation. Prudence should be exercised not to
mistake entrapped and regenerating non-neoplastic
skeletal muscle fibers as rhabdomyoblasts. Other anti-
bodies including epithelial, smooth muscle, hematolym-
DIFFERENTIAL DIAGNOSIS AND PITFALLS phoid, and other markers directed to the entities in the
differential diagnosis may be necessary. When adequate
tissue is available, samples for cytogenetic, molecular,
The differential diagnosis of ARMSs encompasses and ultrastructural examinations should be submitted
other small round cell tumors including lymphoma, to assist in problematic cases.
112 FINE NEEDLE ASPIRATION CYTOLOGY

PATHOLOGIC FEATURES
SMOOTH MUSCLE TUMORS

Histologically, LMSs are composed of spindle-shaped


LEIOMYOSARCOMA tumor cells forming short fascicles that intersect at
right angles. The tumor cells have rounded ends, and,
occasionally, epithelioid variants may be encountered.
CLINICAL FEATURES Mitotic activity may be brisk. The stroma is usually
fibrocollagenous. The histologic criteria applied for the
diagnosis of LMSs may differ according to the site of
The common sites of leiomyosarcomas (LMSs) are origin.
the uterus, deep large vessels (pulmonary vessels, Uterine LMSs are not sampled by fine needle aspira-
inferior vena cava, and vessels of the extremities), and tion. Primary LMSs of the large vessels and, less com-
dermis. The latter tend to have a more indolent behav- monly, dermal LMSs may be aspirated. Most aspirated
ior. Symptoms secondary to obstruction of large vessels, LMSs are from tumors metastatic to the lungs or other
such as Budd–Chiari syndrome, superior vena cava visceral organs. The aspirates of LMSs are usually
syndrome, renal hypertension, or leg edema, may be cellular and consist of cohesive spindle-shaped tumor
the initial manifestation of the deep-seated LMSs. cells arranged in fascicles (Figs 4-21 & 4-22). The
cytoplasm may have fine granules. The spindle cells
typically have cigar-shaped (blunt-ended) nuclei. Less
differentiated forms of LMSs may consist of atypical
poorly cohesive polygonal cells. Bizarre pleomorphic
tumor cells may be present. Epithelioid LMSs consist
predominantly of uniform or markedly pleomorphic
LEIOMYOSARCOMA – DISEASE FACT SHEET
tumor cells with hyperchromasia, pleomorphism, mitotic
figures, and prominent nucleoli. Necrosis may be
Incidence and Location
present. The stroma in all types of LMSs can be fibro-
៉ Most common in the uterus
collagenous or myxoid.
៉ In the soft tissue, leiomyosarcomas (LMSs) account for 10–15% of
soft tissue tumors; most common in the retroperitoneum

Gender, Race, and Age Distribution


ANCILLARY STUDIES
៉ Soft tissue LMSs: female predilection for retroperitoneal and
inferior vena cava LMS; other sites including large vessel or
dermal LMS, no sex predilection; most common in middle-aged LMSs are invariably positive for SMA (Fig. 4-22B) and
but also affects children
MSA, and less frequently for desmin, particularly in
៉ Uterine LMSs: more common in black women; more common in
middle-aged or elderly patients (median age, 50–55 years)
poorly differentiated types. Caldesmon and calponin
are two other antibodies expressed by smooth muscle-
derived neoplasms. LMSs may be positive for CK and

A
B

FIGURE 4-21
Leiomyosarcoma: FNAB. The aspirated materials are usually cellular. The tumor cells are typically spindled with cigar-shaped nuclei. A fascicular
arrangement may be suggested by tight cohesion of cells (B). A, Diff-Quik stain, high power. B, Papanicolaou stain, high power.
CHAPTER 4 Soft Tissue 113

A B

FIGURE 4-22
Leiomyosarcoma: needle core biopsy. A, The tumor cells form short fascicles characteristically intersecting at right angles. H&E stain, low-power.
B, Smooth muscle actin is invariably expressed. Smooth muscle actin, low power.

There is no specific karyotypic aberration, and chro-


LEIOMYOSARCOMA – PATHOLOGIC FEATURES mosomal analysis usually shows multiple and complex
abnormalities.
Cytopathologic Findings
៉ Fairly cellular aspirates composed of predominantly spindle
cells; cigar-shaped nuclei; occasionally, epithelioid cells and
pleomorphic cells predominate
DIFFERENTIAL DIAGNOSIS AND PITFALLS
៉ Spindle tumor cells have elongated cytoplasm
៉ Stroma is myxocollagenous
Other spindle cell tumors such as dermatofibrosarcoma
protuberans (DFSP), fibrosarcoma, malignant periph-
Immunohistochemistry
eral nerve sheath tumor, synovial sarcoma (SS), angio-
៉ Positive for smooth muscle actin, caldesmon, desmin, and
sarcoma, gastrointestinal stromal tumors (GIST), and
calponin
៉ May be positive for cytokeratin and epithelial membrane
mesenteric fibromatosis enter the differential diagno-
antigen sis. Tumors originating from a vessel wall suggest a
៉ Negative for CD34, S-100, CD117, and β-catenin smooth muscle origin. Expression of muscle markers
(SMA, MSA, caldesmon, calponin, and desmin) sup-
Electron Microscopy ports the diagnosis of LMSs. The immunohistochemi-
៉ Actin filaments with focal densities, cell junctions, pinocytotic cal panel should include S-100 protein, CD34, CD117,
vesicles, and basement membrane and β-catenin to assist in the differential diagnosis.
Rarely, cytogenetic and ultrastructural studies are
Genetics resorted to in the confirmation of the diagnosis.
៉ Complex karyotype Although there is no specific cytogenetic abnormality
៉ RB1 gene abnormality may be detected of LMSs, entities with specific aberrations, including
SS and DFSP, can be detected by genetic analysis.
Differential Diagnosis and Pitfalls
៉ Spindle cell sarcomas including dermatofibrosarcoma
protuberans, fibrosarcoma, synovial sarcoma, malignant
peripheral nerve sheath tumor, and angiosarcoma VASCULAR TUMORS
៉ Gastrointestinal stromal tumors (GIST), mesenteric fibromatosis
៉ Presence of pleomorphic cells broadens the differential
diagnosis
ANGIOSARCOMA

EMA in up to 30% of cases. LMSs do not express S-100 CLINICAL FEATURES


and CD117, and rarely CD34. Whereas uterine LMSs
are positive for estrogen and progesterone receptors,
LMSs from other organs are negative. Ultrastructur- Angiosarcomas (ASs) may arise sporadically, associated
ally, presence of actin fi laments with focal densities in with syndromes (neurofibromatosis, Klippel–Trenaunay
the cytoplasm, suggestive of smooth muscle differentia- syndrome, and Maffucci syndrome), or related to expo-
tion, may be demonstrated. sure to chemicals including therapeutic and diagnostic
114 FINE NEEDLE ASPIRATION CYTOLOGY

ANGIOSARCOMA – DISEASE FACT SHEET ANGIOSARCOMA – PATHOLOGIC FEATURES

Incidence Cytopathologic Findings


៉ Rare tumor ៉ Variable cellularity; predominantly spindle cells, rarely
epithelioid (polygonal) and plasmacytoid cells; intracytoplasmic
Location lumen containing red blood cells may be seen
៉ Sporadic angiosarcoma: deep soft tissue of extremities, head and ៉ Hemorrhagic background
neck, trunk, and visceral organs
៉ Postradiation angiosarcoma: chest wall and breast tissue, and any Immunohistochemistry
tissue in the field of exposure ៉ Positive for vimentin, CD31, CD34, and factor VIII-related
antigen
Gender and Age Distribution ៉ 50% are positive for cytokeratin and epithelial membrane
៉ Sporadic angiosarcoma: more common in elderly patients, without antigen
sex predilection
៉ Postradiation angiosarcoma: middle-aged to elderly women Electron Microscopy
៉ Weibel-Palade bodies are definitive for angiosarcomas but rarely
demonstrated

agents (radiation, Thorotrast, foreign body implants, Genetics


arsenic-containing insecticides, and vinyl chloride). ៉ Complex karyotype
The common locations for sporadic AS are the scalp ៉ No specific genetic defect
and deep soft tissue. Postradiation ASs, most commonly
seen in patients treated for breast carcinoma, often Differential Diagnosis and Pitfalls
arise in the chest wall and breast tissue. ASs may ៉ Spindle angiosarcomas: other vascular tumors including
present as cutaneous plaques, subcutaneous nodules, or hemangioma and Kaposi sarcoma, and other spindle cell
soft tissue mass lesions. Non-specific symptoms related sarcomas including fibrosarcoma, leiomyosarcoma, and malignant
to visceral organ involvement, such as gastrointestinal peripheral nerve sheath tumor
៉ Epithelioid angiosarcomas: epithelioid hemangioendothelioma,
bleeding or obstruction, may precede the discovery of
carcinoma, and epithelioid mesenchymal tumors (epithelioid
visceral AS. Epithelioid and poorly differentiated ASs sarcoma, synovial sarcoma, malignant peripheral nerve sheath
are aggressive tumors and may rapidly metastasize. tumor)

PATHOLOGIC FEATURES
helpful clue to the correct diagnosis is the presence of
intracytoplasmic lumina containing red blood cells.
Histologically, the spectrum of ASs varies from well- Aspirates from ASs are often bloody with a variable
differentiated innocuous vessel-forming lesion to number of tumor cells (Fig. 4-23). The tumor cells may
high-grade malignant tumor that mimics carcinoma. A be spindle-shaped, polygonal (epithelioid), or plasma-

B
A

FIGURE 4-23
Angiosarcoma: FNAB. The aspirates are variably cellular, often in a hemorrhagic background. High-grade angiosarcomas usually exhibit an epithelioid
cytomorphology. Intracytoplasmic lumina are not regularly demonstrated. Note prominent nucleoli in (B). A, Papanicolaou stain, high power.
B, Diff-Quik stain, high power.
CHAPTER 4 Soft Tissue 115

cytoid cells. Epithelioid AS may mimic carcinoma or


other mesenchymal lesions with epithelioid features. GASTROINTESTINAL STROMAL TUMOR – DISEASE FACT SHEET
Sheets of pleomorphic tumor cells are seen in poorly dif-
ferentiated ASs. Mitoses may be frequent, and intracyto- Incidence
plasmic hemosiderin pigment may be seen. Identification ៉ 7–14.5 per million population per year
of intracytoplasmic lumina with red blood cells is an
important feature of ASs; however, this feature may be Location
more difficult to appreciate in cytologic preparations. ៉ Stomach – 51%
៉ Small intestine – 36%
៉ Colon and rectum – 12%
៉ Esophagus – 1%
ANCILLARY STUDIES ៉ Mesentery, omentum, and extra-abdominal sites – rare

Gender, Race, and Age Distribution


Most ASs express commonly used endothelial markers ៉ Slight male predilection
(CD31, CD34, and factor VIII-related antigen). Some ៉ Black women are affected six times more frequently than white
ASs, particularly the epithelioid variants, are also women
immunoreactive to CK and EMA. EM can demonstrate ៉ Mean age – 63 years
Weibel-Palade bodies, which are regarded as specific
organelles for endothelial cells; however, these are
rarely identified in proven cases of ASs.
The karyotype of ASs is usually complex and without mass effect. However, in some instances, the fi rst pre-
specific genetic abnormalities. sentation is a metastasis, usually within the peritoneal
cavity, liver, or mesentery. The diagnosis of GIST is
usually established by endoscopic ultrasound-guided or
percutaneous FNAB.
DIFFERENTIAL DIAGNOSIS AND PITFALLS
PATHOLOGIC FEATURES
The differential diagnosis depends on the degree of
differentiation of an AS. Well-differentiated ASs can be
mistaken for hemangioma or Kaposi sarcoma. Less dif- The histology of GISTs ranges from spindle cells in
ferentiated ASs may present with solid sheets of spindle fascicles to nested epithelioid cells to patternless clus-
cells that may be indistinguishable from other spindle tering of pleomorphic cells.
cell sarcomas such as fibrosarcoma, malignant fibrous
histiocytoma, and leiomyosarcoma. Epithelioid variants
of AS can resemble any epithelial/epithelioid neoplasms. GASTROINTESTINAL STROMAL TUMOR – PATHOLOGIC FEATURES
CK and EMA expression of ASs can further add to the
difficulty in the differential diagnosis. In general, Cytopathologic Findings
unusually bloody aspirates should suggest possibility of ៉ Cellular aspirates; admixture of clustered or singly scattered
a vascular lesion. Cytologically, the presence of cyto- tumor cells; tumor cells form three-dimensional clusters
plasmic lumina with red blood cells, and intracyto- ៉ Myxocollagenous stroma
plasmic hemosiderin are also suggestive of endothelial
differentiation. In the right clinical background and cell Immunohistochemistry
morphology, expression of CD31, CD34, and/or factor ៉ Positive for CD117 (almost all cases), CD34 (60–70%), smooth
VIII-related antigen confirms the diagnosis of ASs. muscle actin (30–40%), and, rarely, desmin and S-100 protein
(<5%)
៉ CD117 negative cases are positive for PDGFRA mutation

Genetics
GASTROINTESTINAL STROMAL TUMOR ៉ Common losses in chromosomes 14 and 22
៉ Mutation in exons 11, 9, or 13 of c-kit gene
៉ Mutation of PDGFR in c-kit-negative GISTs
CLINICAL FEATURES
Differential Diagnosis and Pitfalls
៉ Spindle GISTs: smooth muscle tumors, infl ammatory fibroid
Gastrointestinal stromal tumors (GISTs) arise from the polyp, infl ammatory myofibroblastic tumor, fibromatosis, solitary
wall of the GI tract and occasionally in adjacent tissues fibrous tumor, dedifferentiated liposarcoma, malignant
such as the omentum and mesentery. The stomach is peripheral nerve sheath tumor, spindle cell carcinoma,
most frequently affected, followed in order of frequency melanoma, and reactive nodular fibrous pseudotumor
៉ Epithelioid GISTs: carcinoma and melanoma
by the small intestine, colon, and esophagus. A common
៉ Pleomorphic GISTs: malignant fibrous histiocytoma, carcinoma,
presentation is mucosal ulceration which may lead to and pleomorphic sarcomas
hemorrhage, intestinal obstruction or perforation, and
other non-specific abdominal symptoms secondary to
116 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURES 4-24
Gastrointestinal stromal tumor: FNAB. The aspirates are usually cellular. The component cells may have spindle (A), oval/round (B), epithelioid, or
pleomorphic cytology, and usually with abundant myxocollagenous stroma. A, Diff-Quik stain, low power. B, Diff-Quik stain, high power.

A B

FIGURE 4-25
Gastrointestinal stromal tumor: cell block. A, The spindle tumor cells are arranged in short fascicles with moderate fibrocollagenous stroma. H&E
stain, medium power. B, The tumor cells are strongly positive for CD117. Note the adjacent gastric epithelium. CD117, medium power.

FNAB smears are usually cellular (Fig. 4-24). Cell ANCILLARY STUDIES
block preparation may exhibit typical fascicular arrange-
ment of cells (Fig. 4-25). The tumor cells appear as
clusters, often in three-dimensional forms or palisades Approximately 95% of GISTs express c-kit (CD117) by
admixed with single cells. The individually scattered immunohistochemistry (Fig. 4-25B). The c-kit negative
cells are often devoid of their cytoplasm. Where intact GISTs have been shown to be immunoreactive for
cells are demonstrated, the cytoplasmic processes are PDGFRA (platet-derived growth factor receptor-α). Up
often delicate. Generally, the nuclei are relatively to 90% of these neoplasms also react with CD34, which
uniform, ovoid to spindled, and often wavy with blunt appears to be influenced by the site of the tumor.
or tapered ends. Nuclear grooves and intranuclear inclu- Esophageal and rectal GISTs are known to have a
sions have been reported, particularly in epithelioid higher incidence of CD34 expression compared with
variants. A combination of spindle and epithelioid gastric and small intestine GISTs. Less than one-third
lesional cells may be encountered. The background of GISTs are SMA-positive and much less frequently
usually consists of myxocollagenous stroma. (5%) desmin-positive. GISTs often express caldesmon.
CHAPTER 4 Soft Tissue 117

The genetics of GISTs have been extensively studied.


There is a gain-of-function mutation of c-kit, a growth MYXOMA – DISEASE FACT SHEET
factor transmembrane receptor, leading to activation of
proteins in the signal transduction pathway and result- Incidence
ing in cell proliferation. C-kit mutations often involve ៉ Intramuscular myxoma (IM): relatively common tumor
exons 11 and 9. Some of the c-kit-negative GISTs have ៉ Juxta-articular myxoma (JM): rare
been shown to have mutation of PDGFRA, another ៉ Cutaneous myxoma (CM) or superficial angiomyxoma: rare;
tyrosine kinase receptor. Common cytogenetic abnor- association with Carney syndrome
malities include loss of chromosomes 14, 15, and 22.
Location
៉ IM: thigh, buttocks, shoulder, and trunk
៉ JM: near knee, shoulder, elbow, and ankle joints
៉ CM: trunk, thigh, and head and neck
DIFFERENTIAL DIAGNOSIS AND PITFALLS
Gender, Race, and Age Distribution
There is a long list of entities that cytologically resem- ៉ IM: female predilection; no race predilection; middle-aged to
ble GISTs. These include true smooth muscle tumors elderly patients
of the GI tract, inflammatory myofibroblastic tumor, ៉ JM: male predilection; no race predilection; middle-aged patients
៉ CM: no sex predilection; no race predilection; young and middle-
inflammatory fibroid polyp, fibromatosis, solitary
aged patients
fibrous tumor, dedifferentiated liposarcoma, malignant
peripheral nerve sheath tumor, carcinoma, and meta-
static melanoma. Pleomorphic GISTs are indistinguish-
able from other pleomorphic tumors of both epithelial the knee joint, but can occur in the shoulder or elbow.
and mesenchymal origin. The importance of accurate CMs may be multiple and are distributed in the trunk,
diagnosis lies in the good response of GISTs to imatinib head and neck, and extremities. When CMs are multi-
mesylate (Gleevae), a selective inhibitor of tyrosine ple, Carney complex (familial myxoma syndrome)
kinases including c-kit and PDGFRA. should be suspected.
Melanoma may express CD117, so other antibodies
(S-100 protein, HMB-45, and Melan A) should be
included in the laboratory work-up if melanoma is sus- PATHOLOGIC FEATURES
pected. Although some of the neoplasms listed above are
CD34-positive, none consistently express CD117. Car-
cinomas with small cell component may be positive for Histologically, the tumor cells are usually scant with
CD117, but CK expression is rarely observed in GISTs. abundant myxoid stroma. The tumor vascularity is
More problematic cases are the CD117-negative GIST. often minimal to non-existent in IMs and JMs, but CMs
PDGFRA may be of help in some of these tumors. Muta- may have focally prominent capillaries.
tional analysis in most of these cases will show c-kit or
PDGFRA mutations. In the right histomorphology and
site of origin (most c-kit-negative GISTs arise from the MYXOMA – PATHOLOGIC FEATURES
peritoneum and omentum), these tumors should be con-
sidered as GISTs. A recently described lesion, reactive Cytopathologic Findings
nodular fibrous pseudotumor, is CD117-positive and ៉ Low cellularity and abundant myxoid stroma; tumor cells are
may be difficult to discriminate from GISTs without the spindle-shaped with a low N/C ratio; cytoplasm is bipolar with
benefit of clinical history and ancillary studies. elongated processes; absence of vascular channels

Immunohistochemistry
៉ Positive for vimentin
៉ May be positive for CD34, desmin, and smooth muscle actin
TUMORS OF UNCERTAIN HISTOGENESIS ៉ Negative for S-100 protein

Genetics
៉ Intramuscular myxoma: mutation in GNAS1 gene
MYXOMA ៉ Juxta-articular myxoma: no specific genetic abnormality; GNAS1
gene mutation is absent
៉ Cutaneous myxoma: association with Carney complex; mutation
CLINICAL FEATURES in 17q22–24 locus (PRKAR1A gene)

Differential Diagnosis and Pitfalls


Myxomas are subclassified into three clinicopathologic
៉ Scar, and benign and malignant tumors with myxoid stroma
types: intramuscular myxomas (IMs), juxta-articular
(benign fibrous histiocytoma, fibromatosis, dermatofibrosarcoma
myxomas ( JMs), and cutaneous myxomas (CMs) or
protuberans, myxoid liposarcoma, low-grade fibromyxoid
superficial angiomyxomas. IMs often arise in the thigh sarcoma, and myxoid malignant fibrous histiocytoma)
muscles as a painless, soft and fluctuant mass. JMs are
usually located close to a large joint, most commonly
118 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 4-26
Myxoma: touch preparation. The tumor is often hypocellular and composed mainly of thin myxoid stroma. The bland-appearing tumor cells are
elongated with long cytoplasmic processes. Vascular channels are usually not identified. Diff-Quik stain, high power.

Aspirates from these tumors yield clear viscous fluid. myxoid stroma invariably contain markedly atypical
The smears are generally paucicellular with profuse cells and, in most occasions, moderate amount of
myxoid material in the background (Fig. 4-26). The vascular channels.
tumor cells are elongated and bland with long cytoplas- Immunohistochemistry is of limited use in myxoma
mic processes. The N/C ratio is low and nucleoli are owing to the absence of specific antibody associated
not conspicuous. Aspirated capillary channels are quite with the tumor; however, absence of expression of anti-
rare. Other entrapped or surrounding mesenchymal ele- bodies known to be positive in other tumors (e.g. S-100
ments such as skeletal muscle, fat, or fibrocollagenous protein expression of myxoid liposarcoma) assists in the
tissue may be present in the aspirates. differential diagnosis. Mutational analysis directed to
GNAS1 (for IMs) and PRKAR1A (for CMs) genes may
help establish the diagnosis.

ANCILLARY STUDIES

EPITHELIOID SARCOMA
The lesional cells of myxoma may be positive for CD34,
desmin, and SMA. S-100 protein is not expressed.
Point mutation of the GNAS1 gene appears to be a CLINICAL FEATURES
consistent abnormality of IMs but not JMs and CMs.
CMs associated with Carney complex may have muta-
tion of the suppressor gene – protein kinase A type I-α Epithelioid sarcomas commonly affect young and
(PRKAR1A) gene. middle-aged male patients. The distal types often

DIFFERENTIAL DIAGNOSIS AND PITFALLS


EPITHELIOID SARCOMA – DISEASE FACT SHEET

Several sarcomas may have spindle cells and myxoid Incidence and Location
stroma, including malignant fibrous histiocytoma, low- ៉ Rare tumors
grade fibromyxoid sarcoma, and liposarcoma, particu- ៉ Associated with tendons and aponeurosis of the distal extremities
larly the myxoid variant. Other benign and low-grade (distal type) and deep soft tissue of the proximal extremities
sarcomas with myxoid component in the differential (proximal type)
diagnosis are fibromatosis, benign fibrous histiocy-
Gender, Race, and Age Distribution
toma, and dermatofibrosarcoma protuberans.
៉ Male predilection
The presence of bland-appearing spindle cells with
៉ No race predilection
elongated cytoplasmic processes and absent or minimal
៉ Young and middle aged adults for distal type; older age for
vascular channels in the aspirate support the diagnosis proximal type
of myxoma and exclude most of the entities listed in the
differential diagnosis. The malignant neoplasms with
CHAPTER 4 Soft Tissue 119

present as slow-growing subcutaneous nodules in the


fingers, palm, and wrist. The proximal types often EPITHELIOID SARCOMA – PATHOLOGIC FEATURES
arise from the buttocks or thigh as deep-seated mass
lesions. The mode of spread is through regional lymph Cytopathologic Findings
nodes. Frequently, the tumor tracks along the tendon ៉ Spindle and polygonal cells with moderately pleomorphic nuclei
and aponeurosis before reaching the lymph nodes. and variable nucleolar prominence; necrosis may be present;
Distant metastases are usually to the lungs. palisading of aspirated tumor cells and necrosis may resemble
granulomas
៉ Proximal-type epithelioid sarcomas appear more undifferentiated
with a high N/C ratio and round or polygonal tumor cells
PATHOLOGIC FEATURES
Immunohistochemistry
៉ Positive for cytokeratin, epithelial membrane antigen, and V-
Histologically, the tumor cells form nests separated cadherin; 50% are positive for CD34; variable expression of
by fibrocollagenous stroma. The most characteristic desmin, muscle-specific actin (HHF-35), and smooth muscle
feature of epithelioid sarcomas is the presence of central actin
៉ Negative for E-cadherin and S-100
necrosis surrounded by palisading tumor cells. The
tumor cells are spindle or polygonal with eosinophilic
Genetics
cytoplasm, moderately pleomorphic nuclei, and small
៉ No consistent karyotypic abnormality
nucleoli.
The aspirated material is usually highly cellular (Fig.
Differential Diagnosis and Pitfalls
4-27). Due to the fibrocollagenous elements of some
៉ Granuloma annulare, carcinoma, melanoma, rhabdomyosarcoma,
tumors, occasional paucicellular aspirates can be
alveolar soft part sarcoma, synovial sarcoma, malignant
expected. The tumor cells are usually round or poly- peripheral nerve sheath tumor, and epithelioid angiosarcoma
gonal. Infrequently, spindle-shaped cells may predomi-
nate. The neoplastic cells may also assume a rhabdoid
or plasmacytoid appearance. Moderate to severe atypia,
abundant mitotic figures, necrosis, and prominent
nucleoli are common in the proximal types of epitheli-
oid sarcomas. Loosely cohesive clusters of polygonal and
epithelioid cells and a background of necrosis may
recapitulate a granulomatous histology. are positive for CD34. Epithelioid sarcomas may
stain with muscle markers including desmin, MSA
(HHF-35), and SMA. The tumor is consistently nega-
tive for S-100 protein, but may occasionally express
ANCILLARY STUDIES HMB-45.
The ultrastructural features of tumor cells range
from epithelial to fibroblast-like mesenchymal cells.
Aside from vimentin, epithelioid sarcomas also con- There is no specific chromosomal or genetic abnormal-
sistently express CK and EMA. Approximately 50% ity identified so far.

A
B

FIGURE 4-27
Epithelioid sarcoma: FNAB. FNAB materials are often cellular. The tumor cells are round or polygonal and indistinguishable from other epithelial
malignancies. Necrosis may be apparent on the aspirated material (A). A, Papanicolaou stain, medium power. B, Diff-Quik stain, high power.
120 FINE NEEDLE ASPIRATION CYTOLOGY

DIFFERENTIAL DIAGNOSIS AND PITFALLS


ALVEOLAR SOFT PART SARCOMA – DISEASE FACT SHEET

Epithelioid sarcomas can mimic granuloma annulare Incidence and Location


(GA), carcinoma, and other epithelioid-appearing ៉ Rare tumor: 0.5–0.9% of soft tissue tumors
sarcomas including alveolar soft part sarcoma (ASPS), ៉ Lower extremities (41% in thigh or buttocks); orbit and tongue in
children
synovial sarcoma (SS), and epithelioid angiosarcoma
(AS). The frequent location of the tumor on the hand
Gender, Race, and Age Distribution
and the presence of prominent central necrosis may be
៉ Female predilection before age of 30 years; slight male
interpreted as representing GA. Attention to the cyto-
predilection after 30 years
logic atypia in the surrounding cells should prompt a ៉ No race predilection
consideration of epithelioid sarcoma. The typical
location of the tumor, pure epithelioid lesional cells,
and expression of epithelial markers substantiate the
diagnosis.
Immunohistochemical stains are usually necessary
mainly to rule out other entities in the differential diag-
nosis: expression of CD31 and factor VIII-related antigen
for AS, CD68 for GA. Other ancillary studies are PATHOLOGIC FEATURES
intended to exclude other entities with specific ultra-
structural or genetic features, such as rhomboid crystals The characteristic histology of ASPSs is that of a nested
and t(X;17) of ASPS, Weibel-Palade bodies of AS, or pattern of tumor cells forming a central cavity partially
t(X;18) of SS. fi lled with dissociated tumor cells and thin septae
separating each nest. The tumor cells are polygonal
with ample eosinophilic cytoplasm and eccentrically
ALVEOLAR SOFT PART SARCOMA placed nuclei.
The aspirated material is cellular and composed of
tight aggregates or singly dispersed tumor cells (Fig.
CLINICAL FEATURES 4-28). The lesional cells are composed of relatively large
round cells with a low N/C ratio. Binucleation and
multinucleation are frequently seen with the nuclei
Alveolar soft part sarcomas (ASPSs) are rare neoplasms exhibiting conspicuous nucleoli. The nuclei are round
that present as slow-growing deep-seated tumors in the with smooth outline, and of variable size. The abundant
lower extremities, particularly the thigh, usually in cytoplasm is finely granular. Due to the fragility of the
young males. Other reported sites for ASPSs are the cytoplasm, scattered naked nuclei are often prominent
orbit and tongue in children. ASPSs may also present in the background. Branching capillaries may be associ-
as metastatic lesions, particularly in the lungs, bone, ated with clusters of tumor cells. Myxocollagenous
and brain. stroma is rarely prominent.

A B

FIGURE 4-28
Alveolar soft part sarcoma: touch preparation. The tumor is usually cellular and composed of round or polygonal cells with abundant finely granular
cytoplasm, imparting a deceptively low N/C ratio. The nuclei, some devoid of cytoplasm, are round and smooth-outlined. Diff-Quik stain, high
power.
CHAPTER 4 Soft Tissue 121

tumors. It is prudent to submit a portion of the tumor,


ALVEOLAR SOFT PART SARCOMA – PATHOLOGIC FEATURES when adequate material is available, for EM, cyto-
genetic, and/or molecular studies.
Cytopathologic Findings
៉ Cellular aspirates; relatively large round cells with eosinophilic
and granular cytoplasm; nuclei are round and nucleoli are small;
mitotic figures are rare SYNOVIAL SARCOMA
៉ Stroma is usually scant

Immunohistochemistry CLINICAL FEATURES


៉ No specific immunophenotype
៉ May be positive for desmin and S-100
៉ Negative for synaptophysin, chromogranin, cytokeratin, and The majority of synovial sarcomas (SSs) are usually
epithelial membrane antigen slow-growing tumors which arise from soft tissue of
the extremities, often in the thigh. Less common sites
Electron Microscopy include the upper extremities, trunk, and head and
៉ Rhomboid crystals, free or membrane-bound neck region. Patients may complain of pain, numbness,
paresthesia, or some loss of function of the involved
Genetics extremity. Incompletely excised tumor may locally
៉ der(17) t(X;17) (p11.2;q25) recur. Metastasis is usually to the lungs.
៉ ASPL/TFE3 fusion product

Differential Diagnosis and Pitfalls


៉ Paraganglioma, epithelioid sarcoma, renal cell carcinoma, PATHOLOGIC FEATURES
alveolar rhabdomyosarcoma, and melanoma

Histologically, three types are currently recognized:


biphasic type, monophasic type, and poorly differenti-
ated type. The biphasic SSs consist of spindle cells and
epithelial cells often with glandular differentiation.
ANCILLARY STUDIES The monophasic SSs are composed purely of spindle
cells or plump cells with foci of cellular evidence of
epithelial differentiation manifested immunohisto-
The only antibody that ASPSs consistently express is chemically by expression of CK. Poorly differentiated
vimentin. ASPSs are variably reactive to desmin, and SSs (PDSSs) are composed of rather round or oval cells
occasionally reactive to SMA, MSA, and S-100 protein. that are indistinguishable from other small round cell
The tumor is non-reactive to MyoD1 and myogenin. tumors. Immunohistochemical stains and molecular
Epithelial and neuroendocrine markers are negative. studies are necessary to establish the diagnosis of
The characteristic intracytoplasmic crystals can be PDSSs.
appreciated as periodic acid–Schiff (PAS)-positive FNAB of SSs is usually hypercellular (Figs 4-29 &
needle-shaped crystals in tumor cells. These membrane- 4-30). Singly dispersed lesional cells and tumor
bound or free rhomboid crystals can be demonstrated tissue fragments, sometimes branched, are frequently
by EM. A non-balanced translocation, t(X;17)(p11;q25), obtained. Myxofibrous stroma accompanies aggregates
resulting in fusion of TFE3 (Xp11) with ASPL (17q25) of spindle cells or chunks of tumor. Singly dispersed
appear to be specific for ASPS. The fusion product neoplastic cells are often stripped of their cytoplasm.
(TFE3/ASPL) can be demonstrated by PCR analysis. The tumor cell nuclei have fine chromatin and
inconspicuous nucleoli. The cytoplasm is elongated
with bipolar processes. Spindle-shaped tumor cells
DIFFERENTIAL DIAGNOSIS AND PITFALLS

The distinctive alveolar pattern of ASPSs and its mod-


SYNOVIAL SARCOMA – DISEASE FACT SHEET
erate, sometimes clear cytoplasm are shared by other
tumors including alveolar rhabdomyosarcoma (ARMS),
Incidence and Location
paraganglioma, renal cell carcinoma (RCC), and granu-
៉ ∼10% of soft tissue tumors
lar cell tumor (GCT). Immunohistochemistry is often
៉ Extremities, trunk, and neck and head regions
performed to exclude other tumors in the differential
diagnosis. ARMSs are consistently positive for MyoD1 Gender, Race, and Age Distribution
and myogenin; paraganglioma, for neuroendocrine
៉ Slight male predilection
markers; RCC, for CK and vimentin; and GCT, for ៉ No race predilection
S-100 protein and CD68. On occasions, ASPSs may ៉ Young adults and children (median age, 30–35 years)
express desmin and S-100 protein, so a complete panel
of antibodies should be run to effectively exclude other
122 FINE NEEDLE ASPIRATION CYTOLOGY

predominate even in biphasic SSs. Epithelial elements


SYNOVIAL SARCOMA – PATHOLOGIC FEATURES of biphasic SSs may stand out as three-dimensional
clusters or gland-forming cuboidal or polygonal cells
Cytopathologic Findings associated with mucinous material. Squamous epithelial
៉ Biphasic SS: cellular aspirates; spindle cells predominate; scant elements are infrequent. The neoplastic cells of mono-
or absent epithelial cells; myxocollagenous stroma phasic SSs appear as ‘comma’-shaped cells and are
៉ Monophasic SS: cellular aspirates; spindle cells with comma- indistinguishable from spindle cells of biphasic SSs. The
shaped nuclei; myxocollagenous stroma tumor cells of PDSSs are round or oval, and relatively
៉ Poorly differentiated SS: cellular aspirates; oval or round cells;
small sized, with a high N/C ratio. In all three variants
spindle and epithelial cells may be present; myxocollagenous
stroma
of SSs, markedly pleomorphic cells are uncommon.

Immunohistochemistry
៉ Biphasic SS:
ANCILLARY STUDIES
៉ Epithelial cells: positive for cytokeratin (CK), epithelial
membrane antigen (EMA), vimentin, HBME1 and calretinin,
and, less frequently, CD99; negative for bcl-2
CK (Fig. 4-30B), EMA, and, less frequently, mesothelial
៉ Stromal cells: positive for vimentin and bcl-2, CD99 (60%),
S-100 protein (30%); negative for desmin and smooth muscle
markers (HBME1 and calretinin) highlight the epithe-
actin; scattered CK-positive cells lial cells of biphasic SSs, and some areas of monophasic
៉ Monophasic SS: phenotype is similar to the stromal cells of biphasic SSs and PDSSs. The stromal cells of biphasic SSs, and
SS; patchy areas are positive for CK, EMA, CD99, and calretinin the majority of lesional cells of monophasic SSs and
៉ Poorly differentiated SS: phenotype is similar to the stromal PDSSs are positive for vimentin, bcl-2, and CD99. CK-
cells of biphasic SS; negative for WT1; patchy areas are positive and EMA-positive foci of PDSSs may be minimal.
for CK, EMA, CD99, and calretinin PDSSs are negative for WT1. SSs occasionally express
S-100 protein.
Genetics All variants of SSs exhibit t(X;18)(p11.2;q11.2), a
៉ t(X;18)(p11.2;q11.2) genetic abnormality not seen in other tumors. The chi-
៉ SYT/SSX fusion transcript meric transcript (SYT/SSX) results from fusion of the
SYT gene in chromosome 18 and SSX1, SSX2, or SSX4
Differential Diagnosis and Pitfalls in the X chromosome. The translocation can be demon-
៉ Biphasic SS: malignant peripheral nerve sheath tumor (MPNST) strated by karyotyping, FISH, and RT-PCR.
with glandular component and carcinosarcoma The epithelial cells have ultrastructural characteris-
៉ Monophasic SS: neurofibroma, fibromatosis, solitary fibrous
tics of glandular cells: desmosomal junctions and short
tumor, fibrosarcoma, low-grade fibromyxoid sarcoma, MPNST,
leiomyosarcoma and other spindle cell soft tissue tumors, and
microvilli. Spindle cells have non-specific ultrastruc-
carcinoma tural features. There is no transition from epithelial to
៉ Poorly differentiated SS: small round cell tumors (Ewing sarcoma/ mesenchymal cells.
primitive neuroectodermal tumor, rhabdomyosarcoma, non-
Hodgkin lymphoma, desmoplastic small round cell tumor, and
other small round cell tumors in children; small cell carcinoma,
Merkel cell carcinoma, and non-Hodgkin lymphoma in adults)

A B

FIGURE 4-29
Synovial sarcoma: FNAB. FNAB usually yields cellular smears. The tumor cells are spindled or round (poorly differentiated type), with a variable
amount of stroma. The epithelial component of the biphasic type is infrequently recognized in aspirates. A, Diff-Quik stain, low power.
B, Diff-Quik stain, high power.
CHAPTER 4 Soft Tissue 123

A B

FIGURE 4-30
Synovial sarcoma: cell block. Spindled stromal cells and strips of epithelial cells are identified in this cell block preparation. The epithelial cells
may be morphologically indistinguishable from the stromal cells, but are readily highlighted by cytokeratin stain (B). A, H&E stain, low power.
B, Cytokeratin stain, low power.

Cytology of Soft Tissue


DIFFERENTIAL DIAGNOSIS AND PITFALLS
Abdul-Karim FW, Rader AE. Fine needle aspiration of soft-tissue lesions.
Clin Lab Med 1998;18:507–540.
The spindle cells in biphasic SSs predominate and the Bennert KW, Abdul-Karim FW. Fine needle aspiration cytology vs. needle
core biopsy of soft tissue tumors. A comparison. Acta Cytol 1994;38:
list of differential diagnoses is similar to that of mono- 381–384.
phasic SS, including benign and malignant spindle cell Das K, Hameed M, Heller D, et al. Liquid-based vs. conventional smears
mesenchymal neoplasms such as neurofibroma, fibro- in fine needle aspiration of bone and soft tissue tumors. Acta Cytol
matosis, fibrosarcoma, and glandular MPNST. 2003;47:197–201.
Gonzalez-Campora R. Fine needle aspiration cytology of soft tissue tumors.
When the epithelial component of biphasic SS is rep- Acta Cytol 2000;44:337–343.
resented in the aspirated material, other biphasic tumors Gonzalez-Campora R. Cytoarchitectural findings in the diagnosis of primary
including MPNST and carcinosarcoma may be con- soft tissue tumors. Acta Cytol 2001;45:115–146.
sidered. The main cytomorphologic feature that favors Guiter GE, Gatscha RM, Zakowski MF. ThinPrep vs. conventional smears in
SS is the uniformity of the malignant stromal cells. In fine-needle aspirations of sarcomas: a morphological and immunocyto-
chemical study. Diagn Cytopathol 1999;21:351–354.
contrast, the sarcomatous elements of MPNST and Jones C, Liu K, Hirschowitz S, et al. Concordance of histopathologic and
carcinosarcoma are more pleomorphic. cytologic grading in musculoskeletal sarcomas: can grades obtained
PDSSs fall under the category of small round cell from analysis of the fine-needle aspirates serve as the basis for thera-
tumors and the set of differential diagnoses vary with peutic decisions? Cancer 2002;96:83–91.
Kilpatrick SE, Cappellari JO, Bos GD, et al. Is fine-needle aspiration biopsy
the patient’s age. Neuroblastoma, non-Hodgkin lym- a practical alternative to open biopsy for the primary diagnosis of
phoma (NHL), Ewing sarcoma/primitive neuro- sarcoma? Experience with 140 patients. Am J Clin Pathol 2001;115:
ectodermal tumor (PNET), rhabdomyosarcoma, and 59–68.
desmoplastic small round cell tumor should be consid- Kilpatrick SE, Geisinger KR. Soft tissue sarcomas: the usefulness and limi-
ered in children. NHL, small cell carcinoma, and Merkel tations of fine-needle aspiration biopsy. Am J Clin Pathol 1998;110:
50–68.
cell carcinoma are the most frequently encountered Kilpatrick SE, Ward WG, Cappellari JO, Bos GD. Fine-needle aspiration
small round cell tumors in adults. Immunohistochemis- biopsy of soft tissue sarcomas. A cytomorphologic analysis with empha-
try, flow cytometry, cytogenetic and/or molecular sis on histologic subtyping, grading, and therapeutic significance. Am
studies are essential in arriving at a definitive J Clin Pathol 1999;112:179–188.
Kilpatrick SE, Ward WG, Chauvenet AR, Pettenati MJ. The role of fine-
diagnosis. needle aspiration biopsy in the initial diagnosis of pediatric bone and
soft tissue tumors: an institutional experience. Mod Pathol 1998;11:
923–928.
SUGGESTED READINGS Maitra A, Ashfaq R, Saboorian MH, et al. The role of fine-needle aspiration
biopsy in the primary diagnosis of mesenchymal lesions: a community
General hospital-based experience. Cancer 2000;90:178–185.
Soft Tissue Tumors Mathur S, Kapila K, Verma K. Accuracy of cytological grading of spindle-
cell sarcomas. Diagn Cytopathol 2003;29:79–83.
Enzinger FM, Weiss SW, Goldblum JR. Enzinger and Weiss’s Soft Tissue Nagira K, Yamamoto T, Akisue T, et al. Reliability of fine-needle aspiration
Tumors, 4th ed. St Louis, Mosby, 2001. biopsy in the initial diagnosis of soft-tissue lesions. Diagn Cytopathol
Fletcher CDM, Unni KK, Mertens F, eds. World Health Organization Classi- 2002;27:354–361.
fication of Tumors. Pathology and Genetics. Tumors of Soft Tissue and Palmer HE, Mukunyadzi P, Culbreth W, Thomas JR. Subgrouping and
Bone. Lyon, France: IARC Press, 2002. grading of soft-tissue sarcomas by fine-needle aspiration cytology: a
Miettinen M. Diagnostic Soft Tissue Pathology. Philadelphia, Churchill histopathologic correlation study. Diagn Cytopathol. 2001;24:307–
Livingstone, 2003. 316.
124 FINE NEEDLE ASPIRATION CYTOLOGY

Powers CN, Berardo MD, Frable WJ. Fine-needle aspiration biopsy: pitfalls Dermatofibrosarcoma Protuberans
in the diagnosis of spindle-cell lesions. Diagn Cytopathol 1994;10:
232–240. Domanski HA, Gustafson P. Cytologic features of primary, recurrent, and
Sapi Z, Antal I, Papai Z, et al. Diagnosis of soft tissue tumors by fine- metastatic dermatofibrosarcoma protuberans. Cancer 2002;96:351–
needle aspiration with combined cytopathology and ancillary tech- 361.
niques. Diagn Cytopathol 2002;26:232–242. Klijanienko J, Caillaud JM, Lagace R. Fine-needle aspiration of primary and
Wakely PE Jr, Kneisl JS. Soft tissue aspiration cytopathology. Cancer recurrent dermatofibrosarcoma protuberans. Diagn Cytopathol
2000;90:292–298. 2004;30:261–265.
Weir MM, Rosenberg AE, Bell DA. Grading of spindle cell sarcomas in fine- Klijanienko J, Caillaud JM, Lagace R. Fine-needle aspiration of primary and
needle aspiration biopsy specimens. Am J Clin Pathol 1999;112:784– recurrent benign fibrous histiocytoma: classic, aneurysmal, and myxoid
790. variants. Diagn Cytopathol 2004;31:387–391.
Mendenhall WM, Zlotecki RA, Scarborough MT. Dermatofibrosarcoma pro-
tuberans. Cancer 2004;101:2503–2508.

Lipoma and Liposarcoma Malignant Fibrous Histiocytoma

Akerman M, Rydholm A. Aspiration cytology of lipomatous tumors: a 10- Kilpatrick SE, Ward WG. Myxofibrosarcoma of soft tissues: cytomorphologic
year experience at an orthopedic oncology center. Diagn Cytopathol analysis of a series. Diagn Cytopathol 1999;20:6–9.
1987;3:295–302. Kilpatrick SE, Ward WG, Bos GD. The value of fine-needle aspiration biopsy
Bassett MD, Schuetze SM, Disteche C, et al. Deep-seated, well differenti- in the differential diagnosis of adult myxoid sarcoma. Cancer (Cancer
ated lipomatous tumors of the chest wall and extremities: the role of Cytopathol) 2000;90:167–177.
cytogenetics in classification and prognostication. Cancer 2005;103: Klijanienko J, Caillaud JM, Lagace R, Vielh P. Comparative fine-needle
409–416. aspiration and pathologic study of malignant fibrous histiocytoma:
Domanski HA, Carlen B, Jonsson K, et al. Distinct cytologic features of cytodiagnostic features of 95 tumors in 71 patients. Diagn Cytopathol
spindle cell lipoma. A cytologic-histologic study with clinical, radio- 2003;29:320–326.
logic, electron microscopic, and cytogenetic correlations. Cancer 2001; Merck C, Hagmar B. Myxofibrosarcoma: a correlative cytologic and histo-
93:381–389. logic study of 13 cases examined by fine needle aspiration cytology.
Kapila K, Ghosal N, Gill SS, Verma K. Cytomorphology of lipomatous tumors Acta Cytol 1980;24:137–144.
of soft tissue. Acta Cytol 2003;47:555–562.
Klijanienko J, Caillaud JM, Lagace R. Fine-needle aspiration in liposar-
coma: cytohistologic correlative study including well-differentiated, Neurofibroma and Schwannoma
myxoid, and pleomorphic variants. Diagn Cytopathol 2004;30:307– Dahl I, Hagmar B, Idvall I. Benign solitary neurilemoma (schwannoma).
312. A correlative cytological and histological study of 28 cases. Acta Pathol
Vicandi B, Jimenez-Heffernan J, Lopez-Ferrer P, et al. Cytologic features Microbiol Immunol Scand [A] 1984;92:91–101.
of round cell liposarcoma: a report on five patients. Cancer 2003;99: Ferretti M, Gusella PM, Mancini AM, et al. Progressive approach to the
28–32. cytologic diagnosis of retroperitoneal spindle cell tumors. Acta Cytol
Walaas L, Kindblom LG. Lipomatous tumors: a correlative cytologic and 1997;41:450–460.
histologic study of 27 tumors examined by fine needle aspiration cytol- Kong CS, Cha I. Nodular fasciitis: diagnosis by fine needle aspiration
ogy. Hum Pathol 1985;16:6–18. biopsy. Acta Cytol 2004;48:473–477.
Mooney EE, Layfield LJ, Dodd LG. Fine-needle aspiration of neural lesions.
Diagn Cytopathol 1999;20:1–5.
Resnick JM, Fanning CV, Caraway NP, et al. Percutaneous needle biopsy
diagnosis of benign neurogenic neoplasms. Diagn Cytopathol 1997;16:
Nodular Fasciitis 17–25.
Slagel DD, Powers CN, Melaragno MJ, et al. Spindle-cell lesions of the
Aydin O, Oztuna V, Polat A. Three cases of nodular fasciitis: primary diag- mediastinum: diagnosis by fine-needle aspiration biopsy. Diagn Cyto-
noses by fine needle aspiration cytology. Cytopathology 2001;12: pathol 1997;17:167–176.
346–347. Stelow EB, Lai R, Bardales RH, et al. Endoscopic ultrasound-guided fine-
Dahl I, Akerman M. Nodular fasciitis a correlative cytologic and histologic needle aspiration cytology of peripheral nerve-sheath tumors. Diagn
study of 13 cases. Acta Cytol 1981;25:215–223. Cytopathol 2004;30:172–177.
Dodd LG, Martinez S. Fine-needle aspiration cytology of pseudosarcoma-
tous lesions of soft tissue. Diagn Cytopathol 2001;24:28–35.
Kong CS, Cha I. Nodular fasciitis: diagnosis by fine needle aspiration
Granular Cell Tumor
biopsy. Acta Cytol 2004;48:473–477.
Plaza JA, Mayerson J, Wakely PE. Nodular fasciitis of the hand. A potential Husain M, Nguyen GK. Cytopathology of granular-cell tumor of the lung.
diagnostic pitfall in fine-needle aspiration cytopathology. Am J Clin Diagn Cytopathol 2000;23:294–295.
Pathol 2005;123:388–393. Liu K, Madden JF, Olatidoye BA, Dodd LG. Features of benign granular cell
Willen H, Akerman M. Fine needle aspiration of nodular fasciitis – no need tumor on fine needle aspiration. Acta Cytol 1999;43:552–557.
for surgery. Acta Orthop Scand Suppl 1995;265:54–55. Liu Z, Mira JL, Vu H. Diagnosis of malignant granular cell tumor by fine
needle aspiration cytology: Acta Cytol 2001;45:1011–1021.
Wieczorek TJ, Krane JF, Domanski HA, et al. Cytologic findings in granular
cell tumors, with emphasis on the diagnosis of malignant granular cell
tumor by fine-needle aspiration biopsy. Cancer 2001;93:398–408.
Fibromatosis
Hayry P, Reitamo JJ, Totterman S, et al. The desmoid tumor. II. Analysis Malignant Peripheral Nerve Sheath Tumor
of factors possibly contributing to the etiology and growth behavior.
Am J Clin Pathol 1982;77:674–680. Ferretti M, Gusella PM, Mancini AM, et al. Progressive approach to the
Raab SS, Silverman JF, McLeod DL, et al. Fine needle aspiration biopsy of cytologic diagnosis of retroperitoneal spindle cell tumors. Acta Cytol
fibromatoses. Acta Cytol 1993;37:323–328. 1997;41:450–460.
Reitamo JJ, Scheinin TM, Hayry P. The desmoid syndrome. New aspects in Klijanienko J, Caillaud JM, Lagace R, Vielh P. Cytohistologic correlations
the cause, pathogenesis and treatment of the desmoid tumor. Am J Surg of 24 malignant peripheral nerve sheath tumor (MPNST) in 17 patients:
1986;151:230–237. the Institut Curie experience. Diagn Cytopathol 2002;27:103–108.
Zaharopoulos P, Wong JY. Fine-needle aspiration cytology in fibromatoses. Mooney EE, Layfield LJ, Dodd LG. Fine-needle aspiration of neural lesions.
Diagn Cytopathol 1992;8:73–78. Diagn Cytopathol 1997;16:17–25.
CHAPTER 4 Soft Tissue 125

Resnick JM, Fanning CV, Caraway NP, et al. Percutaneous needle biopsy Liu K, Layfield LJ. Cytomorphologic features of angiosarcoma on fine
diagnosis of benign neurogenic neoplasms. Diagn Cytopathol 1997;16: needle aspiration biopsy. Acta Cytol 1999;43:407–415.
17–25. Mullick SS, Mody DR, Schwartz MR. Angiosarcoma at unusual sites. A report
Vendraminelli R, Cavazzana AO, Poletti A, et al. Fine-needle aspiration of two cases with aspiration cytology and diagnostic pitfalls. Acta Cytol
cytology of malignant nerve sheath tumors. Diagn Cytopathol 1992;8: 1997;41:839–844.
559–562. Wakely PE Jr, Frable WJ, Kneisl JS. Aspiration cytopathology of epithelioid
angiosarcoma. Cancer. 2000;90:245–251.
Rhabdomyosarcoma
Brahmi U, Rajwanshi A, Joshi K, et al. Role of immunocytochemistry and Gastrointestinal Stromal Tumor
DNA flow cytometry in the fine-needle aspiration diagnosis of malignant Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal
small round-cell tumors. Diagn Cytopathol 2001;24:233–239. stromal tumors: a consensus approach. Hum Pathol 2002;33:459–
de Jong AS, van Kessel-van Vark M, van Heerde P. Fine needle aspiration 465.
biopsy diagnosis of rhabdomyosarcoma. An immunocytochemical study. Heinrich MC, Rubin BP, Longley BJ, Fletcher JA. Biology and genetic
Acta Cytol 1987;31:573–577. aspects of gastrointestinal stromal tumors: KIT activation and cyto-
Lussier C, Klijanienko J, Vielh P. Fine-needle aspiration of metastatic genetic alterations. Hum Pathol 2002;33:484–495.
nonlymphomatous tumors to the major salivary glands: a clinicopatho- Rader AE, Avery A, Wait CL, et al. Fine-needle aspiration biopsy diagnosis
logic study of 40 cases cytologically diagnosed and histologically cor- of gastrointestinal stromal tumors using morphology, immunocytochem-
related. Cancer 2000;90:350–356. istry, and mutational analysis of c-kit. Cancer 2001;93:269–275.
Pohar-Marinsek Z, Anzic J, Jereb B. Topical topic: value of fine needle Rossi G, Valli R, Bertolini F, et al. PDGFRA expression in differential
aspiration biopsy in childhood rhabdomyosarcoma: twenty-six years of diagnosis between KIT negative gastrointestinal stromal tumours and
experience in Slovenia. Med Pediatr Oncol 2002;38:416–420. other primary soft-tissue tumours of the gastrointestinal tract. Histo-
Pohar-Marinsek Z, Bracko M. Rhabdomyosarcoma. Cytomorphology, subtyp- pathology 2005;46:522–531.
ing and differential diagnostic dilemmas. Acta Cytol 2000;44: Wieczorek TJ, Faquin WC, Rubin BP, Cibas ES. Cytologic diagnosis of gastro-
524–532. intestinal stromal tumor with emphasis on the differential diagnosis
Seidal T, Walaas L, Kindblom LG, Angervall L. Cytology of embryonal rhab- with leiomyosarcoma. Cancer 2001;93:276–287.
domyosarcoma: a cytologic, light microscopic, electron microscopic, and Yantiss RK, Nielsen GP, Lauwers GY, Rosenberg AE. Reactive nodular fibrous
immunohistochemical study of seven cases. Diagn Cytopathol pseudotumor of the gastrointestinal tract and mesentery: a clinico-
1988;4:292–299. pathologic study of five cases. Am J Surg Pathol 2003;27:
Valencerina Gopez E, Dauterman J, Layfield LJ. Fine-needle aspiration 532–540.
biopsy of alveolar rhabdomyosarcoma of the parotid: a case report and
review of the literature. Diagn Cytopathol 2001;24:249–252.
Myxoma
Leiomyosarcoma
Akerman M, Rydholm A. Aspiration cytology of intramuscular myxoma. A
Barbazza R, Chiarelli S, Quintarelli GF, Manconi R. Role of fine-needle comparative clinical, cytologic and histologic study of ten cases. Acta
aspiration cytology in the preoperative evaluation of smooth muscle Cytol 1983;27:505–510.
tumors. Diagn Cytopathol 1997;16:326–330. Caraway NP, Staerkel GA, Fanning CV, et al. Diagnosing intramuscular
Ferretti M, Gusella PM, Mancini AM, et al. Progressive approach to the myxoma by fine-needle aspiration: a multidisciplinary approach. Diagn
cytologic diagnosis of retroperitoneal spindle cell tumors. Acta Cytol Cytopathol 1994;11:255–261.
1997;41:450–460. Fornage BD, Romsdahl MM. Intramuscular myxoma: sonographic appear-
Giuntoli RL II, Metzinger DS, DiMarco CS, et al. Retrospective review of ance and sonographically guided needle biopsy. J Ultrasound Med
208 patients with leiomyosarcoma of the uterus: prognostic indicators, 1994;13:91–94.
surgical management, and adjuvant therapy. Gynecol Oncol 2003;89: Gonzalez-Campora R, Otal-Salaverri C, Hevia-Vazquez A, et al. Fine needle
460–469. aspiration in myxoid tumors of the soft tissues. Acta Cytol 1990;34:
Hornick JL, Fletcher CD. Criteria for malignancy in nonvisceral smooth 179–191.
muscle tumors. Ann Diagn Pathol 2003;7:60–66. Silver WP, Harrelson JM, Scully SP. Intramuscular myxoma: a clinicopatho-
Hummel P, Cangiarella JF, Cohen JM, et al. Transthoracic fine-needle logic study of 17 patients. Clin Orthop Relat Res 2002;(403):
aspiration biopsy of pulmonary spindle cell and mesenchymal lesions: 191–197.
a study of 61 cases. Cancer 2001;93:187–198. Wakely PE Jr, Geisinger KR, Cappellari JO, et al. Fine-needle aspiration
Ito E, Saito T, Suzuki T, et al. Cytology of vaginal and uterine sarcomas. cytopathology of soft tissue: chondromyxoid and myxoid lesions. Diagn
Acta Cytol 2004;48:601–607. Cytopathol 1995;12:101–105.
Klijanienko J, Caillaud JM, Lagace R, Vielh P. Fine-needle aspiration of
leiomyosarcoma: a correlative cytohistopathological study of 96 tumors
in 68 patients. Diagn Cytopathol 2003;28:119–125. Epithelioid Sarcoma
Lin O, Olgac S, Zakowski M. Cytological features of epithelioid mesen-
Cardillo M, Zakowski MF, Lin O. Fine-needle aspiration of epithelioid
chymal neoplasms: a study of 21 cases. Diagn Cytopathol 2005;32:
sarcoma: cytology findings in nine cases. Cancer 2001;93:246–251.
5–10.
Gonzalez-Peramato P, Jimenez-Heffernan JA, Cuevas J. Fine-needle aspira-
Otano-Joos M, Mechtersheimer G, Ohl S, et al. Detection of chromosomal
tion cytology of ‘proximal-type’ epithelioid sarcoma. Diagn Cytopathol
imbalances in leiomyosarcoma by comparative genomic hybridization
2001;25:122–125.
and interphase cytogenetics. Cytogenet Cell Genet 2000;90:86–92.
Kitagawa Y, Ito H, Sawaizumi T, et al. Fine needle aspiration cytology of
Tao LC, Davidson DD. Aspiration biopsy cytology of smooth muscle tumors.
primary epithelioid sarcoma. A report of 2 cases. Acta Cytol 2004;48:
A cytologic approach to the differentiation between leiomyosarcoma
391–396.
and leiomyoma. Acta Cytol 1993;37:300–308.
Lin O, Olgac S, Zakowski M. Cytological features of epithelioid mesenchymal
neoplasms: a study of 21 cases. Diagn Cytopathol 2005;32:5–10.
Angiosarcoma Zeppa P, Errico ME, Palombini L. Epithelioid sarcoma: report of two cases
diagnosed by fine-needle aspiration biopsy with immunocytochemical
Boucher LD, Swanson PE, Stanley MW, et al. Cytology of angiosarcoma. correlation. Diagn Cytopathol 1999;21:405–408.
Findings in fourteen fine-needle aspiration biopsy specimens and one
pleural fluid specimen. Am J Clin Pathol 2000;114:210–219.
Klijanienko J, Caillaud JM, Lagace R, Vielh P. Cytohistologic correlations Alveolar Soft Part Sarcoma
in angiosarcoma including classic and epithelioid variants: Institut
Curie’s experience. Diagn Cytopathol 2003;29:140–145. Lopez-Ferrer P, Jimenez-Heffernan JA, Vicandi B, et al. Cytologic features
Lin O, Olgac S, Zakowski M. Cytological features of epithelioid mesenchy- of alveolar soft part sarcoma: report of three cases. Diagn Cytopathol
mal neoplasms: a study of 21 cases. Diagn Cytopathol 2005;32:5–10. 2002;27:115–119.
126 FINE NEEDLE ASPIRATION CYTOLOGY

Shabb N, Sneige N, Fanning CV, Dekmezian R. Fine-needle aspiration cytol- reexamination of 37 cases, including ancillary diagnostics. A Scandina-
ogy of alveolar soft-part sarcoma. Diagn Cytopathol 1991;7:293– vian Sarcoma Group study. Diagn Cytopathol 2003;28:232–238.
298. Ewing CA, Zakowski MF, Lin O. Monophasic synovial sarcoma: a cytologic
spectrum. Diagn Cytopathol 2004;30:19–23.
Klijanienko J, Caillaud JM, Lagace R, Vielh P. Cytohistologic correlations
Synovial Sarcoma in 56 synovial sarcomas in 36 patients: the Institut Curie experience.
Diagn Cytopathol 2002;27:96–102.
Akerman M, Ryd W, Skytting B; Scandinavian Sarcoma Group. Fine-needle
aspiration of synovial sarcoma: criteria for diagnosis: retrospective
5 Breast
Shahla Masood

MASTITIS, ABSCESS, AND FAT NECROSIS MASTITIS, ABSCESS, AND FAT NECROSIS –
PATHOLOGIC FEATURES

Cytopathologic Findings
Inflammation of the breast as the result of lactation,
៉ Cellular aspirates
infection, or trauma often presents as a palpable breast ៉ Abundant infl ammatory cells
lesion with varying degrees of pain and tenderness. ៉ Cytophagocytosis and granular debris in the background
Acute supportive mastitis is typically seen in 1% to ៉ Epithelial cells with nuclear enlargement and prominent nucleoli
3% of lactating women in the postpartum period. The ៉ Occasional multinucleated cells
most common organisms are staphylococci and strep- ៉ Macrophages
tococci. When inflammation localizes, acute mastitis
often results in an abscess and rarely leads to chronic Ancillary Studies
mastitis with periductal inflammation, duct ectasia, ៉ Microbiologic examination
fibrohistiocytic reaction, and mononuclear chronic ៉ Culture and sensitivity
inflammatory infi ltrate.
Cytologically, smears are cellular and contain abun- Differential Diagnosis and Pitfalls
dant numbers of inflammatory infiltrates, histiocytes ៉ Subareolar abscess: cellular aspirate; abundant acute
with evidence of cytophagocytosis and nuclear debris. infl ammatory cells; anucleated squamous cells and keratinous
Isolated and clusters of epithelial cells with reactive material; macrophages and foreign body type giant cells
atypia are also present (Fig. 5-1). The differential ៉ Granulomatous mastitis: cellular aspirate; abundant chronic
infl ammatory cells, mostly lymphocytes and plasma cells;
diagnosis includes subareolar abscess, granulomatous
granulomatous reaction and giant cells; fibroblasts and reactive
mastitis, and fat necrosis with organized hematoma. atypical epithelial cells
Subareolar abscess is a specific clinicopathologic ៉ Fat necrosis and organized hematoma: cellular aspirate; necrotic
entity, which was first described by Zuka et al as a low- background with granular debris; lipid and hemosiderin-
grade infection in the lactiferous duct or sinus with containing macrophages; multinucleated giant cells;
subsequent abscess formation, chronic recurrent infec- mononuclear infl ammatory cells; small, newly formed vessels;
tion, and fistula formation at the base of the nipple. reactive atypical epithelial cells; cholesterol crystals
Suggestion has been made that squamous metaplasia of
columnar epithelial cells of the lactiferous ducts is the
cause of this lesion. The breast aspirate shows rich
cellularity with abundant inflammatory cells and many
anucleated squamous cells, and keratinous material Granulomatous mastitis is characterized by the
(Fig. 5-2). presence of granulomatous reaction and giant cell
formation. This entity is an inflammatory lesion of an
unknown etiology or may be the result of tuberculosis,
MASTITIS, ABSCESS, AND FAT NECROSIS – DISEASE FACT SHEET fungal infection, epidermal inclusion cyst, or foreign
body reaction such as suture and leakage from silicone
Clinical Features implants (Fig. 5-3).
៉ Inflammatory conditions of the breast account for 5–10% of all Fat necrosis and organized hematoma are trauma-
breast diseases induced inflammatory conditions that predominantly
៉ Primary causes of inflammatory conditions of the breast are present with foamy or hemosiderin-containing macro-
lactation, infection, and trauma phages, lymphocytes, plasma cells, fibroblasts, fragments
៉ Approximately 70% of abscesses are non-lactational and only 30% of fibrous tissue, and newly formed vessels (Fig. 5-4).
are related to puerperium Reactive epithelial atypia associated with fat necrosis
៉ Clinically, they present as a generalized cellulitis or a localized
has resulted in false positive diagnosis of cancer. Mam-
mass which may be associated with pain and tenderness
៉ Clinically and mammographically, they may mimic carcinoma
mographically, fat necrosis with subsequent calcifica-
៉ FNAB may serve as a diagnostic and therapeutic procedure
tion may also mimic a neoplastic process.
In the above-mentioned inflammatory conditions,
surgery can be avoided by the use of fine needle
127

Ch005-F06731.indd 127 10/26/2006 10:25:43 AM


128 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 5-1 FIGURE 5-3


Direct smear of breast abscess: cellular aspirate containing abundant Direct smear of a granulomatous infl ammation: non-caseating granu-
acute infl ammatory infiltrate and a few isolated and clusters of epithe- loma with epithelioid cells, mononuclear infl ammatory cells, and giant
lial cells with infl ammatory atypia. cells.

FIGURE 5-2
Direct smear of subareolar abscess: cellular aspirate with abundant
FIGURE 5-4
acute infl ammatory cells and squamous and anucleated cells.
Direct smear of fat necrosis: cellular aspirate with amorphous material
in the background, acute infl ammatory cells, abundant macrophages,
and newly formed vessels.
aspiration biopsy (FNAB). In conjunction with micro-
biologic studies of the aspirated material, FNAB can
provide valuable information about the etiology of an
inflammatory condition of the breast. It can also serve
as a therapeutic modality.
TREATMENT-INDUCED CHANGES – DISEASE FACT SHEET

Clinical Features
TREATMENT-INDUCED CHANGES ៉ Patients often present with thickening and/or a skin nodule at
the site of previous operation or following radiation therapy
៉ Cytologic distinction between a reactive process and a recurrent
tumor is critical in further management
Recent interest in breast cancer conservative therapy ៉ Radiation-induced angiosarcoma can also present as a nodule
and neoadjuvant chemotherapy has brought a new following conservation therapy
diagnostic challenge for pathologists/cytopathologists.
The distinction between recurrent breast carcinoma

Ch005-F06731.indd 128 10/26/2006 10:25:44 AM


CHAPTER 5 Breast 129

FIGURE 5-5 FIGURE 5-6


Direct smear of recurrent breast carcinoma following radiation therapy, Direct smear of recurrent breast carcinoma following radiation therapy:
characterized by isolated and clusters of highly pleomorphic epithelial higher magnification of the case seen in Fig. 5-5, highlighting necrosis
cells, macrophages, and necrosis in the background. and macronucleoli of neoplastic cells.

TREATMENT-INDUCED CHANGES – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Poor cellularity
៉ Granulation tissue
៉ Fat necrosis
៉ Epithelial atypia
៉ Granulatomous reaction
៉ Fibroblastic/spindle cell reaction
៉ Lymphoplasmacytic infiltration

Ancillary Studies
៉ Positive immunostaining reaction with endothelial cell markers

Differential Diagnosis and Pitfalls


៉ Recurrent tumor: rich cellularity; similar cytonuclear features to
the original tumor; conspicuous anisonucleosis with distinct
FIGURE 5-7
cell borders
៉ Angiosarcoma: moderate to rich cell yield; epithelioid and Direct smear of radiation-induced angiosarcoma: isolated, clustered,
and neoplastic spindle cells.
spindle cell pattern; various-sized nuclei with multiple nucleoli;
arborizing vessels and rosette-like structures

diagnosis of recurrent carcinoma in a sparsely cellular


aspirate from an irradiated breast.
and treatment-induced atypia is a difficult task, since Radiation-induced angiosarcoma may also present as
these entities present with overlapping features. It is a skin or subcutaneous nodule. This entity can easily be
suggested that radiation-induced changes in the breast mistaken for a recurrent carcinoma. Typically, patients
exhibit three different patterns in FNAB smears. These present with a discrete skin nodule several years after
include epithelial atypia, fat necrosis, and poor cellular- breast conservation therapy. Cytologically, breast aspi-
ity. In contrast, malignancy in an irradiated breast rates yield moderate to high cellularity with a variable
presents with a cellular aspirate featuring loss of cell pattern. Tumor cells are spindly and show positive
cellular cohesion, arrangement of cells in small reaction for endothelial markers by immunocytochem-
clusters, anisonucleosis, conspicuous nucleoli, an irreg- istry. Often, a network of thin, arborizing vessels is seen
ular nuclear membrane, and necrosis (Figs 5-5 & 5-6). in the background. Tumor cells may form pseudo-
In our own experience, the most important feature rosettes and have large nuclei with multiple nucleoli
in distinction between a radiation-induced change and (Figs 5-7 & 5-8). A high index of clinical suspicion is
recurrent malignancy is the rich cellularity of the the key factor in rendering an accurate diagnosis of
aspirate. It may be advisable to refrain from making a angiosarcoma by FNAB.

Ch005-F06731.indd 129 10/26/2006 10:25:46 AM


130 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 5-8
Cell block preparation of same case as Fig. 5-7, a radiation-induced FIGURE 5-10
angiosarcoma. Positive immunostaining with factor VIII. Direct smear of treatment-induced changes following surgery, showing
an ill-defined granuloma characterized by epithelioid cells, infl amma-
tory cells, and granular fibers.

Lack of cytologic similarity to the primary tumor and


predominance of spindle cells are important diagnostic
clues to exclude a recurrent tumor. Therefore it is criti-
cal to review the original pathology slides of each patient
for comparison.

HORMONAL CHANGES: SECRETORY,


PREGNANCY, AND LACTATIONAL CHANGES

The recognition of the spectrum of changes that reflect


FIGURE 5-9
the biologic attractions in normal mammary epithelial
cells is essential for the accurate interpretation of
Direct smear of treatment-induced changes following surgery, charac-
terized by the granulation tissue, new vessel formation, abundant various entities in breast pathology. The breast is a
infl ammatory cells, and giant cell reaction. hormone-dependent organ which undergoes puberty,
pregnancy, lactation, and involution and is influenced
by exogenous hormone effect. These changes may
Preoperative chemotherapy in locally advanced breast introduce cytonuclear alterations that may be confused
carcinomas may also produce tissue reaction with and with real disease process. The normal breast is a glan-
without residual tumor. These tissue reactions are dular tissue with various amount of mammary adipose
similar to those with other forms of therapy, such as tissue. During prepubertal and involutionary phases,
granulomatous reaction, necrosis, and epithelial atypia. the aspirates are often hypocellular with a few ductal
In addition, the presence of a lymphoplasmacytic reac-
tion may be conspicuous.
Surgery-induced fibrosis and suture granulomas also
present with reactive inflammatory atypia that should HORMONAL CHANGES: SECRETORY, PREGNANCY, AND
be separated from recurrent cancer. The typical form of LACTATIONAL CHANGES – DISEASE FACT SHEET
suture granuloma is characterized by the presence of
granulation tissue, granulomatous inflammation, histio- Clinical Features
cytes, mononuclear inflammatory cells, blood vessels, ៉ Pregnancy-associated lesions may present as galactocele,
and giant cells (Figs 5-9 & 5-10). Occasionally, retract- lactating adenoma, and fibroderma
៉ The distinction between pregnancy in cancer and pregnancy-
able suture material can also be seen. However, exces-
associated changes is critical in evaluation of pregnant patients
sive amount of granulation tissue, atypical fat necrosis, with a palpable breast lesion
and atypical immature fibroblasts in surgery-induced
changes may be misinterpreted as a malignant tumor.

Ch005-F06731.indd 130 10/26/2006 10:25:46 AM


CHAPTER 5 Breast 131

FIGURE 5-12
FIGURE 5-11 Direct smear of a pregnancy-associated change: dispersed cell pattern
Direct smear of a pregnancy-associated change: cellular aspirate with with enlarged cells, prominent nucleoli, granular debris in the back-
bubbly background, dispersed cell pattern, and a few macrophages. ground, and fraying of cytoplasmic borders.

HORMONAL CHANGES: SECRETORY, PREGNANCY, AND


LACTATIONAL CHANGES – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Cellular aspirate
៉ Proteinaceous background with bubbly appearance
៉ Dispersed cells and loosely arranged cell clusters
៉ Large epithelial cells with uniform nuclei and prominent
nucleoli
៉ Cytoplasmic vacuolization with fraying of cytoplasmic borders
៉ Bipolar naked nuclei, foamy macrophages, and occasional
multinucleated giant cells

Ancillary Studies
៉ Immunostain for myoepithelial cell markers

Differential Diagnosis and Pitfalls


FIGURE 5-13
៉ Cancer in pregnancy: high cellular yield; dispersed cell pattern
Direct smear of a galactocele characterized by a cluster of epithelial
with intact cell borders; marked anisonucleosis and prominent
cells surrounded by abundant lipid-laden macrophages.
nucleoli; proteinaceous background

associated changes present as galactocele, lactating


adenoma, or fibroderma.
Galactoceles often present as a discrete and mobile
epithelial cells and fragments of fibroadipose tissue. mass, which may increase in size during breastfeeding.
Secretory changes in mammary epithelial cells occur The mass regresses following FNAB. The smears show
in normal breast or in association with other entities low to moderate cellularity with numerous foamy cells,
such as fibroderma, proliferative breast disease and in proteinaceous background, and a few epithelial cells
association with a neoplastic process. with vacuolated cytoplasm (Fig. 5-13).
Pregnancy results in an increased number of acini Lactating adenomas are initially noticed during preg-
per lobule, with accumulation of secretory material in nancy. Aspirates from lactating adenomas are cellular
the lobular epithelial cells. These changes occur de novo, and show grape-like clusters of epithelial cells similar to
as a lactating adenoma or due to the enlargement of expanded acini, attached to terminal ductules. The cells
pre-existing tumors such as tubular adenoma and fibro- and the background have all the features of hormone-
adenomas. Fine needle aspirates of pregnancy and induced changes.
lactational changes are rich in cellularity with a bubbly Fibrodermas become more conspicuous in size during
appearance and proteinaceous background. The cells pregnancy. The cytologic features are similar to those
contain several secretory vacuoles with fraying of of lactating adenoma. In fibroadenomas there are
cytoplasmic borders (Figs 5-11 & 5-12). Pregnancy- bipolar stripped nuclei, more stromal fragments, less

Ch005-F06731.indd 131 10/26/2006 10:25:47 AM


132 FINE NEEDLE ASPIRATION CYTOLOGY

FIBROADENOMAS – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Highly cellular aspirate
៉ Biphasic pattern of stromal component and epithelial/
myoepithelial cells
៉ Sheets of monolayered ductal epithelial cells forming antler
horns
៉ Bipolar naked nuclei
៉ Occasional apocrine cells
៉ Rare multinucleated cells

Ancillary Studies
៉ Immunostain for myoepithelials cell markers

Differential Diagnosis and Pitfalls


៉ Proliferative breast disease: lack of stromal component; lack of
FIGURE 5-14 antler-horn pattern; lack of naked nuclei in the background
Direct smear of primary breast cancer in pregnancy: isolated and clus- ៉ Papillary lesions: presence of fibrovascular core; abundance of
ters of neoplastic epithelial cells with an intact cell membrane. macrophages; conspicuous columnar cell differentiation
៉ Low-grade carcinoma: absence of naked nuclei in the
background; absence of discrete stromal components; absence
of myoepithelial cells
៉ Phyllodes tumor: cellular stromal component; variable degree of
proteinaceous material, and lipid droplets in the back- atypicality and mitosis
ground. They may contain a few giant cells. Pregnancy
and lactational changes often present with loosely
cohesive cellular aspirates with enlarged cells with con-
spicuous nucleoli. Those features mimic primary breast
carcinoma. This distinction is a diagnostic challenge, a small, solitary and painless nodule. Mammographi-
particularly since pregnancy-associated changes can cally, they appear as well-defined lesions, and in 7% to
also be seen in non-lactating women. 16% of cases, they may show evidence of multiplicity.
Primary breast carcinoma occurs in 1 out of 1000 Carcinoma, often of lobular type, rarely occurs in fibro-
pregnancies. Aside from anisonucleosis and chromatin adenomas. Giant fibroadenomas are rapidly growing
clumping, the most important cytologic feature in favor fibroadenomas that occur in adolescent girls and they
of carcinoma is the presence of intact cell membrane in may become as large as 19 cm.
the aspirated smears (Fig. 5-14). Cytologically, fibroadenomas yield cellular aspirates
FNAB is an effective initial tool in the evaluation of showing a biphasic pattern of epithelial and stromal
pregnant women with a palpable mass. This approach elements. The cell population displays two cell types:
reduces the delay in the diagnosis of cancer in pregnant epithelial and myoepithelial. The epithelial component
and lactating women. Further follow-up and manage- forms sheets of monolayered epithelial cells with clus-
ment of a pregnant breast cancer patient depend on the ters forming antler-horn projections. Myoepithelial cells
stage of pregnancy and the choice of the patient. are smaller and darker, and often spindly in appearance.
These cells appear in the background with bipolar and
naked nuclei and are admixed with epithelial cells (Fig.
5-15). Classical-pattern fibroadenomas are easy to diag-
nose; however, when they are associated with prolifera-
FIBROADENOMAS tive breast disease, undergo metaplastic change, or
are affected by hormones, they become atypical and
cellular. Loose edematous stroma occasionally displays
Fibroadenomas are slow-growing tumors that are under myxoid features mimicking mucinous carcinoma (Figs
the influence of unopposed estrogen. They are the most 5-15–5-20).
common tumor in young women and often present as Cellular fibroadenomas are the most common cause
of false positive diagnosis in breast FNAB. Differential
diagnoses include proliferative breast disease, papillary
lesions, and low-grade carcinomas such as tubular car-
FIBROADENOMAS – DISEASE FACT SHEET
cinoma. In addition, distinction between fibroadenoma
Clinical Features
and benign phyllodes tumor is difficult.
High cellularity, presence of a stromal component
៉ Most common benign tumor of the breast
៉ Often presents as a mobile breast mass
that is distinctly separated from the epithelial compo-
៉ Mammographically presents as a well-defined lesion
nent, naked nuclei in the background, as well as antler-
horn cell clusters are the features that differentiate
between proliferative breast disease and fibroadenomas.

Ch005-F06731.indd 132 10/26/2006 10:25:49 AM


FIGURE 5-15 FIGURE 5-16
Direct smear of a fibroadenoma characterized by a biphasic pattern, Direct smear of a cellular fibroadenoma characterized by a cellular
showing isolated and clusters of epithelial/myoepithelial cells and the aspirate with isolated and clusters of epithelial/myoepithelial cells with
associated stromal components. Note the presence of myoepithelial numerous naked nuclei in the background. The clusters show overriding
cells as naked nuclei in the background. of the nuclei commonly seen in proliferative breast disease.

FIGURE 5-17 FIGURE 5-18


Direct smear of a cellular fibroadenoma, showing higher magnification Direct smear of a cellular fibroadenoma associated with proliferative
of the same features as in Fig. 5-14. breast disease. Another view of the same case as in Fig. 5-16, showing
the presence of a biphasic pattern commonly seen in fibroadenoma.

FIGURE 5-19
Direct smear of a fibroadenoma with loose myxoid background, simulat- FIGURE 5-20
ing mucoid carcinoma. The presence of myoepithelial cells in the back- Direct smear of the fibroadenoma with myxoid stroma as seen in Fig.
ground substantiates the diagnosis of fibroadenoma. 5-17 and the associated antler-horn pattern seen in the same case.

Ch005-F06731.indd 133 10/26/2006 10:25:49 AM


134 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 5-22
FIGURE 5-21
Direct smear of a benign phyllodes tumor: higher view of case shown
Direct smear of a benign phyllodes tumor characterized by a biphasic in Fig. 5-21, exhibiting cellular stroma characteristic of a phyllodes
pattern of epithelial/myoepithelial cells and the associated cellular tumor.
stroma.

Papillary lesions may also resemble a fibroadenoma. PAPILLARY BREAST LESIONS – DISEASE FACT SHEET
Key features in favor of papillary lesions are high cellu-
larity and three-dimensional architecture of branched Clinical Features
epithelial clusters, the presence of fibrovascular cores, ៉ Uncommon tumors of the breast
various numbers of macrophages, apocrine cells, and ៉ Occur in postmenopausal women
columnar epithelial cell clusters. Distinction between ៉ Presenting symptoms are nipple discharge, nipple retraction, and
tubular adenoma and fibroadenoma is usually not association with a palpable mass
possible because of overlapping features. However, this ៉ Complete removal of the lesion is the recommended procedure
distinction has no clinical implication.
Fibroadenomas are known to simulate carcinoma.
Extensive search for stromal elements and the presence
of myoepithelial cells are the features favoring a fibro-
adenoma. The myoepithelial origin of spindle cells can PAPILLARY BREAST LESIONS – PATHOLOGIC FEATURES
be demonstrated by expression of myoepithelial cell
markers by immunocytochemistry. Cytopathologic Findings
The distinction between fibroadenoma and phyllodes ៉ Intraductal papilloma: cellular aspirate; proteinaceous or bloody
tumor in an aspirate is based on higher cellularity of background; three-dimensional papillary cell clusters; foamy or
hemosiderin-laden macrophages; myoepithelial cells; tall
stromal elements seen in phyllodes tumors. These cases
columnar cells
should be checked for the presence of abnormal mitosis ៉ Papillary carcinoma: cellular aspirate; bloody background;
and pleomorphism to exclude the possibility of a malig- hemosiderin-laden macrophages; occasional necrotic debris;
nant phyllodes tumor (Figs 5-21 & 5-22). It is important papillary clusters of atypical cells enriched by a fibrovascular
to note that phyllodes tumors often occur in an older core; large atypical nuclei; tall columnar cells; absence of
age than fibroadenomas and present with a larger pal- myoepithelial cells
pable mass of over 4 cm. A follow-up surgical excision ៉ Intercystic papillary carcinoma: cellular aspirate; hemosiderin-
of fibroadenomas with atypical features is the current laden macrophages; monomorphic cell population;
procedure. anisonucleosis and pleomorphism; absence of myoepithelial
cells; papillary and/or cribriform pattern

Ancillary Studies
៉ Immunostains for myoepithelial cell markers
PAPILLARY BREAST LESIONS

Papillary lesions of the breast constitute a spectrum of women and are frequently seen in the male breast.
pathologic entities ranging from intraductal papillomas Nipple discharge, nipple retraction, and a subareolar
to papillary carcinomas. Intraductal papillomas, the mass are the pre-existing symptoms.
benign forms of papillary lesions, are solitary lesions Cytologic diagnosis of intraductal papillomas can be
of the major ducts, commonly based in the subareolar made by nipple fluid cytology or by the examination of
region. Papillomas often occur in postmenopausal the breast FNAB. Cytologically, the aspirates are cellu-

Ch005-F06731.indd 134 10/26/2006 10:25:51 AM


CHAPTER 5 Breast 135

FIGURE 5-23 FIGURE 5-25


Direct smear of an intraductal papilloma characterized by cellular aspi- Direct smear of an intraductal papilloma exhibiting a cell ball or three-
rate showing proliferating epithelial cells with myoepithelial cells in dimensional structure.
the background.

FIGURE 5-26
FIGURE 5-24 Direct smear of an intraductal papilloma showing clusters of apocrine
cells, columnar cells, and histiocytes in the background.
Direct smear of an intraductal papilloma showing proliferating cells
forming various architectural patterns.

evidence of skin dimpling and nipple retraction. The


lar and contain a proteinaceous background and con- neoplastic cells may display a variety of patterns such
spicuous numbers of macrophages. Three-dimensional as cribriform and micropapillary. The absence of a myo-
cell balls, tall columnar cells, apocrine cells, and myo- epithelial cell layer is one of the most important charac-
epithelial cells are also present (Figs 5-23–5-26). teristics of papillary carcinoma (Fig. 5-30).
Distinction between an intraductal papilloma and Another focus of papillary lesions of the breast is an
well-differentiated papillary carcinoma may be some- intracystic carcinoma. This is a rare breast tumor
what difficult. These entities share common cytologic accounting for 0.7% of all breast carcinomas. It fre-
features. However, papillary carcinomas often present quently occurs in black and obese women, with bloody
with a monomorphic and uniform-appearing cell nipple discharge as the first presenting symptom. Breast
population with no evidence of myoepithelial cell dif- imaging shows a well-circumscribed cystic mass that
ferentiation. There are also no apocrine cells (Figs persists following FNAB. Cytologically, they mimic the
5-27–5-29). features seen in papillary carcinoma.
Papillary carcinomas account for 1% to 2% of breast The diagnosis of papillary lesions of the breast by
cancers and are associated with a good prognosis. Bloody FNAB requires complete excision of the lesion with a
discharge is the most common presenting symptom, and rim of normal breast tissue. It is advisable to refrain
in 90% of cases, a mass can be palpated. There is also from attempting to do frozen section because of

Ch005-F06731.indd 135 10/26/2006 10:25:52 AM


136 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 5-27 FIGURE 5-29


Direct smear of a papillary carcinoma characterized by highly cellular Direct smear of a papillary carcinoma showing a higher magnification
aspirate of clusters of epithelial cells forming different architectural of a monomorphic cell population with columnar cell differentiation.
patterns.

FIGURE 5-28
Direct smear of a papillary carcinoma showing isolated and clusters of FIGURE 5-30
a monomorphic population of tumor cells with columnar cell differentia-
tion surrounding a fibrovascular core. No apocrine cells are seen. Immunostained slide: cell block preparation of a papillary carcinoma
with no evidence of myoepithelial cell differentiation.

experiencing the same difficulty in rendering a definite


diagnosis as in cytology.

FIBROCYSTIC CHANGE – DISEASE FACT SHEET

FIBROCYSTIC CHANGE Clinical Features


៉ The most common breast lesions
៉ Present as a non-palpable breast lesion or as a palpable mass
Fibrocystic change represents a spectrum of changes ៉ Asystematic density, mass effect, and calcification are the
ranging from normal physiologic alterations to high- common features in breast imaging
៉ The degree of proliferation correlates positively with the
risk proliferative breast disease. Cyst formation, apo-
incidence of subsequent development of breast cancer
crine neoplasia, stromal fibrosis, sclerosing adenosis,
៉ As a morphologic risk factor, recognition of atypical ductal
and various degrees of epithelial hyperplasia are the hyperplasia helps to stratify patients for risk-reduction modalities
features of fibrocystic change. The association between
these morphologic changes and the subsequent

Ch005-F06731.indd 136 10/26/2006 10:25:53 AM


CHAPTER 5 Breast 137

Cytologically, a semiquantitative score, the ‘Masood


FIBROCYSTIC CHANGE – PATHOLOGIC FEATURES Cytology Index’, has been developed. This cytologic
grading system is based on the comparison of cytomor-
Cytopathologic Findings phology of mammographically detected breast fine
៉ Non-proliferative breast disease: low to moderate cellularity; needle aspirates and their corresponding needle local-
fragments of stroma and adipose tissue; monolayered cell ization excisional biopsies. Attempts have been made to
population with honeycomb pattern; histiocytes, apocrine cells, incorporate cellular arrangement with the traditional
and myoepithelial cells nucleocytologic features and presence or absence of
៉ Proliferative breast disease without atypia: moderate to high
myoepithelial cells (Tables 5-1 & 5-2). Controversy
cellularity; abundant clusters of two-cell population of
epithelial/myoepithelial cells; conspicuous overriding of the
regarding the use of this cytologic grading system in
nuclei; occasional nucleoli and focal loss of polarity; apocrine clinical practice continues. However, this index has
cells, histiocytes, and occasional calcified particles been used as a morphologic risk factor in chemopreven-
៉ Proliferative breast disease with atypia (atypical ductal tion trials (Fig. 5-31). Recently, an association between
hyperplasia): high cellularity; clustering of epithelial/ the degree of morphologic abnormality defined by this
myoepithelial cells; marked crowding of nuclei forming slit-like index and the frequency of expression of retinoic acid
openings and loss of polarity; nuclear enlargement and receptor-β2 promoter methylation has been reported
macronucleoli; chromatin clumping; occasional apocrine cells (Fig. 5-32).
and microcalcified particles

Ancillary Studies
៉ Immunostain for myoepithelial cell markers

Differential Diagnosis and Pitfalls


៉ Papillary lesions: tall columnar cells; hemosiderin-containing
24
macrophages; fibrovascular core
៉ Fibroadenoma: distinct stromal component; naked nuclei in the P<0.0001
background; antler-horn pattern
16
៉ Low nuclear grade ductal carcinoma in situ: monotonous cell
population; lack of myoepithelial cells Total high risk
cohort (N=480)
8
3.9% at 45 months

No FNA atypia (N=378)


0
development of breast cancer is now well established. 0 12 24 36 48 60 72 84 96 108
In addition, the new terminology of non-proliferative
Time from entry on study (months)
breast disease, proliferative breast disease without
atypia and proliferative breast disease with atypia FIGURE 5-31
(atypical hyperplasia), have replaced the commonly The relationship between cellular atypia in fine needle aspiration biopsy
used term fibrocystic disease. and increased risk for breast cancer.

TABLE 5-1
Masood Cytology Index

Cellular Cellular Myoepithelial Anisonucleosis Nucleoli Chromatin Score


arrangement pleomorphism cells clumping

Monolayer Absent Many Absent Absent Absent 1


Nuclear overlapping Mild Moderate Mild Micronucleoli Rare 2
Clustering Moderate Few Moderate Micro- and/or rare Occasional 3
macronucleoli
Loss of cohesion Conspicuous Absent Conspicuous Predominately Frequent 4
macronucleoli

Sum of scores: 6–10 Normal


11–14 Proliferative
15–18 Atypia
≥19 Suspicious for cancer
Modified from Masood S, Frykberg ER, McLellan GL, et al. Prospective evaluation of radiologically directed fine-needle
aspiration biopsy of nonpalpable breast lesions. Cancer 1990;66:1480–1487.

Ch005-F06731.indd 137 10/26/2006 10:25:55 AM


138 FINE NEEDLE ASPIRATION CYTOLOGY

Methylation present Methylation absent


12
M3 RARbeta P2 methylation
10

Number RPFNA
8 FIGURE 5-32
6 The association between Masood Cytology
Index score and the expression of retinoic acid
4 receptor-β2 promoter methylation in random
periareolar fine needle aspiration.
2

0
10 11 12 13 14 15 >15
Masood cytology index

TABLE 5-2
Concordance Between Cytologic Evaluation and
Histologic Diagnosis in 100 Mammographically Guided
Fine Needle Aspirates

Diagnosis No. of cases Concordance %

Non-proliferative breast 29/34 85


disease
Proliferative breast disease 15/17 88
without atypia
Proliferative breast disease 21/23 91
with atypia
Cancer 17/20 85

Modified from Masood S, Frykberg ER, McLellan GL, et al.


Cytologic differentiation between proliferative and FIGURE 5-33
nonproliferative breast tissue in mammographically guided Direct smear of non-proliferative breast disease characterized by a
fine-needle aspirates. Diagn Cytopathol 1991;7:581–590. monolayered cluster of epithelial/myoepithelial cells, a cluster of
apocrine cells, stromal fragments, and a few histiocytes.

monolayered sheets with a honeycomb pattern. Foam


NON-PROLIFERATIVE BREAST DISEASE cells, apocrine cells, and myoepithelial cells are present
(Figs 5-33 & 5-34).
Non-proliferative breast disease may present as a cystic
lesion. This is a common condition which is often the
subject of FNAB. Cysts produce fluids that vary in PROLIFERATIVE BREAST DISEASE
appearance from clear to opaque, dark brown, yellow– WITHOUT ATYPIA
green to bloody. Benign cysts are acellular with granu-
lar debris. Occasionally, a few epithelial cells/apocrine
cells may be present. If associated with duct ectasia or FNAB of proliferative breast disease without atypia
chronic cystic mastitis, the smears are more cellular presents with a higher cell yield and unique cellular
and may contain inflammatory cells. Bloody cysts are arrangement. The honeycomb monolayered arrange-
also often associated with papillary lesions. Rarely, ment is changed to clusters of epithelial/myoepithelial
medullary carcinomas may become cystic and contain cells that show overriding of the nuclei, occasional loss
atypical malignant cells. Since more than 99% of cysts of polarity, and some variability in nuclear size. Micro-
are benign, there is controversy regarding the need for nuclei are occasionally seen. Cytologic atypia is incon-
cytologic examination of clear cysts. It may be more spicuous. Apocrine cells, histiocytes, and occasional
cost effective to limit cytologic analysis to the cysts that naked nuclei are seen in the background (Figs 5-35 &
are associated with a turbid or bloody appearance. 5-36).
In non-cystic lesions, aspirates from a non-prolifera- Differential diagnosis includes fibroadenoma and
tive breast disease are characterized by scant or moder- papillary lesions. These conditions have overlapping
ate cellularity. The uniform ductal cells are arranged in features. The presence of well-demarcated stroma,

Ch005-F06731.indd 138 10/26/2006 10:25:55 AM


CHAPTER 5 Breast 139

FIGURE 5-34
Direct smear of non-proliferative breast disease: a cluster of reactive
apocrine cells with micronucleoli.
FIGURE 5-36
Direct smear of proliferative breast disease without atypia: higher
magnification of the same case as in Fig. 5-35. Note the conspicuous
crowding of nuclei, with large, oval and pale epithelial cells intermixed
with small, dark and slender myoepithelial cells.

FIGURE 5-35
Direct smear of proliferative breast disease without atypia, character-
ized by clusters of proliferating epithelial/myoepithelial cells with
overriding of the nuclei and loss of honeycomb pattern.

FIGURE 5-37
Direct smear of proliferative breast disease with atypia (atypical ductal
various naked nuclei, and an antler-horn pattern hyperplasia) characterized by a loosely cohesive cluster of proliferating
epithelial/myoepithelial cells with crowding of the nuclei and loss of
are the features of fibroadenoma. In papillary lesions, polarity forming various-sized slits and openings.
hemosiderin-containing macrophages and polymorphic
populations of tall columnar cells forming papillae are
the differentiating features.
polarity of the contour of ductal elements. Nuclei
display a coarse chromatin pattern and there are some
PROLIFERATIVE BREAST DISEASE WITH variations in the cell nuclei. Intermingled with epithe-
ATYPIA (ATYPICAL HYPERPLASIA) lial cells are myoepithelial cells. Rare apocrine cells
and macrophages may also be present (Figs 5-37–
5-40). The differential diagnosis includes a low-grade
Breast aspirates of lesions with atypical hyperplasia are ductal carcinoma in situ. A monotonous cell arrange-
characterized by rich cellularity composed of several ment and the absence of myoepithelial cells are the
clusters of epithelial cells with conspicuous crowding key differentiating features. Immunostaining for
of the nuclei. The degree of overriding of nuclei forms myoepithelial cell markers is an important ancillary
different-sized slits and openings with some loss of study.

Ch005-F06731.indd 139 10/26/2006 10:25:55 AM


140 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 5-38 FIGURE 5-40


Direct smear of proliferative breast disease with atypia: higher magni- Direct smear of proliferative breast disease with atypia, showing an
fication, highlighting the presence of micronucleoli, nuclear atypia, isolated cluster of atypical epithelial cells. Note the persistence of
intermixed myoepithelial cells, and various-sized cribriform-type accompanying myoepithelial cells.
pattern of proliferation.

DUCTAL CARCINOMA IN SITU – DISEASE FACT SHEET

Clinical Features
៉ Increased emphasis in breast cancer screening has resulted in an
increased frequency of detection of in-situ lesions
៉ Microcalcification is the most common mammographic feature of
in-situ lesions
៉ Distinction between an in-situ lesion and an invasive process may
have clinical significance

DUCTAL CARCINOMA IN SITU – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Low nuclear grade: variable cellularity; monomorphic pattern of
FIGURE 5-39 cells forming cribriform, solid, and micropapillary patterns;
small to medium-sized cells with uniform nuclei; absence of
Direct smear of proliferative breast disease with atypia: high magnifica-
tion of another view, displaying nuclear features, architectural change, myoepithelial cells
and hyperchromasia. ៉ High nuclear grade: variable cellularity in a necrotic background;
pleomorphic population of highly anaplastic epithelial cells
with individual cell necrosis and mitosis; absence of
myoepithelial cells
DUCTAL CARCINOMA IN SITU

LOW NUCLEAR GRADE DUCTAL CARCINOMA dures requires a follow-up surgical biopsy to exclude
IN SITU (NON-COMEDO TYPE) the possibility of a more severe lesion. However, the
diagnosis of atypical ductal hyperplasia by FNAB has
the benefit of identifying high-risk individuals for
Distinction between atypical ductal hyperplasia and breast cancer who may benefit from chemoprevention
low nuclear grade ductal carcinoma has remained a and the available risk-reduction modalities. In recent
diagnostic challenge in breast pathology. This difficulty years, breast cancer awareness and screening programs
is compounded in interpretation of minimally invasive have resulted in an increased detection of borderline
specimens such as core needle biopsy and FNAB. It is breast disease and ductal carcinoma in situ. Clustered
generally agreed that the diagnosis of atypical ductal microcalcification is the most frequent feature in breast
hyperplasia by any of these minimally invasive proce- imaging.

Ch005-F06731.indd 140 10/26/2006 10:25:57 AM


CHAPTER 5 Breast 141

FIGURE 5-41 FIGURE 5-43


Direct smear of low nuclear grade ductal carcinoma in situ demonstrat- Direct smear of high nuclear grade ductal carcinoma in situ: pleomor-
ing clusters of a monomorphic cells forming a cribriform pattern. Myo- phic population of neoplastic cells with necrosis in the background.
epithelial cells are seen only at the periphery of cell clusters.

FIGURE 5-44
Direct smear of high nuclear grade ductal carcinoma in situ: pleomor-
phic cell population with individualized cell necrosis.
FIGURE 5-42
Direct smear of low nuclear grade ductal carcinoma in situ: higher view
of the same case as in Fig. 5-41 with similar features. the low nuclear grade ductal carcinoma in situ. It is
associated with a higher rate of microinvasion, expres-
sion of HER-2/neu oncogene, and hormone receptor
Breast aspirates from non-comedo-type ductal carci- negativity. Fine needle aspirates of high nuclear grade
noma in situ is characterized by moderate to high cel- ductal carcinoma in situ are often cellular and display
lularity. Cell population is monomorphic and uniform. highly neoplastic epithelial cells with individual cell
No myoepithelial cells are present. The cell clusters may necrosis. Extreme pleomorphism with anisonucleosis
form cribriform, solid, and micropapillary patterns. occasionally is associated with the microcalcified parti-
Microcalcified particles may also be present (Figs 5-41 cles. Large irregularly shaped nucleoli are also present
& 5-42). (Figs 5-43 & 5-44).
The distinction between high and low nuclear grade
is important since high nuclear grade lesions require
more aggressive therapy and better respond to
HIGH NUCLEAR GRADE DUCTAL chemotherapy. Assessment of the status of invasion
CARCINOMA IN SITU (COMEDO TYPE) may be critical for treatment planning. Attempts have
been made to cytologically differentiate between an in-
situ versus an invasive breast cancer. However, contro-
High nuclear grade ductal carcinoma in situ is known versy remains regarding the reliability of cytologic
to have a higher rate of local recurrence compared to features to make this distinction. In these circumstances,

Ch005-F06731.indd 141 10/26/2006 10:25:58 AM


142 FINE NEEDLE ASPIRATION CYTOLOGY

it is best to consider an open biopsy to determine the


presence or absence of an invasive process. INFILTRATING DUCT CELL CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Cellular aspirate
៉ Conspicuous loss of cell cohesion
PRIMARY BREAST CARCINOMA ៉ Variable cell pattern in a clean or necrotic background
៉ Variable nuclear atypia and cellular pleomorphism
៉ Rare multinucleated tumor giant cells
FNAB is one of the most cost-effective procedures for ៉ Occasionally, small cells with plasmacytoid appearance
the initial diagnosis of primary breast carcinoma in ៉ Absence of myoepithelial cells
palpable breast lesions. Associated with integration of
clinical presentation and breast imaging, in experi- Histopathologic Findings
enced hands FNAB provides a diagnostic accuracy ៉ Infiltrating duct cell carcinoma NOS varies based on the degree
similar to that of surgical biopsy. Diagnosis of malig- of differentiation
nancy in breast cytopathology is based on the combina-
Ancillary Studies
tion of architectural pattern and the nuclear features.
៉ Immunostain for epithelial versus melanoma specific markers
Aside from familiarity with subtle diagnostic pitfalls
associated with various forms of breast carcinoma, rec-
Differential Diagnosis and Pitfalls
ognition of the limitation of this procedure is the key to
៉ Infl ammatory/reactive/hyperplasia: presence of myoepithelial
an accurate diagnosis. It is critically important to study
cells; preservation of cellular cohesion; polymorphic cell
the spectrum of morphologic features of benign and
pattern
borderline breast disease that may present with atypia. ៉ Infiltrating lobular carcinoma: more uniformity of the nuclei;
To achieve this goal, attempts should be made to refrain less hyperchromasia; occasional Indian filing; occasional signet-
from the casual use of the term atypia. Similarly, recog- ring cells; presence of intracytoplasmic mucin vacuoles
nition of breast malignancies that are associated with ៉ Amelanotic melanoma: presence of intranuclear vacuoles;
insignificant atypia, such as lobular, tubular, and muci- frequent binucleation; prominent, often cherry red, nucleoli
nous lesions, is important. Based on the guidelines pro-
posed in the National Cancer Institute (NCI)-sponsored
workshop, rendering the diagnosis of malignancy in a
breast FNAB is sufficient for patient management.
However, typing of breast carcinoma whenever possible
is strongly encouraged.
ity or as a hard mass with or without skin changes. The
more common variety is the stellate lesion, which is
INFILTRATING DUCT CELL CARCINOMA, characterized by extensive fibrosis, also known as scir-
rhous carcinoma. Mammographically, the vast majority
NOT OTHERWISE SPECIFIED (NOS) of these malignancies present with a poorly defined
spiculated mass with or without microcalcifications.
Based on data from the Surveillance, Epidemiology, Sonographically, the most common features of malig-
and End Results (SEER) programs registry of the NCI, nancy are those of a hypoechoic mass with irregular
the most common primary carcinoma of the breast is borders and an uneven texture. Histologically, infi ltrat-
infi ltrating duct carcinoma, not otherwise specified ing duct cell carcinoma is characterized by a spectrum
(NOS). This form of primary breast cancer accounts of changes that vary accordingly to atypia and differ-
for 68% of all breast carcinomas. Infi ltrating duct cell entiation such as the degree tubule formation, nuclear
carcinoma can present as an image-detected abnormal- pleomorphism, and mitotic activity.
Cytologically, variable patterns reflecting the diverse
histology are seen. Breast aspirates of infiltrating duct
cell carcinoma are cellular and often show conspicuous
loss of cell cohesion. Scattered individual tumor cells
and aggregates of various-sized cells demonstrate three-
INFILTRATING DUCT CELL CARCINOMA – DISEASE FACT SHEET dimensional clustering, syncytial groups or, occasion-
ally, gland-like arrangements. Tumor cells are often
Clinical Features large and pleomorphic with prominent nucleoli. The
៉ Most common type of primary breast carcinoma background may be clear or contain red blood cells and
៉ Clinically presents with a palpable mass with or without skin necrotic debris. Occasionally, multinucleated giant cells
changes can also be observed. The above-mentioned morpho-
៉ Mammographically presents as a poorly defined spiculated mass
logic features are characteristic of poorly differentiated
with or without microcalcifications
infiltrating duct cell carcinoma/grade III tumors. Grade
៉ Characterized by a hypoechoic mass with irregular borders and an
uneven echo texture by ultrasound
I and grade II will exhibit sheets of monomorphic cells
with round and bland-appearing nuclei with single
inconspicuous nucleoli (Figs 5-45–5-47).

Ch005-F06731.indd 142 10/26/2006 10:25:59 AM


CHAPTER 5 Breast 143

FIGURE 5-45 FIGURE 5-47


Direct smear of an infiltrating duct cell carcinoma yielding a cellular Direct smear of an infiltrating duct cell carcinoma, high nuclear grade,
aspirate with conspicuous loss of cell cohesion. demonstrating various-sized nucleoli with clumped chromatin and con-
spicuous nucleoli.

FIGURE 5-46
Direct smear of an infiltrating duct cell carcinoma, low nuclear grade,
characterized by uniform small nuclei with inconspicuous nucleoli.
FIGURE 5-48
Direct smear of an infiltrating duct cell carcinoma, small cell variant,
showing numerous small cells with eccentric nuclei.

Another variant of duct cell carcinoma, which may


be difficult to diagnose cytologically, is the small cell Absence of myoepithelial cells is the key to making the
variant. Breast aspirate of this entity is characterized by distinction.
numerous isolated and clusters of small cells with eccen-
tric round nucleoli and a moderate amount of cytoplasm
exhibiting a ‘plasmacytic’ pattern. This entity should be
differentiated from the rare plasmacytomas of the breast
and from a metastatic amelanotic malignant melanoma. TUBULAR CARCINOMA
Immunostaining for epithelial markers aids in making
an accurate diagnosis of the small cell variant of infil-
trating duct cell carcinoma, which commonly occurs in Tubular carcinoma, a previously rare breast tumor,
older women. This tumor also shares a similar pattern now accounts for up to 20% of all carcinomas detected
with infiltrating lobular carcinoma. Higher cellularity through mammographic screening. It occurs in a
and increased hyperchromasia of the nuclei are the younger age group than the classic type of infi ltrating
features more commonly seen in this variant (Fig. duct cell carcinoma and is associated with a higher
5-48). incidence of bilaterality and multiplicity. Because of
Infiltrating duct cell carcinoma should be differenti- their small size and infrequent lymph node involve-
ated from borderline and atypical benign lesions. ment, tubular carcinomas are associated with a good

Ch005-F06731.indd 143 10/26/2006 10:25:59 AM


144 FINE NEEDLE ASPIRATION CYTOLOGY

TUBULAR CARCINOMA – DISEASE FACT SHEET

Clinical Features
៉ Accounts for up to 20% of mammographically detected
carcinomas
៉ Mammographically, it shares similar features with radial scar
៉ Associated with a low incidence of axillary lymph node metastasis
and a favorable prognosis
៉ Suitable for conservative therapy

TUBULAR CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings FIGURE 5-49


៉ Variable cellularity
Direct smear of tubular carcinoma characterized by isolated and clusters
៉ Presence of epithelial cells forming angulated tubular structures of epithelial cells with tubular structures and cell balls.
៉ Uniform nuclei with occasional nuclear grooving
៉ Cytoplasmic vacuoles

Ancillary Studies
៉ Immunostain for myoepithelial cell markers

Differential Diagnosis and Pitfalls


៉ Radial scar: low to moderate cellularity; admixed myoepithelial
cells with epithelial cells; absence of nuclear grooves and
intracytoplasmic vacuoles
៉ Microglandular adenosis: low cellularity; absence of angulated
tubules; cell balls; myoepithelial cells
៉ Fibroadenoma: complex branching of ductal epithelial cells;
myoepithelial cells within the clusters of epithelial cells and in
the background; absence of nuclear grooving and
intracytoplasmic vacuoles

prognosis and are most suitable for conservative FIGURE 5-50


therapy. Direct smear of tubular carcinoma showing an angulated tubular struc-
Mammographically, tubular carcinomas present as a ture with no myoepithelial cells within the cluster of epithelial cells.
spiculated mass, similar to that of a radical scar, which
may require surgical excision. Grossly, tubular carcino-
mas present as stellate or scirrhous areas of gray–white
discoloration with induration and retroaction. Histo-
logically, the tumor consists of elongated round tubular
structures lined by a single layer of cells, some of which
may exhibit apocrine snouts.
Cytologically, the aspirates are moderately cellular,
containing bland-appearing tumor cells which form
angular branching and three-dimensional tubular struc-
tures. Tumor cells are uniform with a finely granular
chromatin and small nucleoli. Groved nuclei and solitary
intracytoplasmic vacuoles are often features of tubular
carcinomas (Figs 5-49–5-51). The differential diagnosis
includes radial scar, fibroadenoma, and microglandular
adenosis. There are overlapping features between tubular
carcinoma and radial scar. Radial scars, however,
present with lower cellularity and show the presence of
myoepithelial cells within the cell clusters and have FIGURE 5-51
inconspicuous nucleoli. If radial scar is associated with Direct smear of tubular carcinoma showing a uniform population of cells
proliferative breast disease with atypia, the higher with uniform nuclei and no myoepithelial cells.

Ch005-F06731.indd 144 10/26/2006 10:26:00 AM


CHAPTER 5 Breast 145

cellularity, loss of polarity, and some degree of anisonu- are well-circumscribed lesions; on ultrasound, they
cleosis may mimic a neoplastic process. Immunostaining are solid and hypoechoic. Histologically, they present
for myoepithelial cell markers is an effective tool in as pure or mixed type, with two prognostically
making the distinction. different behaviors. Pure forms present with a well-
Similarly, tubular carcinomas may mimic fibro- defined gelatinous mass and show small epithelial
adenomas. However, fibroadenomas often present with islands floating in abundant extracellular mucin. Pure
complex branching of ductal epithelial cells admixed mucinous carcinomas are associated with a favorable
with myoepithelial cells. Tubular carcinomas may occa- prognosis.
sionally lead to false negative diagnosis, particularly if Cytologically, the aspirates contain thick mucinous
the aspirate is not cellular. When in doubt, it is reason- material on smears, and show abundant mucin in the
able to recommend total surgical excision of the background. Clusters of relatively small and uniform
lesion. epithelial cells with bland-appearing cytologic features
are often seen within the mucinous background. Tumor
cells may appear in three-dimensional clusters, mono-
layered sheets, or as dissociated isolated cells. The epi-
MUCINOUS CARCINOMA thelial cells are relatively uniform with wispy cytoplasm
and a vesicular chromatin pattern (Figs 5-52 & 5-53).

Mucinous carcinomas represent approximately 2% of


breast cancers. It is also known as colloid, mucoid,
gelatinous, and mucin-producing carcinoma. These
tumors generally occur in older women. Clinically, they
present as a soft mobile mass. Mammographically, they

MUCINOUS CARCINOMA – DISEASE FACT SHEET

Clinical Features
៉ A rare breast tumor that occurs in an older age group
៉ Clinically, it presents as a soft mobile mass
៉ Breast imaging shows a well-defined lesion
៉ Prognosis in pure mucinous carcinomas is favorable
៉ Mixed variety of mucinous carcinoma is associated with the same
prognosis as an infiltrating duct cell carcinoma

FIGURE 5-52
Direct smear of mucinous carcinoma characterized by abundant mucin
in the background and clusters of epithelial cells.

MUCINOUS CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Variable cellularity
៉ Mucinous background
៉ Cohesive clustering of small aggregates of epithelial cells with
minimal nuclear atypia
៉ Fragments of stroma and newly formed small vessels

Ancillary Studies
៉ Immunostain for myoepithelial cell markers

Differential Diagnosis and Pitfalls


៉ Mucocele-like tumor: occurs in younger women; associated with
proliferative and premalignant breast disease; contains both
epithelial and myoepithelial cell components
៉ Myxoid fibroadenoma: occurs in younger women; demonstrates
stromal and mixed epithelial components; presence of antler-
horn pattern
៉ Signet-ring cell carcinoma: abundance of signet-ring cells with FIGURE 5-53
intracellular mucin production Direct smear of mucinous carcinoma showing the same features in
higher magnification. The epithelial cells are uniform and there are no
myoepithelial cells.

Ch005-F06731.indd 145 10/26/2006 10:26:01 AM


146 FINE NEEDLE ASPIRATION CYTOLOGY

The cellularity varies based on the amount of extra-


cellular mucin. In hypocellular aspirates, the bland
appearance of tumor cells may lead to false negative
diagnosis.
The mixed form of mucinous carcinoma is an infil-
trating duct cell carcinoma with mucinous differentia-
tion. The prognosis of this variant is the same as for the
classical form of infiltrative duct cell carcinoma. In the
mixed form, cellular pleomorphism is more conspicuous
with occasional necrosis and a smaller amount of mucin
in the background. Regardless of the cellularity and the
presence of mucinous material, it is best to classify these
lesions as ‘breast carcinoma with a mucinous compo-
nent’ by FNAB.
Mucinous carcinomas must be distinguished from
‘mucocele-like lesions’ and myxoid fibroadenomas.
Mucocele-like tumors of the breast are benign entities
that are associated with a variety of metaplastic and FIGURE 5-54
hyperplastic conditions. They occur in young and in Direct smear of signet-ring cell carcinoma characterized by isolated and
premenopausal women, in contrast to mucinous carci- clusters of neoplastic epithelial cells with a signet-ring pattern.
noma, which is a tumor of old age. Aside from age, the
aspirates from mucoceles display the presence of
both epithelial and myoepithelial cells, which can be
supported by immunostains for myoepithelial cell
markers. SIGNET-RING CELL CARCINOMA – PATHOLOGIC FEATURES
Another entity which should be differentiated from
mucinous carcinoma is fibroadenoma with myxoid Cytopathologic Findings
stroma. Clues to the correct diagnosis are young age of ៉ Moderate to high cellularity
the patient and the presence of stroma and epithelial/ ៉ Abundance of small to medium-sized cells with crescent-shaped
nuclei and intracellular mucin
myoepithelial components. Immunostain for myoepi-
thelial cell markers will provide the supporting evidence.
Ancillary Studies
Signet-ring cell carcinoma of the breast may also mimic
៉ Special stains
mucinous carcinoma. However, in this tumor, the
៉ Electron microscopy
majority of cells show evidence of intracytoplasmic ៉ Panel of immunostains for organ-specific antibodies
mucin.
Differential Diagnosis and Pitfalls
៉ Metastatic adenoma: pertinent clinical history; review of the
original pathology slides; use of ancillary studies
SIGNET-RING CELL CARCINOMA ៉ Infiltrating lobular carcinoma: presence of other forms of
neoplastic epithelial cells; Indian file pattern
៉ Secretory carcinoma: numerous branching sheets of neoplastic
cells with cytoplasmic vacuoles and extracellular lumina; red
Signet-ring cell carcinomas are rare breast tumors that blood cells, cellular debris with tumor diathesis in the
are associated with an unfavorable prognosis. This background; multiple intracytoplasmic vacuoles
lesion occurs in the mid to late fifties. The origin of this ៉ Lipid-rich carcinoma: moderate cellular yield, loose cohesive cell
as a variant of mucinous carcinoma versus an infi ltrat- arrangement; atypical, indented nuclei with nuclear vacuoles;
ing lobular carcinoma remains controversial. Cytologi- abundant intracytoplasmic lipid material, which can be
cally, signet-ring cell carcinomas present with moderate demonstrated on air-dried smears and by the use of oil res
to rich cellularity. The cells are arranged singly or in O staining

SIGNET-RING CELL CARCINOMA – DISEASE FACT SHEET small loose clusters. The cells are small with crescent-
shaped nuclei compressed to the cell periphery by
Clinical Features mucin (Figs 5-54 & 5-55).
៉ A rare breast tumor with an unfavorable prognosis The differential diagnosis includes metastatic carci-
៉ May mimic other forms of malignancies
noma (especially from gastrointestinal tumors), infil-
៉ Distinction from metastatic tumors is important and requires
trating lobular carcinomas, secretory carcinomas, and
clinical information, review of the original tumor, and ancillary
studies
lipid-secreting carcinomas. The clinical history, review
of the original pathology slides, and use of ancillary
studies such as special stains, electron microscopy, and

Ch005-F06731.indd 146 10/26/2006 10:26:02 AM


CHAPTER 5 Breast 147

FIGURE 5-57
FIGURE 5-55
Direct smear of secretory carcinoma: higher magnification of the same
Direct smear of signet-ring cell carcinoma: higher magnification of the case as in Fig. 5-56, showing multiple vacuoles in the cytoplasm.
same case as in Fig. 5-54, showing signet-ring cells with mucin
droplets.

MEDULLARY CARCINOMA

Medullary carcinomas of the breast are rare tumors,


accounting for 5% to 7% of all breast cancers, and are
associated with a good prognosis. Medullary carcino-
mas occur in patients aged between 21 and 95 years,
with a mean age of 50 years. In young patients, medul-
lary carcinomas may be associated with genetically
mutated cancer in high-risk individuals. Medullary
carcinomas present as a well-defined lesion, both clini-
cally and mammographically.
On histology, typical medullary carcinomas are
characterized by sharp circumscription and a syncytial
growth pattern of highly pleomorphic epithelial cells
with no glandular pattern and surrounded by a lympho-
cytic infiltration. Tumors that exhibit any or few of
these criteria are called atypical medullary carcinomas.
FIGURE 5-56 The behavior and prognosis of these tumors are similar
Direct smear of secretory carcinoma characterized by clusters of to those seen in the usual forum of infiltrating duct cell
neoplastic epithelial cells with abundant vacuolated cytoplasm. carcinoma. Therefore, the designation of ‘ductal carci-
noma with medullary features’ is more appropriate for
cytologic diagnosis of medullary carcinoma.
a panel of immunostains for organ-specific antibodies Cytologically, the aspirates from a medullary carci-
are the tools to differentiate between a signet-ring cell noma are hypercellular with a background showing
carcinoma and a metastatic tumor. lymphocytes and plasma cells. The neoplastic epithelial
The distinction between signet-ring cell carcinomas cells show an extreme degree of pleomorphism,
and other tumors requires special attention to subtle
morphologic differences. Infiltrating lobular carcinomas
often show other characteristics of this tumor, including
the presence of Indian file pattern. Secretory carcino- MEDULLARY CARCINOMA – DISEASE FACT SHEET
mas present with several prominent intracytoplasmic
Clinical Features
vacuoles (Figs 5-56 & 5-57). Lipid-rich carcinomas
៉ A rare breast tumor that occurs in all age groups
contain small cytoplasmic vacuoles that present in dif-
៉ In young patients, may be associated with breast cancer genes
ferent forms. The vacuoles are often seen in the peri-
៉ A well-circumscribed lesion, both clinically and mammographically
nuclear area, and nuclei are indented. Special stain ៉ Associated with a good prognosis
demonstrates the presence of fat droplets in the cyto-
plasm of tumor cells.

Ch005-F06731.indd 147 10/26/2006 10:26:02 AM


148 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 5-58
FIGURE 5-59
Direct smear of medullary carcinoma characterized by clusters of
epithelial cells surrounded by lymphocytes. Direct smear of medullary carcinoma showing syncytial growth
pattern.

APOCRINE CARCINOMA
MEDULLARY CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings Apocrine carcinoma, a rare breast tumor, occurs in an


៉ High cell yield older age group. The apocrine features and biomarker
៉ Pleomorphic cell population forming syncytial growth pattern findings may suggest a difference in steroid metabo-
៉ Pronounced lymphoplasmacytic infiltration lism, particularly a higher frequency of androgen recep-
៉ Necrotic background tor positivity compared to the ductal carcinoma not
otherwise specified. It has been suggested that patients
Ancillary Studies
៉ Immunophenotyping by immunocytochemistry and/or fl ow
cytometry

Differential Diagnosis and Pitfalls APOCRINE CARCINOMA – DISEASE FACT SHEET


៉ Malignant lymphoma: monotonous cell population; absence
of epithelial cells; evidence of monoclonality by Clinical Features
immunocytochemistry or flow cytometry ៉ A rare breast tumor that occurs in an older age group
៉ Infiltrating duct cell carcinoma with lymphocytic pattern: ៉ May have a different pathway of steroid metabolism
absence of syncytial growth pattern; lesser degree of
anisonucleosis and pleomorphism

APOCRINE CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
anisonucleosis, a high mitotic rate and arrangement in ៉ Cellular aspirate
syncytial sheets (Figs 5-58 & 5-59). Medullary carcino- ៉ Pleomorphic population of cells with abundant granular
mas rarely present as a cystic lesion. They are frequently cytoplasm
hormone receptor-negative and have an abnormal DNA ៉ Marked anisonucleosis with large nuclei and prominent nucleoli
pattern.
The differential diagnosis includes poorly differenti- Differential Diagnosis and Pitfalls
ated infiltrating duct cell carcinoma with lymphocytic ៉ Hyperplastic/metaplastic apocrine cells: presence of
infiltrate and malignant lymphoma. Ductal cell carci- accompanying proliferating epithelial cells; lack of significant
noma with lymphocytic pattern does not present with anisonucleosis and hyperchromasia
៉ Secretory carcinoma: neoplastic cells with prominent
a syncytial growth pattern. Malignant lymphomas are
intracellular spaces; cytoplasmic vacuoles; signet-ring formation
characterized by a monotonous cell population with no
៉ Lipid-rich carcinoma: abundant multivacuolated cells with a
evidence of epithelial differentiation. Immunopheno- more uniform nuclear appearance; lack of multiple nucleoli
typing by immunocytochemistry or flow cytometry will
aid in making the differentiation.

Ch005-F06731.indd 148 10/26/2006 10:26:03 AM


CHAPTER 5 Breast 149

INFILTRATING LOBULAR CARCINOMA

Infi ltrating lobular carcinoma accounts for approxi-


mately 6.3% of primary breast carcinomas. It occurs in
wide range of age groups, ranging from 26 to 86 years
old. Clinical presentation is similar to that of other
primary breast carcinomas. However, it is not associ-
ated with Paget’s disease and occasionally it may present
as an indurated area without any discrete mass. Infi l-
trating lobular carcinomas have a different pattern of
metastatic presentations and tend to involve skeletal,
visceral, serosal, and meningeal areas. Similarly, ovary,
bone, and uterus are the common sites of metastasis
from this tumor.
FIGURE 5-60
Mammographically, it can present as an asymmetric
density with no clearly delineated margin with no or
Direct smear of apocrine carcinoma characterized by a population of
neoplastic epithelial cells with granular cytoplasm and conspicuous little architectural distortion. Multifocal infiltrating
nucleoli. lobular carcinoma may present with minimal distor-
tion with no significant mass or increased density or
microcalcification. These subtle mammographic fea-
tures warrant careful examination of the breast and
sampling of any suspicious area. Infiltrating lobular
carcinoma is often bilateral and shows evidence of
multiplicity.
On histology, infiltrating lobular carcinomas are
characterized by diffuse infiltration of mammary stroma

INFILTRATING LOBULAR CARCINOMA – DISEASE FACT SHEET

Clinical Features
៉ Variable clinical and mammographic presentations
៉ Associated with bilaterality and multiplicity
៉ Has a different pattern of metastasis from tumors of ductal origin

FIGURE 5-61 INFILTRATING LOBULAR CARCINOMA – PATHOLOGIC FEATURES


Direct smear of apocrine carcinoma showing multinucleation and
prominent nucleoli. Cytopathologic Findings
៉ Variable cellularity
៉ Monomorphic cell population of small cells with insignificant
atypia
with apocrine carcinoma may experience a longer sur- ៉ Eccentrically located nuclei
vival. Cytologically, the aspirates are often cellular and ៉ Cells with intracytoplasmic lumina
show pleomorphic tumor cells with abundant granular ៉ Indian file pattern
cytoplasm. The nuclei are large, vesicular and are cen- ៉ No myoepithelial cells
trally or eccentrically located. The nucleoli are promi-
nent and multinucleation is common (Figs 5-60 & Ancillary Studies
5-61). ៉ Immunostain for myoepithelial cell markers and E-cadherin
Apocrine carcinomas should be distinguished from
atypical changes seen in proliferative and metaplastic Differential Diagnosis and Pitfalls
apocrine cells. The clues are the presence of an accom- ៉ Fibrocystic change: presence of polymorphic cell pattern;
panying polymorphous cell population commonly seen presence of myoepithelial cells
៉ Low nuclear grade ductal carcinoma: positive immunostain for
in proliferative breast disease. Absence of anisonucleo-
E-cadherin
sis and significant hyperchromasia are also other fea-
៉ Atypical lobular hyperplasia and lobular carcinoma in situ: less
tures seen in atypical apocrine lesions. Other lesions cellularity; more cohesive cell pattern
included in the differential diagnosis are lipid-rich and
secretory carcinomas.

Ch005-F06731.indd 149 10/26/2006 10:26:04 AM


150 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 5-62 FIGURE 5-64


Direct smear of an infiltrating lobular carcinoma characterized by Direct smear of an infiltrating lobular cell carcinoma exhibiting an
isolated and a cluster of epithelial cells with uniform nuclei and Indian file pattern.
intracytoplasmic lumina.

FIGURE 5-65
FIGURE 5-63 Direct smear of an infiltrating lobular carcinoma, pleomorphic variant,
Direct smear of an infiltrating lobular cell carcinoma showing clusters characterized by a cellular aspirate showing an isolated population
of epithelial cells with no myoepithelial cells. of epithelial cells with eccentric nuclei, anisonucleosis. and
hyperchromasia.

and ductal structures with neoplastic cells in a pagetoid


growth pattern. An area of carcinoma in situ is com- insignificant atypia associated with this entity. The
monly seen. Infiltrating lobular carcinoma may present presence of a monomorphic pattern of small cells
in a variety of patterns with similar prognostic behav- arranged in single cells, cords, or clusters with no
ior, except for the pleomorphic variant, which is associ- recognizable myoepithelial cells is the key feature to
ated with a more unfavorable outcome. differentiate infiltrating lobular carcinoma from benign
Cytologically, infiltrating lobular carcinomas present breast lesions.
with variable cellularity. The cells are small and uniform Differentiation between infiltrating lobular carci-
with small nucleoli, and present as cell balls or with noma and other entities may be difficult. With a hypo-
Indian file appearance. Tumor cells present with eccen- cellular specimen, it is best to consider surgical excision
tric nuclei, occasional intracytoplasmic lumina with for further characterization of suspicious cells for lobular
mucin droplets, and rare signet-ring cells (Figs 5-62– carcinoma. There are overlapping features between
5-64). The pleomorphic variant features more pleomor- atypical lobular hyperplasia, lobular carcinoma in situ,
phism and nuclear atypia (Figs 5-65 & 5-66). Infiltrating and an infiltrating lobular carcinoma. These entities are
lobular carcinomas are one of the main reasons for false collectively called lobular neoplasia if the distinction is
negative diagnosis in breast FNAB. This is often due to not possible. Higher cellularity and a higher proportion

Ch005-F06731.indd 150 10/26/2006 10:26:04 AM


CHAPTER 5 Breast 151

FIGURE 5-66
FIGURE 5-68
Direct smear of an infiltrating duct cell carcinoma, pleomorphic variant,
showing intracytoplasmic lumina with mucin droplets. Direct smear of lobular carcinoma in situ showing acini and clustering
of uniform-appearing epithelial cells.

FIGURE 5-67
Direct smear of lobular carcinoma in situ characterized by clusters of
small uniform-appearing epithelial cells with no myoepithelial cells.

of dissociated cells are commonly seen in an infiltrating


lobular carcinoma (Figs 5-67–5-70). The differential FIGURE 5-69
diagnosis also includes low nuclear grade carcinoma of Direct smear of atypical lobular hyperplasia characterized by prolifera-
ductal origin. Immunostain for E-cadherin can differen- tion of small cells simulating acini.
tiate between these two entities.

METASTASES TO THE BREAST


METASTASES TO THE BREAST – DISEASE FACT SHEET

The breast is not a common site for metastases from Clinical Features
other primary malignancies. The distinction between ៉ Rare
a primary breast carcinoma and a metastasis is criti- ៉ Clinical and mammographic features may be similar to those of
cally important because of different therapeutic impli- primary breast carcinoma
៉ Cytologic distinction is important for the management and
cations and the differences in outcome. Clinically, the
clinical outcome
presentation of the two entities may be similar; however,
៉ Clinical history, review of the original pathology slides, and
nipple discharge and retraction are not seen in meta- ancillary studies are essential to make an accurate diagnosis
static tumors. Metastatic tumors have a variety of
mammographic features, ranging from features similar

Ch005-F06731.indd 151 10/26/2006 10:26:05 AM


152 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 5-70 FIGURE 5-71


Direct smear of atypical lobular hyperplasia: higher magnification of Direct smear of metastatic squamous cell carcinoma showing isolated
the same case as shown in Fig. 5-69. and clusters of neoplastic epithelial cells with evidence of
keratinization.

METASTASES TO THE BREAST – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ High cellularity
៉ Dispersed cell pattern of various types and shapes, dissimilar to
the classic types of breast cancer
៉ Variable cytomorphology similar to the primary site of origin

Ancillary Studies
៉ Special stains
៉ Electron microscopy
៉ Panel of immunostains
៉ Flow cytometry

Differential Diagnosis and Pitfalls


៉ Primary breast carcinoma: clinical history; classic pattern of
cytomorphology for primary breast cancer
FIGURE 5-72
Direct smear of mucin-producing metastatic adenocarcinoma character-
ized by isolated epithelial cells with centrally located nuclei and
to those of proliferative breast disease, cyst, or fibroad- abundant cytoplasm and intracellular mucin.
enoma to those of a malignant lesion such as medullary
carcinoma. Microcalcification is not frequent except in
rare cases of psammoma bodies in metastatic tumors Cytologically, metastatic tumors present with rich
from the ovary. Patients with metastatic breast lesions cellularity. Based on the site of the origin, the aspirates
experience a poor outcome and about 80% of them die present with various cell types, sizes, and cellular
within a year. arrangements. Malignant melanomas, squamous cell
The differentiation between a primary breast tumor carcinoma, various adenocarcinomas, Hodgkin disease,
and a metastasis may not be always possible. The most large cell lymphoma, and pleomorphic sarcomas exhibit
important step is the recognition of a cytologic pattern pleomorphic large cells. Melanomas, lymphomas, leuke-
not commonly seen in a primary breast carcinoma. This mias, and poorly differentiated carcinomas present with
should alert the pathologist to explore the possibility of a dispersed cell pattern. Malignant melanoma cells
a metastasis. Review of the clinical history and the may contain pigment, intranuclear inclusions and
pathology slides of the primary tumor associated with binucleation (Figs 5-71–5-78).
special stains and often ancillary studies such as Neuroendocrine tumors and small cell carcinoma of
EM and/or organ-specific immunohistochemical stains the lung often present with small-sized tumor cells.
usually resolves the quandary. This process can be com- Spindle cell tumors often represent various types of
pleted by the use of special stains and ancillary studies sarcomas. The most common metastatic tumors to the
such as electron microscopy and of immunostaining for breast are lung and malignant melanoma. Metastasis
a panel of specific antibodies. from other sites have also been reported. In men,

Ch005-F06731.indd 152 10/26/2006 10:26:06 AM


CHAPTER 5 Breast 153

FIGURE 5-73 FIGURE 5-74


Direct smear of non-Hodgkin lymphoma characterized by dispersed cell Direct smear of Hodgkin lymphoma characterized by atypical lymphoid
pattern and various-sized malignant lymphoid cells. elements and classic Reed-Sternberg cell.

FIGURE 5-75 FIGURE 5-76


Direct smear of malignant melanoma: dispersed cell pattern with Immunostained cell block showing expression of HMB-45 (same case as
binucleation and intracytoplasmic vacuoles. Fig. 5-75).

FIGURE 5-77 FIGURE 5-78


Direct smear of metastatic prostate carcinoma characterized by uniform- Metastatic prostate carcinoma: immunostained cell block showing
appearing epithelial cells. expression of prostate-specific antigen.

Ch005-F06731.indd 153 10/26/2006 10:26:07 AM


154 FINE NEEDLE ASPIRATION CYTOLOGY

prostate cancer is the most common form of metastatic


tumor to the breast. Lymphoma including Hodgkin
disease of the breast is commonly seen as a manifesta-
tion of a known generalized lymphoma and presents no
difficulty in rendering a diagnosis. However, rarely they
may occur as the primary breast carcinoma. Immuno-
phenotyping with immunocytochemistry or flow cyto-
metry can establish the diagnosis.
Sarcomas can also occur as a primary breast carci-
noma or as metastasis from other sites. The aspirate of
sarcomas is cellular with a spindle cell pattern. Electron
microscopy and immunostaining for mesenchymal
markers are helpful in establishing the diagnosis of a
sarcomatous lesion.

MISCELLANEOUS CONDITIONS FIGURE 5-79


Direct smear of granular cell tumor characterized by uniform-appearing
tumor cells with conspicuous granular cytoplasm.

GRANULAR CELL TUMOR

Granular cell tumors are examples of stromal tumors


of the breast, which are rare and present as a palpable
mass. These tumors may resemble breast carcinomas,
both clinically and mammographically, and FNAB is
quite helpful to provide an accurate diagnosis.
Cytologically, the aspirates show moderately cellular
smears with isolated and clusters of cells with granular
and foamy cytoplasm. The nuclei are uniform and the
chromatin is fine. Immunostain for S-100 and electron
microscopy are often used to substantiate the diagnosis
of granular cell tumor (Figs 5-79 & 5-80). Malignant

GRANULAR CELL TUMOR – DISEASE FACT SHEET FIGURE 5-80


Granular cell tumor: immunostained cell block showing expression
Clinical Features of S-100 protein.
៉ Rare form of stromal tumors of the breast
៉ Clinically and mammographically, may mimic primary breast
carcinoma granular cell tumors show extensive pleomorphism
with a high degree of mitotic activity.

GRANULAR CELL TUMOR – PATHOLOGIC FEATURES


GYNECOMASTIA

Cytopathologic Findings
Gynecomastia refers to unilateral or bilateral enlarge-
៉ Variable cellularity ment of the male breast. This condition may be due
៉ Isolated and clusters of cells with uniform nuclei, conspicuous
to hormonal changes, testicular or hepatic tumors,
nucleoli, and fine chromatin pattern
៉ Tumor cells with granular cytoplasm
Klinefelter’s syndrome, drug therapy, liver or renal
disease.
Differential Diagnosis and Pitfalls Cytologically, gynecomastias present with a cellular
៉ Malignant granular cell tumor: pronounced cellular
aspirate that shows tightly cohesive groups of epithelial
pleomorphism; abundant mitosis cells and stromal components, features seen in fibro-
adenomas in the female breast. Apocrine metaplasia,
histiocytes, fragments with cribriform or micropapillary

Ch005-F06731.indd 154 10/26/2006 10:26:09 AM


CHAPTER 5 Breast 155

FIGURE 5-82
FIGURE 5-81
Direct smear of gynecomastia: a cluster of epithelial/myoepithelial cells
Direct smear of gynecomastia characterized by uniform-appearing
with an architectural pattern similar to a fibroadenoma.
epithelial/myoepithelial cells with stromal fragments.

lung cancer, prostrate cancer, and lymphoma being


GYNECOMASTIA – DISEASE FACT SHEET
common metastatic tumors.

Clinical Features
៉ Most frequent breast lesion in men SUGGESTED READINGS
៉ Commonly caused by hormone-induced conditions Mastitis, Abscess, and Fat Necrosis
៉ FNAB is most effective in establishment of diagnosis
Al Nazer MA. Idiopathic granulomatous lobular mastitis. A forgotten clini-
cal diagnosis. Saudi Med J 2003;24:1377–1380.
Asoglu O, Ozmen V, Karanlik H, et al. Feasibility of surgical management
in patients with granulomatous mastitis. Breast J 2005;11:108–114.
Das DK, Sodhani P, Kashyap V, Parkash S, Pant JN, Bhatnagar P. Inflam-
matory lesions of the breast: diagnosis by fine needle aspiration.
GYNECOMASTIA – PATHOLOGIC FEATURES Cytopathology 1992;3:281–289.
Galblum LI, Oertel YC. Subareolar abscess of the breast: diagnosis by fine
needle aspiration. Am J Clin Pathol 1983;80:496–499.
Cytopathologic Findings
Karstrup S, Solvig J, Nolsoe CP, et al. Acute puerperal breast abscesses:
៉ Variable cellularity US-guided drainage. Radiology 1993;188:807–809.
៉ Uniform population of small groups of epithelial/myoepithelial Khoda J, Lantsberg L, Yegev Y, Sebbag G. Management of periareolar
cells abscess and mamillary fistula. Surg Gynecol Obstet 1992;175:306–308.
៉ Tall columnar cells Macansh S, Greenberg M, Barraclough B, Pacey F. Fine needle aspiration
៉ Stromal fragments cytology of granulomatous mastitis. Report of a case and review of the
literature [see comments]. Acta Cytol 1990;34:38–42.
Maier WP, Au FC, Tang CK. Nonlactational breast infection. Am Surg
Differential Diagnosis and Pitfalls 1994;60:247–250.
៉ Primary and metastatic breast cancer: obvious evidence of Maygarden SJ, Novotny DB, Johnson DE, Frable WJ. Subclassification of
morphologic features of malignancy; history of other primary benign breast disease by fine needle aspiration cytology. Comparison
carcinomas of cytologic and histologic findings in 265 palpable breast masses. Acta
Cytol 1994;38:115–129.
Meguid MM, Oler A, Numann PJ, Khan S. Pathogenesis-based treatment
of recurring subareolar breast abscesses. Surgery 1995;118:775–782.
Tse GM, Poon CS, Ramachandram K, et al. Granulomatous mastitis: a
clinicopathological review of 26 cases. Pathology 2004;36:254–257.
arrangements, and cystic changes may be associated Versluijs-Ossewaarde FN, Roumen RM, Goris RJ. Subareolar breast abscesses:
characteristics and results of surgical treatment. Breast J 2005;11:
with gynecomastia (Figs 5-81 & 5-82). Occasionally, the 179–182.
cellular patterns, presence of small groups and isolated Weiss RL, Matsen JM. Group B streptococcal breast abscess. Arch Pathol
cells, and columnar cell differentiation may cause diag- Lab Med 1987;111:74–75.
nostic difficulty. However, a diagnosis of malignancy Zuka JJ, Crite G Jr, Aryes WW. Fistulas of lactiferous ducts. Am J Surg
1951;81:312–317.
should be considered only if the aspirate presents
obvious features of malignancy.
Primary breast cancers in men are similar to those in Treatment-Induced Changes
women. Intracystic papillary carcinomas are frequent in Boyages J, Bilous M, Barraclough B, Langlands AO. Fat necrosis of the
men. In addition, there is relatively equal frequency of breast following lumpectomy and radiation therapy for early breast
primary and metastatic lesions in the male breast, with cancer. Radiother Oncol 1988;13:69–74.

Ch005-F06731.indd 155 10/26/2006 10:26:10 AM


156 FINE NEEDLE ASPIRATION CYTOLOGY

Costa MJ, Stewart G, Perez EA, Goodnight JE. Fine-needle aspiration Walters TK, Zuckerman J, Nisbet Smith A, Hudson E, Chia Y, Burke M. Fine
cytology in locally advanced breast adenocarcinoma: a case with needle aspiration biopsy in the diagnosis and management of fibro-
complete response to preoperative chemotherapy in association with adenoma of the breast. Br J Surg 1990;77:1215–1217.
granulomatous inflammatory reaction. Diagn Cytopathol 1994;10:357–
361.
Dey P, Karmakar T. Fine needle aspiration cytology in operative Papillary Breast Lesions
scar nodules in breast carcinoma [letter]. Acta Cytol 1995;39:137–
138. Al-Kaisi N. The spectrum of the ‘gray zone’ in breast cytology. A review
Filomena CA, Jordan AG, Ehya H. Needle aspiration cytology of the irradi- of 186 cases of atypical and suspicious cytology. Acta Cytol
ated breast. Diagn Cytopathol 1992;8:327–332. 1994;38:898–908.
Ku NN, Mela NJ, Fiorica JV, et al. Role of fine needle aspiration cytology Jeffrey PB, Ljung BM. Benign and malignant papillary lesions of the
after lumpectomy. Acta Cytol 1994;39:927–932. breast. A cytomorphologic study. Am J Clin Pathol 1994;101:500–
Malberger E, Edoute Y, Toledano O, Sapir D. Fine needle aspiration and 507.
cytologic findings of surgical scar lesions in women with breast cancer. Kline TS. Masquerades of malignancy: a review of 4241 aspirations from
Cancer 1992;69:148–152. the breast. Acta Cytol 1981;25:263–266.
Maygarden SJ, Novotny DB, Johnson DE, Powers CN, Frable WJ. Fine needle Koss LG, Woyke S, Olszewski W. Aspiration Biopsy: Cytologic Interpretation
aspiration cytology of suture granulomas of the breast: a potential and Histologic Bases. New York: Igaku-Shoin, 1984.
pitfall in the cytologic diagnosis of recurrent breast cancer. Diagn Levine PH, Waisman J, Yang GC. Aspiration cytology of cystic carcinoma
Cytopathol 1994;19:175–179. of the breast. Diagn Cytopathol 2003;28:39–44.
Pedio G, Landolt U, Zobeli L. Irradiated benign cells of the breast: a Masood S, Loya A, Khalbuss W. Is core needle biopsy superior to fine
potential diagnostic pitfall in fine needle aspiration cytology [letter]. needle aspiration breast biopsy in papillary breast lesions? Diagn
Acta Cytol 1988;32:127–128. Cytopathol 2003;28:329–334.
Solin LJ, Fowble BL, Schultz DJ, Rubenstien JR, Goodman RL. The detec- Saphir O, Parker ML. Intracystic papilloma of the breast. Am J Pathol
tion of local recurrence after definitive irradiation for early stage car- 1940;16:189–210.
cinoma of the breast. An analysis of the results of breast biopsies
performed in previously irradiated breasts. Cancer 1990;65:2497–
2502. Fibrocystic Change
[Anonymous]. Final version: the uniform approach to breast fine needle
aspiration biopsy. Breast J 1997;3:149–168.
Hormonal Changes: Secretory, Pregnancy, and Lactational Changes Bibbo M, Scheiber M, Cajulius R, Keebler CM, Wied GL, Dowlatshahi K.
Bottles K, Taylor RN. Diagnosis of breast masses in pregnant and lactating Sterotaxic fine needle aspiration cytology of occult malignant and pre-
women by aspiration cytology. Obstet Gynecol 1985;66(3 Suppl): malignant breast lesions. Acta Cytol 1988;32:193–201.
76S–78S. Dizura BR, Bonfiglio TA. Needle cytology of the breast. Acta Cytol
Gupta RK, McHutchinson AGR, Dowle CS, et al. Fine needle aspiration 1979;23:332–340.
cytodiagnosis of breast masses in pregnant and lactating women and Dupont WD, Page DL. Risk factors for breast cancer in women with prolif-
its impact on management. Diagn Cytopathol 1993;9:156–159. erative breast disease. N Engl J Med 1985;312:146–151.
Hughes LE. Classification of benign breast disorders. The ANDI classifica- Fabian C, Kimler B, Brady D, et al. Phase II chemoprevention trial of
tion based on physiological processes within the normal breast. Br Med DFMO using the random FNA model. Breast Cancer Res Treat 2000;
Bull 1991;47:251–257. 64:48.
Hughes LE, Mansel RE, Webster DJ. Aberrations of normal development Fabian CJ, Kimler BF, Brady D, et al. A phase II breast cancer chemo-
and involution (ANDI): a new perspective on pathogenesis and nomen- prevention trial of oral α-difluoromethylornithine. Clin Cancer Res
clature of benign breast disorders. Lancet 1987;2:1316–1319. 2002;8:3105–3117.
Hutson SW, Cowen PN, Bird CC. Morphologic studies of age related changes Fabian CJ, Kimler BF, Zalles CM, et al. Short term breast cancer prediction
in normal human breast and their significance to evolution of mammary by random periareolar fine needle aspiration cytology and the Gail risk
cancer. J Clin Pathol 1985;38:281–287. model. J Natl Cancer Inst 2000;92:1217–1227.
Novotny DB, Maygarden SJ, Shermer RW, Frable WJ. Fine needle aspiration Khan SA, Masood S, Miller L, Numann PJ. Random fine needle aspiration
of benign and malignant breast masses associated with pregnancy. Acta of the breast of women at increased risk and standard risk controls.
Cytol 1991;35:676–686. Breast J 1998;4:420–425.
Lim JC, Al-Masri H, Salhadar A, Xie HB, Gabram S, Wojcik EM. The signifi-
cance of the diagnosis of atypia in breast fine-needle aspiration. Diagn
Fibroadenomas Cytopathol 2004;31:285–288.
Marshall CJ, Schumann GB, Ward JH, Riding JM, Cannon-Albright L,
Benoit JL, Kara R, McGregor SE, Duggan MA. Fibroadenoma of the breast: Skolnik M. Cytologic identification of clinically occult proliferative
diagnostic pitfalls of fine needle aspiration. Diagn Cytopathol 1992; breast disease in women with a family history of breast cancer. Am J
8:643–647. Clin Pathol 1991;95:157–165.
Bottles K, Chan JS, Holly EA, Chiu SH, Miller TR. Cytologic criteria for Masood S. Cytopathology of the Breast. Chicago: ASCP Press, 1995.
fibroadenoma. A step-wise logistic regression analysis. Am J Clin Pathol Masood S. Diagnostic terminology in breast fine needle aspiration biopsy:
1988;89:707–713. redefining the term atypia. Cancer (Cancer Cytopathol) 1999;87:1–4.
Dejmek A, Lindholm K. Frequency of cytologic features in fine needle Masood S. Expanding role of breast cytopathology as a risk predictor. Adv
aspirates from histologically and cytologically diagnosed fibroadeno- Anat Pathol 2001;8:255–263.
mas. Acta Cytol 1991;35:695–699. Masood S, Frykberg ER, McClellan GL, Scalapino MC, Mitchum DG, Bullard
Dusenberry D, Frable WJ. Fine needle aspiration cytology of phyllodes JB. Prospective evaluation of radiologically detected fine needle aspira-
tumor: potential diagnostic pitfalls. Acta Cytol 1992;36:215–221. tion biopsy of nonpalpable breast lesions. Cancer 1990;66:1480–
Rogers LA, Lee KR. Breast carcinoma simulating fibroadenoma or fibro- 1487.
cystic change by fine needle aspiration. A study of 16 cases. Am J Clin Masood S, Frykberg ER, McLellan GL, et al. Cytologic differentiation
Pathol 1992;98:155–160. between proliferative and nonproliferative breast disease in mammo-
Simi U, Moretti D, Iacconi P, et al. Fine needle aspiration cytopathology graphically guided fine needle aspirates. Diagn Cytopathol 1991;7:581–
of phyllodes tumor. Differential diagnosis with fibroadenoma [see com- 590.
ments]. Acta Cytol 1988;32:63–66. Masood S, Hardy NM, Assaf N. The value of muscle specific actin immuno-
Stanley MW, Tani EM, Rutquist LE, et al. Cystosarcoma phyllodes of the staining in differentiation between atypical hyperplasia and carcinoma
breast: a cytologic and clinicopathologic study of 23 cases. Diagn in breast fine needle aspirates [abstract]. Acta Cytol 1994;38:
Cytopathol 1989;5:29–34. 861.
Stanley MW, Tani EM, Skoog L. Fine needle aspiration of fibroadenomas Masood S, Hardy NM, Assof-Munasifi N. Cytologic grading system in diag-
of the breast with atypia: a spectrum including cases that cytologically nosis of high-risk and malignant breast lesions: merits and pitfalls.
mimic carcinoma. Diagn Cytopathol 1990;6:375–382. Acta Cytol 1994;38:797–798.

Ch005-F06731.indd 156 10/26/2006 10:26:11 AM


CHAPTER 5 Breast 157

Masood S, Lu L, Assaf Munasifi N, McCaulley K. Application of immuno- Fisher ER, Gregorio R, Redmond C, Fisher B. Tubulolobular invasive breast
staining for muscle specific actin for detection of myoepithelial cells in cancer: a variant of lobular invasive cancer. Hum Pathol 1977;8:679–
breast fine needle aspirates. Diagn Cytopathol 1995;13: 683.
71–74. Frierson JF Jr, Iezzoni J, Covell JL. Stereotaxic fine needle aspiration
Masood S, Rasty G. Potential value of cytology in the detection of breast cytology of radial scars [abstract]. Acta Cytol 1993;37:814.
cancer precursors by fine needle aspiration biopsy: ‘The future Pap Gupta RK, Dowle C. Fine needle aspiration cytology of tubular carcinoma
smear for breast cancer.’ Acta Cytol 1999;43:890. of the breast in a young woman. Diagn Cytopathol 1991;7:72–74.
Shiels LA, Mulford D, Dawson AG. Cytomorphology of proliferative breast Silverman JF. Breast. In: Bibbo M, ed. Comprehensive Cytopathology.
disease [abstract]. Acta Cytol 1993;37:768. Philadelphia: WB Saunders, 1991:703–770.
Sidawy M, Stoler M, Frable W, et al. Interobserver variability in the
classification of proliferative breast lesions by fine-needle aspiration:
results of the Papanicolaou Society of Cytopathology Study. Diagn Mucinous Carcinoma
Cytopathol 1998;18:150–165.
Silverman J, Masood S, Ducatman BS, et al. Can FNA biopsy separate Duane GB, Kanter MH, Branigan T, Chang C. A morphologic and morpho-
atypical hyperplasia, carcinoma in situ and invasive carcinoma of the metric study of cells from colloid carcinoma of the breast obtained by
breast? Cytomorphic criteria and limitations in diagnosis. Diagn Cyto- fine needle aspiration. Distinction from other breast lesions. Acta Cytol
pathol 1993; 9:713–728. 1987;31:742–750.
Sneige N, Staerkel GA. Fine needle aspiration cytology of ductal hyper- Fisher CJ, Millis RR. A mucocele-like tumour of the breast associated with
plasia with and without atypia and ductal carcinoma in situ. Hum Pathol both atypical ductal hyperplasia and mucoid carcinoma. Histopathology
1994;25:485–492. 1992;21:69–71.
Stanley MA, Henry-Stanley MJ, Zera R. Prospective study of high-risk Ro JY, Sneige N, Sahin AA, Silva EG, Del Junco GW, Ayala AG. Mucocele
proliferative lesions of breast duct epithelium by fine needle aspiration like tumor of the breast associated with atypical ductal hyperplasia or
[abstract]. Acta Cytol 1991;35:611. mucinous carcinoma. A clinicopathologic study of seven cases. Arch
Pathol Lab Med 1991;115:137–140.
Stanley MW, Tani EM, Skoog L. Mucinous breast carcinoma and mixed
Ductal Carcinoma In Situ mucinous-infiltrating ductal carcinoma: a comparative cytologic study.
Diagn Cytopathol 1989;5:134–138.
Frable WJ. Fine needle aspiration biopsy: a review. Hum Pathol 1983;14: Weaver MG, Abdul Karim FW, al Kaisi N. Mucinous lesions of the breast.
9–28. A pathological continuum. Pathol Res Pract 1993;189(8):873–876.
Frykberg ER, Masood S, Copeland EM, et al. Ductal carcinoma in situ of
the breast. Surg Gynecol Obstet 1993;177:425–440.
Klijanienko J, Katsahian S, Vielh P, Masood S. Stromal infiltration as a Signet-Ring Cell Carcinoma
predictor of tumor invasion in breast fine needle aspiration biopsy.
Diagn Ctyopathol 2004;30:182–186. Craig JP. Secretory carcinoma of the breast in an adult. Correlation of
Lagios MD, Westdahl PR, Margopin FR, et al. Duct carcinoma in situ. aspiration cytology and histology on the biopsy specimen. Acta Cytol
Relationship of extent of noninvasive disease to the frequency of occult 1985;29:589–592.
invasion, multiplicity, lymph node metastasis and short-term treatment Fisher ER, Gregoriao R, Kim WS, et al. Lipid invasive cancer of the breast.
failures. Cancer 1982;50:1309–1314. Am J Clin Pathol 1977;68:558–561.
Lilleng R, Hagmar B. The comedo subtype of intraductal carcinoma. Cyto- Insabato L, Russo R, Cascone AM, et al. Fine needle aspiration cytology
logic characteristics. Acta Cytol 1992;36:345–352. of lipid-secreting breast carcinoma. A case report. Acta Cytol
Malamud YR, Ducatman BS, Wang HH. Comparative features of comedo 1993;37:752–755.
and noncomedo ductal carcinoma in situ of the breast on fine needle Merino MY, Livolsi VA. Signet ring carcinoma of the female breast. Cancer
aspiration biopsy. Diagn Cytopathol 1991;8:571–576. 1981;48:1830–1837.
Wang HH, Ducatman BS, Eick D. Comparative features of ductal carcinoma Nguyen GK, Neifer R. Aspiration biopsy cytology of secretory carcinoma
in situ and infiltrating ductal carcinoma of the breast on fine needle of the breast. Diagn Cytopathol 1987;3:234–237.
aspiration biopsy. Am J Clin Pathol 1989;92:736–740. Ramos CV, Taylor HB. Lipid-rich carcinoma of the breast. A clinicopatho-
logic analysis of 13 examples. Cancer 1974;33:812–819.
Shinagawa T, Tadokoro M, Kitamura H, et al. Secretory carcinoma of the
Primary Breast Carcinoma breast: correlation of aspiration cytology and histology. Acta Cytol
1994;38:909–914.
Abendroth CS, Wang HH, Ducatman BS. Comparative features of carcinoma Shinagawa T, Tadokoro M, Takeuchi E, et al. Aspiration biopsy cytology
in situ and atypical ductal hyperplasia of the breast on fine needle of secretory carcinoma of the breast. A case report. Acta Cytol 1992;36:
aspiration biopsy. Am J Clin Pathol 1991;96:654–659. 189–193.
Ductaman BS, Emery ST, Wang HH. Correlation of histologic grade of
breast carcinoma with cytologic features on fine needle aspiration of
the breast. Mod Pathol 1993;6:539–543. Medullary Carcinoma
Patel JJ, Gartel PC, Smallwood JA, et al. Fine needle aspiration cytology
of breast masses: an evaluation of its accuracy and reasons for diag- Howell LP, Kline TS. Medullary carcinoma of the breast. A rare cytologic
nostic failure. Ann R Coll Surg Engl 1987;69:156–159. finding in cyst fluid aspirate. Cancer 1990;65:277–282.
Meyer JE, Amin E, Lindfors KK, Lipman JC, Stomper PC, Genest D. Medullary
carcinoma of the breast: mammographic and US appearance. Radiology
Tubular Carcinoma 1989;179(1 Pt 1):79–82.
Moore OSJ, Foote FWJ. The relatively favourable prognosis of medullary
Bondeson L, Lindholm K. Aspiration cytology of tubular breast carcinoma. carcinoma of the breast. Cancer 1949;2:635–642.
Acta Cytol 1990;34:15–20. Pedersen L, Schiodt T, Holck S, Zedeler K. The prognostic importance of
Cleary PMG, Mayze JD. Recognition of tubular breast carcinoma on fine syncytial growth pattern in medullary carcinoma of the breast. APMIS
needle aspiration cytology [abstract]. Acta Cytol 1992;36:773. 1990;98:921–926.
de la Torre M, Lindholm K, Lindgren A. Fine needle aspiration cytology Pedersen L, Zedeler K, Holck S, Schiodt T, Mouridsen HT. Medullary carci-
of tubular breast carcinoma and radial scar. Acta Cytol 1994;38:884– noma of the breast, proposal for a new simplified histopathological
890. definition. Based on prognostic observations and observations on inter-
Dei Tos AP, Della-Giustina D, Martin VD, et al. Aspiration biopsy cytology and intraobserver variability of 11 histopathological characteristics in
of tubular carcinoma of the breast. Diagn Cytopathol 1994;11:146– 131 breast carcinomas with medullary features. Br J Cancer 1991;63:
150. 591–595.
Evans AT, Hussein KA. A microglandular adenosis-like lesion simulating Ridolfi RF, Rosen PP, Post A, Kinne T, Mike V. Medullary carcinoma of the
tubular adenocarcinoma of the breast. A case report with cytological breast. A clinicopathologic study with 10 year follow-up. Cancer 1977;
and histological appearances. Cytopathology 1990;1:311–316. 40:1365–1385.

Ch005-F06731.indd 157 10/26/2006 10:26:11 AM


158 FINE NEEDLE ASPIRATION CYTOLOGY

Wargotz ES, Silverberg SG. Medullary carcinoma of the breast: a clinico- Levine PH, Zamuco R, Yee HT. Role of fine-needle aspiration cytology in
pathologic study with appraisal of current diagnostic criteria. Hum breast lymphoma. Diagn Cytopathol 2004;30:332–340.
Pathol 1988;19:1340–1346. Mattia AR, Ferry JA, Harris JA. Breast lymphoma: a B-cell spectrum includ-
ing the low grade B-cell lymphoma of mucosa associated lymphoid
tissue. Am J Surg Pathol 1993;17:574–587.
Apocrine Carcinoma Miura H, Konaka C, Kamate N, et al. Fine needle aspiration cytology of
metastatic breast tumor originating from leukemia. Diagn Cytopathol
Abati AD, Kimmel M, Rosen PP. Apocrine mammary carcinoma. A clinico- 1992;8:605–608.
pathologic study of 72 cases. Am J Clin Pathol 1990;94:371–377. Nielsen M, Andersen JA, Henriksen FW, et al. Metastases to the breast
Bryant J. Male breast cancer: a case of apocrine carcinoma with psam- from extramammary carcinomas. Acta Pathol Microbiol Scand [A]
moma bodies. Hum Pathol 1981;12:751–753. 1981;89:251–256.
Frable WJ, Kay S. Carcinoma of the breast. Histologic and clinical features Pettinato G, Manivel JC, Petrella G, De Chiara A. Primary multilobated
of apocrine tumors. Cancer 1968;21:756–763. T-cell lymphoma of the breast diagnosed by fine needle aspiration
Gupta RK, McHutchison AG, Simpson JS, Dowle CS. Fine needle aspiration cytology and immunocytochemistry. Acta Cytol 1991;35:294–299.
cytodiagnosis of apocrine carcinoma of the breast. Cytopathology Silverman JF, Feldman PS, Covell JL, Frable WJ. Fine needle aspiration
1992;3:321–326. cytology of neoplasms metastatic to the breast. Acta Cytol 1987;31:
Ito F, Avai, Suzuki M, et al. Apocrine carcinoma of the breast with cyto- 291–300.
logic, histologic and electron microscopic findings: a case report Sneige N, Zachariah S, Fanning TV, Dekmezian RH, Ordonez NG. Fine
[abstract]. Acta Cytol 1992;36:792. needle aspiration cytology of metastatic neoplasms in the breast. Am
Japaze H, Emina J, Diaz C, et al. Pure invasive apocrine carcinoma of the J Clin Pathol 1989;92:26–35.
breast: a new clinicopathological entity? Breast 2005;14:3–10.
Makunura CN, Curling OM, Yeomans P, Perry N, Wells CA. Apocrine adenosis
within a radial scar: a case of false positive breast cytodiagnosis. Granular Cell Tumor
Cytopathology 1994;5:123–128.
Page DL, Anderson TJ. Miscellaneous features of carcinoma. In: Diagnostic Adeniran A, Al-Ahmadie H, Mahone M, Robinson-Smith T. Granular cell
Histopathology of the Breast. Edinburgh: Churchill Livingstone, 1987: tumor of the breast: a series of 17 cases and review of the literature.
294–295. Breast J 2004;19:528–531.
Simpson JF, Page DL, Dupont WD. Apocrine adenosis – a mimic of DeMay RM, Kay S. Granular cell tumor of the breast. Pathol Annu
mammary carcinoma. Surg Pathol 1990;3:289–299. 1984;19:121–148.
Zajdela A, Ghossein NA, Pilleran JP, et al. The experience of aspiration Geisinger KR, Kawamoto EH, Marshall EB, et al. Aspiration and exfoliative
cytology in the diagnosis of breast cancer. Experience at the Foundation cytology, including ultrastructure of a malignant granular cell tumor.
Curie. Cancer 1975;35:499–506. Acta Cytol 1985;29:593–597.
Ingram DL, Mossler JA, Snowwhite I, et al. Granular cell tumors of the
breast. Steroid receptor analysis and localization of carcinoembryonic
Infiltrating Lobular Carcinoma antigen, myoglobin, and S-100 protein. Arch Pathol Lab Med 1984;108:
897–901.
Dixon JM, Anderson TJ, Page DL, et al. Infiltrating lobular carcinoma of Lowhagen T, Rubio CA. The cytology of the granular cell myoblastoma of
the breast: an evaluation of the incidence and consequence of bilateral the breast. Report of a case. Acta Cytol 1977;21:314–315.
disease. Br J Surg 1983;70:513–516. Strobel SL, Shah NT, Lucas JG, et al. Granular cell tumor of the breast: a
Ermier WJ, Gaffey TA, Welch JS, et al. Linitis plastica caused by metastatic cytologic immunohistochemical and ultrastructural study of two cases.
lobular carcinoma of the breast. Mayo Clin Proc 1980;55:747–753. Acta Cytol 1985;29:598–601.
Eusebi V, Maghales F, Azzopardi JG. Pleomorphic lobular carcinoma of the
breast. An aggressive tumor showing apocrine differentiation. Hum
Pathol 1992;23:655–662. Gynecomastia
Fechner RE. Histologic variants of infiltrating lobular carcinoma of the
breast. Hum Pathol 1975;6:373–378. Gorczyca W, Olszewski W, Tuziak T, et al. Fine needle aspiration biopsy of
Kline TS. Handbook of Fine Needle Aspiration Biopsy Cytology. St. Louis, rare malignant tumors of the breast. Acta Cytol 1992;36:918–926.
MO: CV Mosby, 1988:199–252. Hajdu SI, Espinosa MH, Robbins GF. Recurrent cystosarcoma phyllodes. A
Kline TS, Joshi LP, Neal HS. Fine needle aspiration of the breast: diag- clinicopathologic study of 32 cases. Cancer 1976;38:1402–1406.
nostic pitfalls. Cancer 1979;4:1458–1464. Hoeven KV, Hibbard C, Jones J, et al. Fine needle aspiration diagnosis of
Koss LG, Woyke J, Olszewski W. Cytologic Interpretation and Histologic tumors metastatic to the breast. Acta Cytol 1991;35:613.
Bases. New York: Igaku-Shoin, 1984. Koss LG, Woyke S, Olszewski W. Aspiration Biopsy: Cytologic Interpretation
Rajesh L, Dey P, Joshin K. Fine needle aspiration cytology of lobular breast and Histologic Bases. New York: Igaku-Shoin, 1984.
carcinoma. Comparison with other breast lesions. Acyta Cytol 2003;47: London G, Senige N, Ordonez NG, et al. Carcinoid metastatic to breast
177–182. diagnosed by fine needle aspiration biopsy. Diagn Cytopathol 1987;3:
Salhany KE, Page DL. Fine needle aspiration of mammary lobular carcinoma 230–233.
in situ and atypical lobular hyperplasia. Am J Clin Pathol 1989; Martinez-Onsurbe P, Fuentes-Vaamonde E, Gonzalez-Estecha A,
92:22–26. Hernandez-Ortiz MJ, Ruiz-Villaespesa A. Neurilemmoma of the breast in
Smith DB, Howell A, Harris M, et al. Carcinomatous meningitis associated a man. A case report. Acta Cytol 1992;36:511–513.
with infiltrating lobular carcinoma of the breast. Eur J Surg Oncol Miura H, Konaka C, Kamate N, et al. Fine needle aspiration cytology of
1985;11:33–36. metastatic breast tumor originating from leukemia. Diagn Cytopathol
1992;8:605–608.
Rao CR, Narasimhamurthy NK, Jaganathan K, Mukherjee G, Hazarika D.
Metastases to the Breast Cystosarcoma phyllodes. Diagnosis by fine needle aspiration cytology.
Acta Cytol 1992;36:203–207.
Gorczyca W, Olszewski W, Tuziak T, et al. Fine needle aspiration biopsy of Simi U, Moretti D, Iacconi P, et al. Fine needle aspiration cytopathology
rare malignant tumors of the breast. Acta Cytol 1992;36:918–926. of phyllodes tumor. Differential diagnosis with fibroadenoma [see com-
Gorczyca W, Wlodzimierz O, Tuziak T, Kram A, Woyke S, Ucinski M. Fine ments]. Acta Cytol 1988;32:63–66.
needle aspiration cytology of rare malignant tumors of the breast. Acta Toker C, Tang CK, Whitely JF, Verkheiser SW, Rachman R. Benign spindle
Cytol 1992;36:918–926. cell breast tumour. Cancer 1981;48:1615–1622.

Ch005-F06731.indd 158 10/26/2006 10:26:11 AM


6 Lung
Ema A Berbescu
Celeste N Powers
• Michael O Idowu •

Alveolar macrophages (Fig. 6-1B) have a similar


INTRODUCTION morphology to macrophages elsewhere in the body.
They have round, oval, or bean-shaped eccentric nuclei.
They usually have abundant cytoplasm and are typically
Fine needle aspiration (FNA) of the lung has increased laden with dust, carbon, and other particulate matters.
in popularity and frequency over the last two decades. Hemosiderin-laden macrophages usually indicate prior
It is often the preferred method for sampling solid hemorrhage or infarction; however, this diagnosis can
lesions and can be performed safely and accurately via be overlooked if the pigment is attributed to the normal
transbronchial (during fiberoptic bronchoscopy) or ‘dust burden’.
transthoracic (radiology-guided) methods; however, these Alveolar pneumocytes. Type I pneumocytes are not
methods are not useful for sampling diffuse pulmonary usually encountered in aspirate specimens. Type II
infiltrates. Tables 6-1 and 6-2 summarize the indications pneumocytes are relatively small with finely vacuolated
for transbronchial and transthoracic lung FNA. cytoplasm and centrally placed small nuclei. They may
Transbronchial FNA uses a retractable needle (Wang be difficult to distinguish from macrophages unless the
needle) attached at the tip of a flexible catheter that is latter have pigment-laden cytoplasm. Type II pneumo-
inserted into the flexible bronchoscope. Transthoracic cytes can increase in number in reparative states and in
FNA is performed under radiologic guidance using com- response to chronic injury such as pneumonia, diffuse
puted tomography (CT), fluoroscopy, or ultrasonogra- alveolar damage, drugs, radiotherapy, etc. In such cases,
phy. The needle types more commonly used are Chiba they may be in three-dimensional clusters or singly with
and Franseen, with needle gauges ranging from 18G to a high nuclear to cytoplasmic (N/C) ratio, coarse chro-
25G. Selection of the size, gauge, and type of needle is matin, and prominent nucleoli – and may resemble
based on location and predicted consistency of any adenocarcinoma, so much so, that the history of pneu-
given lesion, as well as experience and personal prefer- monia becomes very important.
ence. Many radiologists tend to select large diameter Mesothelial cells may be seen in the lung FNA, espe-
needles (18–20G) for these procedures; however, when cially in transthoracic FNA. This is due to the transfer
these needles are used, this procedure should not be of the pleural mesothelial lining by the aspiration needle,
termed fine needle aspiration biopsy. FNA uses 22G or often when the FNA target is a peripheral mass. The
smaller-sized needles (23G, 25G) and has much less risk morphology of the mesothelial cells in this case is similar
of complications, such as hemorrhage and/or pneumo- to that of the mesothelial cells covering visceral organs.
thorax, which are associated with the use of large- Mesothelial cells on FNA present as honeycomb
diameter biopsy needles. Complications of lung FNA are sheets of evenly spaced, uniform cells with abundant
presented in Table 6-3; contraindications of lung FNA cytoplasm, narrow gaps or ‘windows’ (due to the inter-
are summarized in Table 6-4. action of the microvilli), and small, discrete nucleoli
(Fig. 6-2).

NORMAL CONSTITUENTS NON-NEOPLASTIC CONDITIONS

Normal cellular constituents that may be encountered


SARCOIDOSIS
in lung FNA comprise bronchial cells, alveolar macro-
phages, pneumocytes, and mesothelial cells (in trans-
thoracic FNA). CLINICAL FEATURES
Bronchial cells are usually columnar or cuboidal and
ciliated (Fig. 6-1A). They have basal nuclei with small
nucleoli. Cilia and terminal bars are usually visible at Sarcoidosis is a multisystem granulomatous disease of
the apical poles. The presence of cilia in a cell group is unknown etiology. It affects predominantly the lungs
a major clue to its benign nature. (90% of patients), but can also involve extrapulmonary
159

Ch006-F06731.indd 159 10/26/2006 10:27:24 AM


160 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 6-1
Normal cellular constituents. A, Bronchial cells are columnar cells with small, round–oval, basally located nuclei. Note the presence of cilia and
terminal bars. Identifying cilia in a group of cells indicates their benign nature. Bronchial cells are frequently seen in transbronchial FNA materials.
Papanicolaou stain, medium power. B, Alveolar macrophages have abundant, somewhat vacuolated cytoplasm and a small, round nucleus. Charac-
teristically, macrophages hold on their cytoplasm during slide preparation. Diff-Quik stain, high power.

TABLE 6-1 TABLE 6-3


Indications for Transbronchial FNA Complications of Lung FNA

• Centrally located lesions: 1. Pneumothorax


• beneath the bronchial mucosa • Most common complication (its incidence increases
• intrabronchial polypoid lesions with the number of aspirations)
• Mediastinal adenopathy (sarcoidosis, lymphoma, staging • Most are clinically insignificant
of patients with lung cancer) • 5% of them require chest tube
2. Hemorrhage
3. Air embolism

TABLE 6-2
Indications for Transthoracic FNA

• Peripherally located nodules that cannot be sampled by TABLE 6-4


bronchoscopy
• Evaluation for metastasis in a patient with multiple lung Contraindications of Lung FNA
nodules
• Confirm the diagnosis of lung cancer in an inoperable • Bleeding diathesis and coagulation abnormalities
patient • Pulmonary hypertension (increased risk of bleeding)
• Confirm the diagnosis of small cell carcinoma and avoid • Severe respiratory distress
unnecessary surgery • Severe emphysema with bullae (risk of pneumothorax)

sites such as eye, skin, heart, liver, spleen, salivary The diagnosis of sarcoidosis includes compatible
glands, and central nervous system. Between 30% and clinicoradiologic features, pathologic evidence of non-
50% of patients are asymptomatic and are diagnosed caseating epithelioid granulomas, and exclusion of
on routine chest radiographs. One-third of patients similar diseases. The pathologic proof of epithelioid
have non-specific symptoms, such as fever, fatigue, granulomas is obtained most commonly by transbron-
weight loss, malaise, and respiratory symptoms (dry chial lung biopsy, but also lymph node or skin biopsy
cough, dyspnea, chest discomfort). Pulmonary sarcoid- can be helpful. Bronchoscopy with transbronchial lung
osis has an unpredictable course that may result in biopsy is non-diagnostic in 30% of patients with sus-
spontaneous remission or lead to progressive loss of pected sarcoidosis and has a risk of pneumothorax and
lung function with fibrosis. hemoptysis. Recently, endoscopic ultrasound-guided

Ch006-F06731.indd 160 10/26/2006 10:27:24 AM


CHAPTER 6 Lung 161

FIGURE 6-2
Mesothelial cells. They present as
fl at, two-dimensional sheets of
evenly spaced, bland cells. Meso-
thelial cells can be sampled during
transthoracic FNA and usually they
are sparsely present within a given
specimen. It is very important not
to confuse them with the cells
aspirated from a bronchioloalveolar
carcinoma. Clues to the origin of
these cells are sparse cellularity
and lack of single, discohesive cells.
Diff-Quik stain, low power.

particularly methotrexate (Rheumatrex) and azathio-


SARCOIDOSIS – DISEASE FACT SHEET prine (Imuran).

Definition
៉ Granulomatous disease of unknown etiology
R ADIOLOGIC FEATURES
Gender, Race, and Age Distribution
៉ More common in women than in men (incidence of 6.3 cases and Chest X-ray findings are classified in four stages. Stage
5.9 cases per 100,000 person-year, respectively) 0 refers to an apparently normal chest radiograph; stage
៉ Affects all races and ages, but more common in patients younger I consists of bilateral hilar adenopathy without paren-
than 40 years and more prevalent in US Blacks, Swedes, and chymal changes; stage II refers to hilar adenopathy and
Danes
parenchymal involvement; stage III consists of only
parenchymal involvement; and stage IV refers to
Clinical Features
advanced fibrosis with bullae formation.
៉ 30–50% of patients are asymptomatic
៉ One-third of patients have non-specific symptoms

Radiologic Features CYTOPATHOLOGIC FEATURES


៉ Four stages

Prognosis and Treatment The characteristic feature of sarcoidosis is tight, non-


៉ Unpredictable clinical course
caseating epithelioid granulomas (Fig. 6-3). In general,
៉ Corticotherapy, cytotoxic agents in the FNA material from hilar lymph nodes, the
epithelioid granulomas are numerous, in a background
of lymphocytes.
The characteristic cell is the epithelioid histiocyte,
which is a large, polygonal cell with abundant eosino-
philic cytoplasm and indistinct cell borders. The nucleus
FNA of enlarged hilar lymph nodes has been performed is elliptical, slightly curved (‘footprint’) and pale, with
in patients with suspected sarcoidosis, with 95% sensi- fine chromatin and one or two small nucleoli. Admixed
tivity and specificity. with the epithelioid histiocytes are multinucleated
Oral corticosteroids are the mainstay therapy for giant cells. In general, caseous necrosis is absent, but
pulmonary sarcoidosis. Patients who do not respond occasionally a small focus of necrosis can be seen in
to corticosteroids may benefit from cytotoxic agents, large granulomas.

Ch006-F06731.indd 161 10/26/2006 10:27:25 AM


162 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 6-3
Sarcoidosis. The characteristic feature of sarcoidosis is the presence of non-caseating epithelioid granulomas. They consist of tight clusters of epi-
thelioid histiocytes admixed with lymphocytes. The inset shows the characteristic features of epithelioid histiocytes: abundant eosinophilic cyto-
plasm with indistinct cell borders and a slightly curved (‘footprint’) nucleus with fine nuclear chromatin. Diff-Quik stain, low power (inset: Diff-Quik
stain, high power).

SARCOIDOSIS – PATHOLOGIC FEATURES


ANCILLARY STUDIES

Cytopathologic Findings
Sarcoidosis is a diagnosis of exclusion. Infectious and
៉ Tight, non-caseating epithelioid granulomas
non-infectious etiologies need to be excluded before
៉ Epithelioid histiocyte is a large cell with abundant eosinophilic
making the diagnosis of sarcoidosis. Special stains for
cytoplasm, indistinct cell borders, and a slightly curved
(‘footprint’) nucleus
microorganisms (Ziehl-Neelsen, Gomori methenamine
៉ Multinucleated giant cells can be present silver [GMS]) need to be performed on the cytology
៉ It is a diagnosis of exclusion material. Cultures can be obtained at the time of FNA.
Foreign material (talc, beryllium, etc.) can be identified
Ancillary Studies by light microscopy using polarized light or by laser
៉ Special stains (Ziehl-Neelsen, GMS) mass spectrometric analysis.
៉ Cultures

Differential Diagnosis and Pitfalls


៉ Mycobacterial infections DIFFERENTIAL DIAGNOSIS AND PITFALLS
៉ Histoplasmosis
៉ Granulomas accompanying malignancy
Non-caseating granulomas are not specific for sarcoid-
osis and they can be seen in a variety of other

Ch006-F06731.indd 162 10/26/2006 10:27:25 AM


CHAPTER 6 Lung 163

conditions. The differential diagnosis for epithelioid R ADIOLOGIC FEATURES


granulomas in the hilar lymph nodes includes mycobac-
terial infection (including atypical mycobacteria), histo-
plasmosis, and non-specific granulomas accompanying Typically, tuberculosis involves the apical and poste-
malignancy (T-cell lymphomas, Hodgkin lymphoma, rior segments of upper lobes, but any lung segment can
seminomas). be affected. The chest X-ray shows hazy infi ltrates that
progress to cavitation and fibrosis.

TUBERCULOSIS
CYTOPATHOLOGIC FEATURES
CLINICAL FEATURES
The aspirated material shows granulomas, plump his-
tiocytes, and/or necrosis (Figs 6-4 & 6-5). Routine
Worldwide it is estimated that 1 billion people are cytologic stains do not stain the actual bacilli; however,
infected with Mycobacterium tuberculosis. In the United there may be significant clues that suggest their pres-
States, the annual tuberculosis case rate is estimated to ence. Mycobacteria cannot be identified on Papanico-
be 9.5 per 100,000 persons. Forty percent of tubercu- laou stain. While the Diff-Quik stain does not stain the
losis cases in the United States occur in foreign-born individual organisms per se, it does outline them. The
persons. unstained bacilli appear as slender, straight or slightly
Tuberculosis is the result of infection with Mycobac- curved, colorless rods highlighted against a dirty blue–
terium tuberculosis complex (M. tuberculosis, M. bovis), grey background. Thus, the terms ‘negative images’ or
transmitted by infected aerosols, by direct person- ‘ghost bacilli’ are used as descriptors. This characteris-
to-person contact (M. tuberculosis), or by ingestion of tic ‘negative image’ is presumably the result of hydro-
contaminated food (milk; M. bovis). After the initial phobic interactions of the water-based Diff-Quik stain
infection (primary tuberculosis), the disease is latent with the lipid within the cell walls of the bacilli (Fig.
for a variable period of time and reactivates when the 6-6). The identification of these negative images within
immune system is weakened (secondary tuberculosis). histiocytes and in the background is virtually patho-
Non-tuberculous mycobacterial infections (M. avium- gnomonic for mycobacteria. Lowering the substage
intracellulare complex) cannot be distinguished on clini- condenser often increases the refractivity of the bacilli
cal and radiologic grounds from M. tuberculosis complex and enhances their identification when they are scat-
infections. This distinction is made only by cultures tered in the smear background. In FNA material from
with biochemical tests or using polymerase chain reac- mediastinal lymph nodes, the presence of numerous
tion (PCR) techniques. organisms both within distended histiocytes and scat-
Treatment is characterized by using multiple drugs tered in the background tends to be from atypical myco-
for a long period of time to prevent resistance. Major bacterial infections rather than from tuberculosis.
antituberculous drugs used are isoniazid, rifampin,
pyrazinamide, ethambutol, and streptomycin.

TUBERCULOSIS – PATHOLOGIC FEATURES

TUBERCULOSIS – DISEASE FACT SHEET Cytopathologic Findings


៉ Necrotizing granulomatous infl ammation
Definition
៉ Routine cytologic stains do not stain the bacilli
៉ Infectious disease caused by Mycobacterium tuberculosis ៉ ‘Negative images’ or ‘ghost bacilli’ can be seen on Diff-Quik
stain (unstained bacilli as colorless slender rods against a
Clinical Features dirty-blue background)
៉ Pathogenic non-tuberculous mycobacteria species (such as M.
avium-intracellulare complex) produce similar clinical and Ancillary Studies
radiologic findings; distinction made only by cultures or PCR ៉ Acid-fast (Ziehl-Neelsen) or auramine rhodamine stains, or
៉ In the US, certain populations are at increased risk: foreign-born culture are necessary for confirmation
individuals, HIV-positive individuals, nursing home residents, and ៉ Material can be obtained for rapid detection of Mycobacterium
prison inmates tuberculosis by PCR techniques

Radiologic Features Differential Diagnosis and Pitfalls


៉ More commonly involves the apical and posterior segments of ៉ Other conditions associated with granulomatous infl ammation
upper lobes (sarcoidosis, fungal infections, etc.)
៉ Hazy infiltrates that progress to cavitation and fibrosis ៉ Clofazimine crystals within macrophages can mimic ‘negative
images’
Treatment ៉ Nocardia species can appear as acid-fast, beaded filamentous
៉ Multiple antituberculous drugs over a long period of time organisms

Ch006-F06731.indd 163 10/26/2006 10:27:26 AM


164 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 6-4
Tuberculosis. The FNA yields abundant necrosis and scattered granulomas, characteristic findings in any necrotizing granulomatous process. The
inset shows a Ziehl-Neelsen stain performed on the cell block material that reveals scattered acid-fast bacilli. Diff-Quik stain, low power (inset:
Ziehl-Neelsen stain, high power).

FIGURE 6-5
Tuberculosis. The granulomas in
tuberculosis are loosely formed,
with admixed necrosis and infl am-
matory cells. Contrast this with the
tight granulomas and clean back-
ground seen in sarcoidosis. Papani-
colaou stain, high power.

Ch006-F06731.indd 164 10/26/2006 10:27:26 AM


CHAPTER 6 Lung 165

FIGURE 6-6
Atypical mycobacteria. The FNA yields a dirty-grey background in which the unstained bacilli appear as slender, straight or slightly curved colorless
rods (‘negative images’). Ziehl-Neelsen stain performed on an alcohol-fixed slide confirms the presence of numerous acid-fast bacilli (inset). Diff-
Quik stain, low power (inset: Ziehl-Neelsen stain, high power).

Acid-fast stain (Ziehl-Neelsen) can be performed on inflammation with or without necrosis. The presence
smears, cytospins, and cell block sections and should be of M. tuberculosis or atypical mycobacteria should be
used for confirmation. The organisms appear as thin, suspected in every case of granulomatous inflamma-
slender, red beaded bacilli. tion, especially when the patient is immunosuppressed,
and confirmed with acid-fast (Ziehl-Neelsen) or aura-
mine rhodamine stains, cultures, or PCR. ‘Negative
images’ are not specific. Patients treated with clofazi-
ANCILLARY STUDIES mine show crystal formation within macrophages that
mimic negative images. Nocardia species can appear
beaded on acid-fast stain; however, they are slender
The diagnosis of tuberculosis is confirmed by identify- fi laments, branching at acute angles, and are larger in
ing acid-fast bacilli in respiratory specimens or cul- diameter than mycobacteria.
tures. In many instances, when the patient is known
to be immunocompromised or has had prior infections,
the index of suspicion for infection is high and addi-
tional specimens are obtained for culture or fluores-
cence microscopy. PCR can now be used for rapid FUNGAL INFECTIONS
detection of M. tuberculosis.
Pulmonary infections with dimorphic fungi (hyphae at
room temperature and yeasts at body temperature) are
DIFFERENTIAL DIAGNOSIS AND PITFALLS increasing in importance as causes for opportunistic
infections in immunocompromised patients, especially
those with AIDS. The three dimorphic fungi discussed
Sarcoidosis, fungal infections, and reaction to an adja- in this chapter are Histoplasma capsulatum, Blasto-
cent neoplasm (such as Hodgkin lymphoma, seminoma, myces dermatitidis, and Coccidioides immitis. Other
T-cell lymphomas) also present with granulomatous important fungi involved in pulmonary infections in

Ch006-F06731.indd 165 10/26/2006 10:27:27 AM


166 FINE NEEDLE ASPIRATION CYTOLOGY

immunocompromised individuals that will be dis- Acute histoplasmosis is characterized by flu-like


cussed in this chapter are Cryptococcus neoformans, symptoms (fever, chills, myalgia, headache, cough) and
Aspergillus fumigatus, and fungi belonging to Zygomy- chest X-ray abnormalities. Chronic pulmonary histo-
cetes class (Rhizopus, Absidia, and Mucor species). plasmosis develops in patients with emphysema exposed
to H. capsulatum. They present with cough, dyspnea,
fatigue, fever, and sweats, mimicking tuberculosis.
Granulomatous mediastinitis produces obstruction of
HISTOPLASMOSIS mediastinal structures (airways, superior vena cava,
pulmonary vessels, esophagus). The mediastinal lymph
nodes are enlarged, forming cystic masses with central
CLINICAL FEATURES necrosis and surrounded by granulomatous inflamma-
tion and encased in a fibrous capsule. Disseminated
Histoplasmosis is produced by inhalation of microco- histoplasmosis occurs in 1 in 2000 acute infections.
nidia of Histoplasma capsulatum or by reactivation of It is rare in immunocompetent individuals and more
latent infection in individuals with past histoplasmosis common in immunosuppressed individuals (in particu-
who become immunosuppressed. H. capsulatum can lar, AIDS). The infection spreads to extrapulmonary
be found throughout the world. In the United States, sites (liver, spleen, bone marrow, skin, gastrointestinal
H. capsulatum is endemic in the Mississippi and Ohio tract, adrenal glands). Fibrosing mediastinitis is an
river valleys. Bird and bat excrement enhance growth excessive fibrotic response to past infection. This is a
of the organism in the soil. rare complication (less than 1 in 5000 infections); 80%
In endemic areas, 50–80% of young adults are of cases occur in individuals between the ages of 20 and
infected with H. capsulatum and are asymptomatic. 40 years.
After low-level exposure, less than 5% of individuals Antifungal therapy (triazole agents and liposomal
develop symptomatic disease. After high-level exposure formulations of amphotericin B) is indicated in patients
(work or recreational activities in contaminated sites), with symptomatic acute pulmonary histoplasmosis,
more than 75% of individuals develop symptoms. granulomatous mediastinitis, chronic pulmonary
There are several clinical manifestations of infection histoplasmosis, and in disseminated disease. Antifungal
with H. capsulatum: acute pulmonary histoplasmosis, therapy is not indicated in acute, self-limited forms
chronic pulmonary histoplasmosis, granulomatous of pulmonary histoplasmosis or in fibrosing
mediastinitis, disseminated histoplasmosis, and fibros- mediastinitis.
ing mediastinitis.

HISTOPLASMOSIS – DISEASE FACT SHEET R ADIOLOGIC FEATURES

Definition
Chest X-ray in symptomatic acute pulmonary histoplas-
៉ Infection resulting from inhalation of Histoplasma capsulatum or
reactivation of latent infection
mosis shows enlarged hilar and mediastinal lymph
nodes with focal patchy or nodular pulmonary infi l-
Incidence and Geographic Distribution trates. In granulomatous mediastinitis, radiologic
៉ Annual incidence: 5/100,000
studies show enlarged, often cystic, mediastinal lymph
៉ Endemic in Mississippi and Ohio river valleys nodes (measuring up to 10 cm). Chest X-ray in fibrosing
mediastinitis shows subcarinal and mediastinal widen-
Clinical Features ing, but high-resolution chest CT with contrast is
៉ 50–80% of infected young adults are asymptomatic required to delineate the mediastinal abnormalities.
៉ Several clinical manifestations: acute pulmonary histoplasmosis
(mimics flu); chronic histoplasmosis (mimics tuberculosis);
granulomatous mediastinitis (less than 10% of patients);
disseminated histoplasmosis and fibrosing mediastinitis CYTOPATHOLOGIC FEATURES
Radiologic Features
៉ Acute form: hilar and mediastinal adenopathy with pulmonary Material can be obtained by FNA from enlarged medi-
infiltrates astinal lymph nodes and cavitary lesions. H. capsula-
៉ Granulomatous mediastinitis: enlarged, cystic mediastinal lymph tum is one of the smallest dimorphic fungi (3 to 5 μm).
nodes It is predominantly intracellular, as foamy or vacuo-
៉ Fibrosing mediastinitis: mediastinal widening
lated oval structures within the cytoplasm of histio-
cytes (Fig. 6-7). Yeast forms of H. capsulatum are easiest
Treatment
to identify when they are within macrophages; their
៉ Antifungal therapy in symptomatic patients
engulfment results in a vacuolated or foamy cytoplasm
៉ Antifungal therapy not indicated for self-limited forms of acute
disease or for fibrosing mediastinitis
with small halos surrounding each organism (due to
retraction from the wall creating the appearance of a
pseudocapsule). Macrophages with engulfed organisms

Ch006-F06731.indd 166 10/26/2006 10:27:28 AM


CHAPTER 6 Lung 167

ANCILLARY STUDIES

PCR studies can be done in cases in which the morphol-


ogy of the yeasts identified in tissue specimens is incon-
clusive. Detection of Histoplasma antigen in body fluids
(blood, urine, bronchoalveolar lavage) allows a rapid
diagnosis (24 to 48 hours) in patients with severe
disease. Cultures are also useful, particularly in the
disseminated form. The highest yield is from blood and
bone marrow specimens.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

GMS stain must be interpreted carefully, as other


organisms (Candida species, Cryptococcus neoformans,
FIGURE 6-7 Blastomyces dermatitidis, Toxoplasma gondii, Leishma-
Histoplasmosis. The yeast forms of Histoplasma capsulatum appear as nia species, Pneumocystis carinii) or staining artifacts
foamy or vacuolated oval structures within the cytoplasm of histio- can be misinterpreted as H. capsulatum. When the
cytes. The small halos surrounding each organism are the result of wall
retraction. This artifact can mimic the Cryptococcus neoformans capsule. organism retracts from its wall, it creates a clear space
Diff-Quik stain, high power. that mimics the capsule of C. neoformans. If the differ-
ential diagnosis includes leishmaniasis, recognition of
a nucleus and kinetoplast and the absence of budding
will exclude H. capsulatum.
HISTOPLASMOSIS – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Histoplasma capsulatum is a small yeast (3 to 5 μm), with BLASTOMYCOSIS
narrow-based budding
៉ Usually intracellular (engulfed by macrophages)
៉ Vacuolated cytoplasm with small halos surrounding the CLINICAL FEATURES
organism (appearance of pseudocapsule)
៉ On Papanicolaou stain, it is difficult to visualize
៉ On Diff-Quik stain: dark-purple organisms Blastomycosis is the result of inhalation of spores of
Blastomyces dermatitidis. In the United States, the
Ancillary Studies
៉ Confirmatory stain: GMS
៉ PCR performed when morphology is inconclusive BLASTOMYCOSIS – DISEASE FACT SHEET
៉ Histoplasma antigen detection and cultures in severe cases,
particularly in disseminated disease Definition
៉ Infection caused by inhalation of Blastomyces dermatitidis
Differential Diagnosis and Pitfalls
៉ Other organisms (Candida species, Cryptococcus neoformans, Incidence and Geographic Distribution
Blastomyces dermatitidis, Toxoplasma gondii, Leishmania species, ៉ Annual incidence: 0.5/100,000
Pneumocystis carinii) ៉ Endemic areas in the US are Central, South-central, and
៉ Staining artifacts Southeastern
៉ Endemic area overlaps with that of histoplasmosis

Clinical Features
៉ Primary infection in the lung with subsequent dissemination to
are typically present in a background of granulomatous
skin and bone
inflammation that can also include necrosis, acute
inflammatory cells, and lymphocytes. Yeast forms of Radiologic Features
H. capsulatum demonstrate narrow-based (teardrop)
៉ Variable: round densities, consolidations, mass-like infiltrates,
budding. If present in small numbers, H. capsulatum etc.
can be very difficult to visualize on Papanicolaou stain.
The Diff-Quik stain is useful because it stains these Treatment
organisms a dark purple that is usually in stark con- ៉ Itraconazole (in sporadic cases) and amphotericin B (in severely
trast to the pale-staining cytoplasm. Confirmatory stain ill and immunosuppressed patients)
(GMS) should be performed when this organism is
presumptively identified.

Ch006-F06731.indd 167 10/26/2006 10:27:28 AM


168 FINE NEEDLE ASPIRATION CYTOLOGY

endemic areas are Central, South-central, and South- multinucleated giant cells. The organisms will be found
eastern, with the highest incidence along the Missis- in this non-specific background. Only the yeast forms
sippi and Ohio river valleys and the western shore of are identified in vivo. Yeast forms of B. dermatitidis are
Lake Michigan. The endemic area overlaps with the large (8–15 μm), spherical, and double-contoured. They
endemic area for histoplasmosis. The annual incidence have a rigid, refractile cell wall with a centrally retracted
is roughly 0.5/100,000. cytoplasm (Fig. 6-8). The yeast forms are generally
Clinically, patients can be asymptomatic or can larger than those of Cryptococcus neoformans. Because
present with life-threatening pulmonary and dissemi- of their double-contoured rigid cell wall, they often
nated disease. The majority of patients with sporadic appear slightly out of the plane of focus, making detec-
pulmonary blastomycosis do well with itraconazole tion difficult on routine screening. Characteristically,
therapy, which is safe, highly effective, and non-toxic. they have a broad-based budding. Broad-based budding
Severely ill patients require amphotericin B therapy. is a useful criterion to differentiate B. dermatitidis
Immunosuppressed patients (on prednisone or organ from the other dimorphic fungi. The yeast forms appear
transplant recipients) require sequential therapy of blue–green on Papanicolaou stain and blue on Diff-
amphotericin B until improved clinically, followed by Quik stain. Although most often identified extra-
6–12 months of itraconazole. HIV-infected patients cellularly, these organisms can also be engulfed by
require a similar sequential therapy, but they are not macrophages. Special stain (GMS) can be performed to
permanently cured and lifelong therapy with itracon- highlight the yeast forms. Cytomorphology is usually
azole is necessary to maintain control. sufficient for diagnosis. This is the easiest and fastest
way to make a diagnosis. Culture identification takes
usually several weeks.

R ADIOLOGIC FEATURES
DIFFERENTIAL DIAGNOSIS AND PITFALLS
Radiologic findings are variable and include single
or multiple round densities scattered throughout,
segmental/lobar consolidation, mass-like perihilar in- B. dermatitidis yeasts can be confused with the imma-
fi ltrates, and diffuse nodular, interstitial, or alveolar ture spherules of Coccidioides immitis. The endospores
infi ltrates. of C. immitis are not visible in the immature spherules.
This, combined with the thick wall of C. immitis,
results in an erroneous identification as B.
dermatitidis.
CYTOPATHOLOGIC FEATURES Another potential problem is that the pathologist
may overlook these yeast forms. Although larger, yeast
forms of B. dermatitidis are very difficult to identify on
B. dermatitidis produces a granulomatous suppurative cytologic preparations, chiefly because they are not
inflammatory reaction. FNA cytology aspirate shows numerous and are often out of the plane of focus. The
acute suppurative inflammation, necrotic debris, and latter problem is due to their rigid cell walls, which
clusters of epithelioid histiocytes, macrophages, and resist compression by a coverslip.

BLASTOMYCOSIS – PATHOLOGIC FEATURES


COCCIDIOIDOMYCOSIS
Cytopathologic Findings
៉ Blastomyces dermatitidis is a large yeast (8–15 μm), spherical,
double-contoured with broad-based budding CLINICAL FEATURES
៉ Has a rigid, refractile cell wall appearing slightly out of plane
of focus
៉ Background shows acute suppurative infl ammation, necrosis, Coccidioidomycosis is produced by inhalation of arthro-
and macrophages conidia of Coccidioides immitis. This is a dimorphic
៉ Yeast forms can be extracellular or engulfed by macrophages fungus limited to the Western hemisphere, from Cali-
៉ In Papanicolaou stain, appears blue–green
fornia to Argentina. Endemic areas in the United States
៉ In Diff-Quik stain, appears blue
are Southwest desert regions (Arizona, California,
Nevada, Utah, New Mexico, and Texas). Patients with
Ancillary Studies
impaired cell-mediated immunity (HIV infection,
៉ GMS highlights the yeast forms
prolonged corticotherapy, organ transplant recipients,
៉ Culture takes several weeks
patients under chemotherapy, pregnant women) are
Differential Diagnosis and Pitfalls
most susceptible to C. immitis infection.
Between 60% and 70% of individuals exposed to
៉ Immature spherules of Coccidioides immitis
៉ Can be completely overlooked because is out of plane of focus
C. immitis are completely asymptomatic. The rest of
patients develop signs and symptoms of primary pulmo-
nary coccidioidomycosis. A small group of patients can

Ch006-F06731.indd 168 10/26/2006 10:27:28 AM


CHAPTER 6 Lung 169

FIGURE 6-8
Blastomycosis. Yeast forms of Blastomyces dermatitidis are spherical, double-contoured with a rigid, refractile wall. They appear blue on Diff-Quik
stain. The inset shows several yeasts of B. dermatitidis engulfed by a macrophage. Note the broad-based budding. Diff-Quik stain, high power (inset:
GMS stain, high power).

develop disseminated disease, with predilection for skin,


bones, soft tissues, meninges, and disseminated pulmo-
COCCIDIOIDOMYCOSIS – DISEASE FACT SHEET
nary disease. The sequelae of pulmonary disease are
Definition
nodules and cavities that are single, asymptomatic, less
than 5 cm, and can persist for a long period of time.
៉ Fungal disease caused by inhalation of Coccidioides immitis
Cavities can be complicated by infection (bacteria from
Distribution
upper respiratory tract or Aspergillus species), hemo-
ptysis or rupture into the pleural space with pyopneu-
៉ Endemic in Southwest desert regions
៉ Most susceptible are HIV-positive individuals, organ transplant
mothorax. The antifungal therapy (with amphotericin
recipients, patients under chemotherapy or prolonged B and azoles) is reserved for immunocompromised
corticotherapy, and pregnant women patients with pulmonary or disseminated disease and
for immunocompetent patients whose symptoms have
Clinical Features persisted for more than 6 weeks.
៉ 60–70% of patients are asymptomatic

Radiologic Features
៉ Alveolar infiltrates that progress to cavities
R ADIOLOGIC FEATURES

Prognosis and Treatment


In primary pulmonary coccidioidomycosis, the chest
៉ Pulmonary sequelae: cavities that can get infected or rupture
X-ray shows segmental or lobar alveolar infi ltrates with
within pleural space
occasional hilar and mediastinal adenopathy. The cavi-
៉ Antifungal therapy for immunocompromised individuals or
patients with prolonged symptoms
ties are thin-walled, and, when infected, they will
appear with air–fluid level, opacified, and surrounded
by parenchymal infi ltration (Fig. 6-9).

Ch006-F06731.indd 169 10/26/2006 10:27:28 AM


170 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 6-10
Coccidioidomycosis. The thick-walled spherules of Coccidioides immitis
are identified engulfed by macrophages. They appear orangeophilic in
Papanicolaou stain, being easy to identify. Papanicolaou stain, high
power.

phages (Figs 6-10 & 6-11A). This organism presents as


endospores within thick-walled spherules that can vary
tremendously in size. Endospores are not visible in
immature spherules; as the spherules mature, they will
contain numerous small (2–5 μm) endospores. The
endospores are expelled from the spherule, creating an
FIGURE 6-9 acute inflammatory reaction. The now-empty spherule
Cavitary lesion: radiologic findings. An isolated cavitary lesion with will appear fractured and folded (Fig. 6-11B). This
air–fluid level is seen in the left lower lobe. The differential diagnosis organism occasionally stains orangeophilic by Papani-
includes infectious process (abscess; tuberculosis; fungal infection, colaou stain, making it easy to detect (Fig. 6-11C). The
especially coccidioidomycosis, aspergillosis, and zygomycosis) versus
malignant neoplasm (especially squamous cell carcinoma). spherules stain lightly or not at all with the Diff-Quik
stain, although, with experience, this lack of staining
is a useful clue. Periodic acid–Schiff (PAS) stain after
CYTOPATHOLOGIC FEATURES diastase digestion with a light-green counterstain is a
very useful confirmatory stain, since the spherules
stain a vivid magenta against a pale-green background
C. immitis is one of the largest of the dimorphic fungi. (Fig. 6-11D). Rarely, the cavities may be exposed to air,
The organism can be identified extracellularly within resulting in the development of mycelial forms with
a background of acute fibrinopurulent exudate and barrel-shaped arthroconidia (5% of cases).
granulomatous inflammation or engulfed by macro-

COCCIDIOIDOMYCOSIS – PATHOLOGIC FEATURES


ANCILLARY STUDIES

Cytopathologic Findings
Direct examination of respiratory specimens is nega-
៉ Coccidioides immitis is the largest of the dimorphic fungi
tive in approximately 60% of patients with culture-
៉ Presents as endospores within thick-walled spherules; the
positive pulmonary coccidioidomycosis. Fungal culture
endospores are expelled and the empty spherule appears folded
and fractured
is a sensitive method of diagnosis. The organism grows
៉ The spherules are extracellular or engulfed by macrophages rapidly (3 to 5 days), at 35ºC, on a variety of media. It
should be remembered that in the culture C. immitis
Ancillary Studies grows as a mold and is highly infectious.
៉ Fungal culture is sensitive (C. immitis in culture grows as a mold
and is highly infectious)

Differential Diagnosis and Pitfalls DIFFERENTIAL DIAGNOSIS AND PITFALLS


៉ Other fungi (Blastomyces dermatitidis, Histoplasma capsulatum)
៉ Pollen grains
C. immitis can resemble a variety of other fungi, pollen
៉ Inorganic contaminants
៉ Helpful clue: look for older, folded and fractured spherules
grains, and even inorganic contaminants. Endospores
are not visible in immature spherules; combined with
the thick wall of C. immitis, this often results in an

Ch006-F06731.indd 170 10/26/2006 10:27:29 AM


CHAPTER 6 Lung 171

A B

C D

FIGURE 6-11
Coccioidomycosis: FNA findings. A, The spherules of Coccidioides immitis are scattered in an acute fibrinopurulent background. They stain very light
with Diff-Quik, and this lack of staining can be a useful clue. Diff-Quik stain, low power. B, After the endospores are expelled, the empty spherule of
C. immitis appears fractured and folded. Diff-Quik stain, high power. C, A mature spherule of C. immitis containing numerous endospores is engulfed
by a macrophage. Note the orangeophilic staining. Papanicolaou stain, high power. D, Three spherules of C. immitis are engulfed by a macrophage.
They appear vivid magenta colored against a pale-green background. PAS stain after diastase digestion with light-green counterstain, high power.

erroneous identification as Blastomyces dermatitidis. CRYPTOCOCCOSIS – DISEASE FACT SHEET


Once the endospores are separated from the spherule,
they may resemble Histoplasma capsulatum and even Definition
Toxoplasma gondii, and a careful search for older, folded ៉ Infection caused by inhalation of Cryptococcus neoformans
and fractured spherules is necessary. Conversely, when
these older spherules are seen without intact, mature Incidence
spherules, they may be overlooked as inorganic ៉ Most common fungal infection in immunosuppressed individuals
contaminants. ៉ Isolated pulmonary cryptococcosis is rare

Clinical Features
៉ Affects brain, lung, skin, and bone
CRYPTOCOCCOSIS
Radiologic Features
៉ Pulmonary infiltrates, mimicking neoplasm or other infections
CLINICAL FEATURES
Treatment
៉ Amphotericin B with 5-fluorocytosine
Cryptococcosis is an infection caused by inhalation
from the environment of Cryptococcus neoformans,

Ch006-F06731.indd 171 10/26/2006 10:27:30 AM


172 FINE NEEDLE ASPIRATION CYTOLOGY

which is spread worldwide. The infection affects the CYTOPATHOLOGIC FEATURES


brain, lung, skin, or bone, both in immunocompetent
and in immunosuppressed patients. C. neoformans is
the most common fungal infection complicating HIV C. neoformans is a round or oval encapsulated yeast (5
infection. Meningitis is the most common form of to 10 μm in diameter). The capsule measures from 1 to
disease. Isolated pulmonary cryptococcosis is very 30 μm in thickness and is made of polysaccharides. Due
rare. The majority of patients clear the pulmonary to its large capsule, C. neoformans resists phagocytosis.
infection but later develop meningitis, while 90% of T-cell-mediated immunity with granuloma formation
patients with cryptococcal meningitis (excluding is necessary to stop the infection. In patients with
AIDS patients) have a clear chest X-ray and no history impaired T-cell-mediated immunity (HIV-positive,
of pneumonia. Treatment of cryptococcal infection in organ transplant recipients), granuloma formation does
immunosuppressed patients includes amphotericin B not occur and the growth continues, producing large
with 5-fluorocytosine; when clinical improvement is gelatinous masses of cryptococci with no tissue
obtained, then the treatment is switched to fluconazole reaction.
for life. In FNA material, this organism is usually abundant,
being found individually or as clusters of organisms
with accentuated capsular halos (Fig. 6-12). In Diff-
Quik-stained preparations, the purple yeasts with accen-
RADIOGRAPHIC FEATURES tuated clear halos against the deep-purple background
give the smear a punched-out appearance (Fig. 6-13). A
useful feature is the appearance of teardrop, narrow-
The chest radiographic fi lms show pulmonary infi l- based budding. At times, the daughter bud will not
trates, single or multiple, mimicking other pulmonary detach and repetition of budding yields small chains
infections or neoplasms. Occasionally, patients can of daughter progeny. Occasionally, the yeast can be
develop large, round masses up to 10 cm in diameter. engulfed by histiocytes.

FIGURE 6-12
Cryptococcosis. In the FNA material, the organisms are abundant, single or in small clusters. The inset shows the characteristic ‘teardrop’, narrow-
based budding. Diff-Quik stain, low power (inset: GMS stain, high power).

Ch006-F06731.indd 172 10/26/2006 10:27:31 AM


CHAPTER 6 Lung 173

based budding is a distinctive, helpful feature to iden-


tify C. neoformans.

ASPERGILLOSIS

CLINICAL FEATURES

Aspergillosis refers to infection, through inhalation,


with molds of the genus Aspergillus. The most common
pathogen encountered in lung infections is A. fumiga-
tus (which is the most pathogenic of all species). Pul-
monary manifestations are numerous and include
FIGURE 6-13 allergic bronchopulmonary aspergillosis, hypersensi-
Cryptococcosis. The yeast forms of Cryptococcus neoformans have a large tivity pneumonitis, asthma, aspergilloma, and invasive
polysaccharide capsule. They appear as purple yeasts with accentuated
clear halos giving the smear a punched-out appearance. Diff-Quik stain,
pulmonary aspergillosis. From all these manifesta-
high power. tions, invasive pulmonary aspergillosis and aspergil-
loma present most commonly as a distinct mass on
radiologic studies and can be sampled by FNA.
Invasive pulmonary aspergillosis is a progressive and
CRYPTOCOCCOSIS – PATHOLOGIC FEATURES usually fatal disease (mortality rate is 80%). Immuno-
suppression predisposes to invasive pulmonary asper-
Cytopathologic Findings
gillosis. Risk factors are prolonged neutropenia,
៉ Cryptococcus neoformans is a round to oval yeast (5 to 10 μm)
corticosteroid therapy, advanced HIV infection, bone
with a polysaccharide capsule
marrow and solid-organ transplant, and chronic granu-
៉ Teardrop, narrow-based budding
៉ In Diff-Quik stain: purple yeasts with accentuated clear halos
lomatous disease. The incidence of invasive aspergillo-
against a deep-purple background sis is 0.7% in renal transplant recipients, 6.4% in bone
marrow transplant recipients, 8.4% in lung transplant
Ancillary Studies recipients, and 5–24% in patients with chemotherapy-
៉ Special stains: GMS (stains the organism), PAS, mucicarmine, induced neutropenia. Clinical manifestations of
Alcian blue (stain the capsule) invasive aspergillosis are fever, non-productive cough,

Differential Diagnosis and Pitfalls


៉ Histoplasma capsulatum
ASPERGILLOSIS – DISEASE FACT SHEET

Definition
៉ Infection caused by inhalation of molds of the genus Aspergillus
Diagnosis of C. neoformans still rests primarily on its
cytomorphology, with confirmation, as necessary, by Clinical Features
special stains. Silver stains (GMS) stain the organism
៉ Most common in lung infections is Aspergillus fumigatus
itself, while mucicarmine, PAS, and Alcian blue stain ៉ Invasive pulmonary aspergillosis and aspergilloma present as
the mucopolysaccharide capsule. masses
៉ Risk factors for invasive aspergillosis (80% mortality rate):
neutropenia, HIV infection, bone marrow and solid organ
transplant, chronic granulomatous disease, corticosteroid
ANCILLARY STUDIES therapy
៉ Aspergilloma: hyphae of Aspergillus species growing within a
pre-existing cavity
The capsule is the target of serologic tests both in serum
and in cerebrospinal fluid (CSF). The best single test Radiologic Features
for cryptococcal meningitis is the cryptococcal antigen ៉ Invasive aspergillosis: non-specific lung infiltrate that rapidly
assay. progresses to a cavitary mass or infarct
៉ Aspergilloma: cavitary mass with fungus ball (moves with the
position)

DIFFERENTIAL DIAGNOSIS AND PITFALLS Treatment


៉ Invasive aspergillosis: intravenous amphotericin B
៉ Aspergilloma: surgical excision
When the yeasts are engulfed by histiocytes, they can
be mistaken for Histoplasma capsulatum. Narrow-

Ch006-F06731.indd 173 10/26/2006 10:27:31 AM


174 FINE NEEDLE ASPIRATION CYTOLOGY

dyspnea, and hemoptysis, which can be life-threatening vague lung infi ltrate. This infi ltrate will progress in the
(due to the ability of A. fumigatus to invade blood course of days to a classic wedge-shaped infarct or a
vessels). consolidated mass with cavitation.
An aspergilloma consists of Aspergillus species Chest radiography in aspergilloma shows a cavitary
hyphae growth within a pre-existing cavity that mass, usually in the upper lobes. The fungus ball will
communicates with the bronchial tree. The etiology of move with the position (when standing and decubitus
such a cavity includes neoplasm, tuberculosis, healed films are compared).
abscess, cystic fibrosis, congenital cysts, and bullous
emphysema.
The standard therapy in invasive pulmonary asper-
gillosis is intravenous amphotericin. Resection of the CYTOPATHOLOGIC FEATURES
localized consolidated mass has been performed with
good results. Granulocyte–monocyte colony-stimulating
The FNA will show the organisms commonly present
factor may improve the outcome and should be consid-
as tangled clusters of branched hyphae that are accom-
ered in neutropenic patients.
panied by acute inflammation, necrosis, and cellular
The treatment of choice for aspergilloma is surgical
debris. When a cytologic specimen contains hyphal
excision. In patients with compromised lung function,
structures that are relatively large (3–6 μm in diame-
in whom surgical excision is not a viable solution, intra-
ter), septate, with regular, progressive dichotomous
cavitary instillation of amphotericin can be tried.
branching at 45-degree angles, Aspergillus species
should be immediately suspected (Fig. 6-14). Aspergil-
lus species stain fairly well by Papanicolaou and hema-
R ADIOLOGIC FEATURES toxylin–eosin (H&E) stains, which clearly delineate
the hyphal septations. The organisms also stain with
Diff-Quik; however, many times the staining is extreme,
In invasive pulmonary aspergillosis, the initial radio- i.e. either too dark to distinguish internal structure or
logic studies can appear normal or show a non-specific, too light such that the fungus blends into the dirty

FIGURE 6-14
Aspergillosis. The FNA material contains tangled clusters of branched hyphae, acute infl ammation, necrosis, and debris. The inset shows septate
hyphae of Aspergillus species with characteristic progressive, dichotomous, 45-degree branching. Papanicolaou stain, low power (inset: Diff-Quik
stain, high power).

Ch006-F06731.indd 174 10/26/2006 10:27:32 AM


CHAPTER 6 Lung 175

ZYGOMYCOSES
ASPERGILLOSIS – PATHOLOGIC FEATURES

Cytopathologic Findings CLINICAL FEATURES


៉ Septate hyphae with regular, progressive, dichotomous
branching at 45-degree angle
៉ Background shows abundant necrosis with reactive atypical Zygomycoses are an infrequent, often fatal, group of
squamous cells infections caused by fungal organisms from the class
៉ When fungus cavity exposed to air: fruiting bodies with conidia of Zygomycetes (Rhizopus, Absidia, and Mucor species).
៉ Rosettes of needle-like calcium oxalate crystals
The most common pathogen is Rhizopus species.
Pulmonary zygomycosis is most common in patients
Differential Diagnosis and Pitfalls
with hematologic malignancies (leukemia or lymphoma)
៉ The scattered atypical squamous cells within a necrotic
with severe neutropenia. The incidence of zygomycosis
background can be interpreted as necrotic squamous cell
carcinoma
in patients with hematologic malignancies is approxi-
៉ Clue: low cellularity should preclude a diagnosis of malignancy;
mately 1%. Other risk factors are uncontrolled diabetic
look for septate hyphae ketoacidosis, corticosteroid therapy, neutropenia, burns,
៉ Other fungi (Fusarium species, Pseudoallescheria boydii) HIV infection, and desferrioxamine therapy.
Patients will present with fever and pulmonary
infiltrates. The organism has a propensity for angio-
invasion, with thrombosis and tissue infarction. This
can lead to massive, life-threatening hemorrhage. The
infection can extend to invade the chest wall, diaphragm,
background. Septations are often colorless, creating a pericardium, etc. A definite diagnosis is made by
negative image that contrasts with the darkly stained histologic examination and cultures. A negative culture
hyphae. Occasionally, when the fungus cavity is result does not exclude the possibility of infection, and
exposed to air, fruiting bodies with conidia can be the most important is direct histopathologic examina-
detected in cytologic specimens; their presence helps tion. Pulmonary zygomycosis in immunosuppressed
confirm the presence of Aspergillus species and can be patients is a progressive disease, usually fatal within
used in further classification. Sheaves or rosettes of a few days or weeks. Successful treatment requires
needle-like, birefringent crystals, representing calcium a high index of suspicion, with diagnosis made
oxalate, may be identified. early, and aggressive therapy with amphotericin B,
surgical excision of infarcted tissues, and decrease of
immunosuppression.
DIFFERENTIAL DIAGNOSIS AND PITFALLS

Since infection with Aspergillus species can cause a R ADIOLOGIC FEATURES


necrotizing pneumonitis, which presents radiologically
as a cavitary lesion, the immediate concern is the pos- Pulmonary zygomycosis presents on CT scan as a seg-
sibility that this mass may represent a squamous cell mental or lobar consolidation with central cavitation.
carcinoma with central necrosis. Although malignant
squamous cells do not resemble fungal hyphae, active
infection by Aspergillus species can cause the surround-
ing lung parenchyma to become quite atypical. Severely ZYGOMYCOSIS – DISEASE FACT SHEET
reactive squamous cells may mimic carcinoma – this
can result in a false positive diagnosis, with potentially Clinical Features
serious consequences for the patient. Unlike most ៉ Most common pathogen in lung infections is Rhizopus species
FNAs that attempt to sample the center of a mass in ៉ Risk factors: neutropenia, hematologic malignancies, burns, HIV
order to maximize recovery of material, FNAs of infection, uncontrolled diabetic ketoacidosis, desferrioxamine
cavitary lesions are directed away from the presumed therapy
necrotic center in favor of the peripheral viable lesional ៉ Propensity for angioinvasion with thrombosis, infarction, and
tissue. In aspergillomas, this is precisely where viable hemorrhage
fungus will be located. Recognition of these branched
Radiologic Features
structures can still be difficult if necrotic debris and
៉ Segmental/lobar consolidation with cavitation
inflammation are also present. Even if close scrutiny
reveals no organisms, in the absence of a positive diag-
Prognosis and Treatment
nosis of carcinoma, silver stains are recommended to
៉ Progressive disease, fatal in a few days in immunosuppressed
completely exclude the possibility of an infectious
patients
agent.
៉ Amphotericin B, excision of infarcted tissues, decrease of
A diagnosis of Aspergillus species is sometimes used immunosuppression
as a wastebasket for all morphologically similar fungi,
such as Fusarium species and Pseudoallescheria boydii.

Ch006-F06731.indd 175 10/26/2006 10:27:32 AM


176 FINE NEEDLE ASPIRATION CYTOLOGY

CYTOPATHOLOGIC FEATURES ACTINOMYCOSIS

Zygomycetes are characterized by their wide (3–25 μm), CLINICAL FEATURES


waxy, ribbon-like hyphae that branch irregularly and
are pauci-septate. Side branches are short and at 90- Actinomyces species are normal constituents of oral
degree angle (Fig. 6-15). Usually associated with promi- cavity flora. The most common is A. israelii, but several
nent acute inflammation and necrosis, these organisms other species are seen in humans. There are three dis-
can be overlooked on Diff-Quik stain and are best seen tinctive syndromes associated with A. israelii: cervico-
on Papanicolaou or H&E stains, while silver stains facial, pulmonary and abdominal actinomycosis.
(GMS) may be used for confirmation.

ZYGOMYCOSIS – PATHOLOGIC FEATURES


ACTINOMYCOSIS – DISEASE FACT SHEET
Cytopathologic Findings
Clinical Features
៉ Zygomycetes class (Rhizopus, Absidia, and Mucor species)
៉ Wide (3–25 μm), waxy, ribbon-like, pauci-septate hyphae with ៉ Risk factors: dental caries and periodontal disease
90-degree branching ៉ Actinomyces species are normal constituents of oral flora
៉ Side branches are short
៉ Background shows marked acute infl ammation and necrosis Radiologic Features
៉ Can be overlooked on Diff-Quik stain ៉ Dense, nodular infiltrates (mimic lung cancer)
៉ Best seen on Papanicolaou stain
Treatment
Ancillary Studies ៉ Penicillin for long period of time
៉ GMS used for confirmation ៉ Surgery to drain the abscess

FIGURE 6-15
Zygomycosis. The Zygomycetes are wide, waxy, ribbon-like hyphae embedded within a prominent acute fibrinopurulent exudate. The inset shows the
pauci-septate hyphae with 90-degree branching. Papanicolaou stain, low power (inset: Papanicolaou stain, high power).

Ch006-F06731.indd 176 10/26/2006 10:27:32 AM


CHAPTER 6 Lung 177

Pulmonary actinomycosis occurs after aspiration of


infective material. Individuals with dental caries or peri- ACTINOMYCOSIS – PATHOLOGIC FEATURES
odontal disease are predisposed to pulmonary actinomy-
cosis. The incidence is not precisely known; the disease Cytopathologic Findings
is uncommon, but not rare. Diagnosis is established by ៉ Gram-positive bacteria, non-acid-fast, non-motile, strict or
transbronchial biopsy, open lung biopsy, or radiology- facultative anaerobes
guided FNA of the nodular cavities. Evaluation of the ៉ Fragments of slender, delicate, beaded filaments with acute-
upper respiratory specimens or samples obtained by angle branching
៉ Background shows suppurative infl ammation
bronchoscopy is of limited value because they may be
contaminated by upper respiratory flora, which nor-
Ancillary Studies
mally contains Actinomyces species. The treatment of
៉ Special stains: Gram, PAS, GMS
choice for actinomycosis is penicillin, given for a long
៉ Modified acid-fast (Fite’s) stain is negative
period of time (6 to 12 months). Shorter treatment is
associated with relapses. Surgery is necessary to drain Differential Diagnosis and Pitfalls
abscesses. If the disease is recognized early and treated
៉ Nocardia species (but Nocardia is acid-fast with Fite’s stain)
appropriately, the prognosis is very good.

RADIOGRAPHIC FEATURES
fragments and tangles of slender, delicate, beaded fi la-
ments with acute-angle branching, usually in a back-
Chest radiograph fi lms show a dense, nodular infiltrate ground of suppurative inflammation (Fig. 6-16). The
that mimics lung cancer. Half of the patients can have equivalent of sulfur granules is accumulations of these
also nodular cavities. organisms with a very dark granular center with
peripherally radiating fi laments. Splendore-Hoeppli
reaction consists of elongated, club-like structures radi-
ating centrifugally from a dense band at the periphery
CYTOPATHOLOGIC FEATURES of the granules. Necrosis and abscess formation may
also accompany these infections.
Actinomyces species are Gram-positive bacteria, non-
acid-fast, non-motile, strict or facultative anaerobes. In
tissues, they grow in microcolonies that appear as DIFFERENTIAL DIAGNOSIS AND PITFALLS
yellow granules in the drainage from actinomycotic
lesions. They are called sulfur granules because of the
yellow color, but their content in sulfur is actually A bacteria morphologically similar to Actinomyces
low. In cytology specimens, the organisms present as species is Nocardia species; however, Actinomyces

FIGURE 6-16
Actinomycosis. The FNA shows clus-
ters of delicate, slender filamentous
organisms. Papanicolaou stain, low
power.

Ch006-F06731.indd 177 10/26/2006 10:27:33 AM


178 FINE NEEDLE ASPIRATION CYTOLOGY

species organisms are slightly larger (1–1.5 μm in


diameter, 20–70 μm in length) than Nocardia species
and branch at acute angles. Most cytologic and histo-
logic stains, including silver stains, can be used to
identify Actinomyces species; however, unlike Nocardia
species, Actinomyces species are not acid-fast when
stained with a modified acid-fast (Fite’s) stain. Other
useful ‘special stains’ include PAS and a modified
Gram stain.

BENIGN NEOPLASMS

HAMARTOMA

CLINICAL FEATURES

Patients are usually asymptomatic and the condition


is discovered incidentally on radiologic images. The
common type is usually in the periphery of the lung.
In the rare endobronchial variant, the patients present
with symptoms of airway obstruction, such as cough
and dyspnea. There is a wide age range, with the peak
age of incidence in the sixth decade. It is relatively rare FIGURE 6-17
in children. Hamartoma: radiologic findings. A well-circumscribed, discrete, ‘coin’
lesion is present in the right upper lobe. This is usually an incidental
finding. This presentation is characteristic for benign pulmonary neo-
plasms such as hamartoma, clear cell (sugar) tumor, granular cell tumor,
R ADIOLOGIC FEATURES and sclerosing hemangioma.

Hamartoma appears as a well-circumscribed, discrete CYTOPATHOLOGIC FEATURES


(‘coin’) lesion (Fig. 6-17). The characteristic ‘popcorn’
calcification can be detected in about 15–30% of cases
on plain radiographs. Pulmonary hamartoma is a tumor-like malformation
composed of both an epithelial and a mesenchymal
component. There is usually a mixture of cartilage,
adipose tissue and some cellular fibrous tissue (mesen-
chymal component), and a non-specific epithelial com-
ponent (usually entrapped lung epithelium) (Fig. 6-18).
The cartilage may be predominant with a chondroid
PULMONARY HAMARTOMA – DISEASE FACT SHEET
matrix and recognizable chondrocytes. When the epi-
thelial component predominates, a diagnosis of malig-
Incidence
nancy may be considered (Fig. 6-19). Careful search for
៉ Uncommon, benign
the mesenchymal component resolves the dilemma.
៉ Peak age of incidence is the 6th decade
The aspirate smears have variable cellularity, with
Clinical Features
metachromatic chondromyxoid stroma on Diff-Quik
stain and fibrillar-myxoid connective tissue with bland,
៉ Usually asymptomatic and discovered incidentally
៉ Rarely presents with symptoms of airway obstruction
oval, or spindle nuclei embedded within the stroma,
similar to benign mixed tumors of the salivary gland.
Radiologic Features This matrix may be the only indication of a cartilage
៉ Well-circumscribed ‘coin’ lesion
component of the tumor. The chondromyxoid matrix is
៉ May have ‘popcorn’ calcifications
less characteristic on Papanicolaou stain and may stain
a pale blue, making it difficult to distinguish it from
Treatment other extracellular elements (Fig. 6-20). In any case,
៉ Surgical excision
the presence of adipose tissue and cartilage in a lung
aspirate should alert one to the possibility of pulmo-
nary hamartoma.

Ch006-F06731.indd 178 10/26/2006 10:27:33 AM


FIGURE 6-18
Hamartoma. The FNA yields a
mixture of fibrillary metachromatic
chondromyxoid stroma and adipose
tissue. Diff-Quik stain, low power.

FIGURE 6-19
Hamartoma: FNA findings. The epithelial component of a hamartoma
consists usually of fl at, honeycomb groups of cytologically bland cells.
These cells can be confused with mesothelial cells or with a malignant
process, such as bronchioloalveolar carcinoma. Diff-Quik stain, medium
power.

FIGURE 6-20
Hamartoma. The mesenchymal com-
ponent with bland, spindle-shaped
nuclei appears very pale with
Papanicolaou stain and can be over-
looked. Papanicolaou stain, high
power.

Ch006-F06731.indd 179 10/26/2006 10:27:34 AM


180 FINE NEEDLE ASPIRATION CYTOLOGY

PULMONARY HAMARTOMA – PATHOLOGIC FEATURES CLEAR CELL (SUGAR) TUMOR – DISEASE FACT SHEET

Cytopathologic Findings Definition


៉ Variable cellularity with epithelial and mesenchymal ៉ Benign tumor of uncertain origin
components
៉ Metachromatic chondromyxoid stroma with embedded bland, Gender and Age Distribution
oval or spindle nuclei (best seen on Diff-Quik stain) ៉ Equal incidence in males and females
៉ Occasionally adipose tissue ៉ Wide age range, mean age is 49 years

Differential Diagnosis and Pitfalls Clinical Features


៉ Carcinoid tumor, bronchioloalveolar carcinoma, small cell ៉ Usually asymptomatic, discovered incidentally
carcinoma
៉ Predominance of epithelial cells may lead to a diagnosis of
Radiologic Features
carcinoma; careful search for the mesenchymal component is
៉ Well-circumscribed ‘coin’ lesion
necessary

Treatment
៉ Surgical excision

DIFFERENTIAL DIAGNOSIS AND PITFALLS

Evaluation of the accuracy of FNA biopsy of pulmo- CYTOPATHOLOGIC FEATURES


nary hamartoma by the College of American Patholo-
gists (CAP) revealed that FNA has a specificity of 78%
with a false positive rate of 22%. The usual false posi- The aspirate is cellular, composed of small irregular
tive diagnoses are carcinoid tumor, adenocarcinoma, clusters or individual, single, polygonal and spindle-
and small cell carcinoma. Contributing factors to erro- shaped cells. The neoplastic cells are cytologically
neous diagnoses are the fact that the cartilaginous and bland. They have abundant clear cytoplasm with
the fibromyxoid components may not be easily identi- ill-defined borders. The cytoplasm can be occasionally
fied in Papanicolaou-stained preparations and the fact vacuolated or granular. The tumor cells are easily
that the reactive bronchial cells (the epithelial compo- stripped of their abundant vacuolated (glycogen-rich)
nent) may look worrisome with large nuclei, prominent cytoplasm, resulting in a very granular, foamy back-
nucleoli, intranuclear invaginations, and multinucle- ground. The nuclei show minimal pleomorphism, with
ation. Careful search for the mesenchymal component fine, evenly distributed chromatin and inconspicuous
helps avoiding diagnostic pitfalls. nucleoli. The nuclei are round–oval or may be only
slightly indented, resembling histiocytes. Naked
(stripped) nuclei are usually present in the background,
as the cells lose their cytoplasm during smearing
(Fig. 6-21). Mitoses and necrosis are usually absent.
CLEAR CELL (SUGAR) TUMOR

CLINICAL FEATURES CLEAR CELL (SUGAR) TUMOR – PATHOLOGIC FEATURES

Cytopathologic Findings
This is a rare benign tumor of uncertain nature, which
៉ Variable cellularity with large irregular clusters of polygonal
is usually asymptomatic and found incidentally. There
and spindle-shaped bland cells
is an equal occurrence in males and females. There is
៉ Cytoplasm is usually clear with indistinct cell borders; may be
a wide age range, from 8 to 73 years, with a mean age vacuolated or granular
of 49 years. The tumor is usually in the lung periphery, ៉ Bland round, oval, or spindle-shaped nuclei
and circumscribed but not encapsulated. There is ៉ Naked nuclei in the background
usually no obvious connection with major airways,
blood vessels, or pleura. Ancillary Studies
៉ Positive for HMB-45 and PAS

Differential Diagnosis and Pitfalls


៉ Metastatic renal cell carcinoma
R ADIOLOGIC FEATURES ៉ Carcinoid tumor, clear cell variant
៉ Granular cell tumor
៉ Metastatic clear cell sarcoma
The tumor presents as a circumscribed, ‘coin’
lesion.

Ch006-F06731.indd 180 10/26/2006 10:27:35 AM


CHAPTER 6 Lung 181

FIGURE 6-21
Clear cell (sugar) tumor. The FNA
yields groups of bland-appearing
cells with round–oval nuclei with
finely distributed chromatin. The
cytoplasm is fine, with indistinct
cell borders, and is frequently
detached from the nucleus during
slide preparation. Note naked,
stripped nuclei in the background.
Diff-Quik stain, high power.

ANCILLARY STUDIES squamous cell carcinoma, adenocarcinoma, small cell


carcinoma, large cell carcinoma, and pleomorphic car-
cinoma. From a practical point of view, based on spe-
Immunohistochemical studies are usually necessary to cific cytologic criteria, lung carcinoma can be divided
confirm the diagnosis. Although the histogenesis of in two main categories: small cell (20% of lung cancer
the ‘sugar’ tumor is still unclear, the tumor cells are cases) and non-small cell carcinoma (NSCLC) (80%).
positive for HMB-45 (similar to angiomyolipoma and This distinction has clinical and prognostic signifi-
lymphangioleiomyomatosis), leading to the speculation cance. There is no age difference between the patients
that they may have the same histogenesis. The clear with small cell carcinoma and those with NSCLC. The
cells are positive for PAS (due to glycogen content). average age at presentation is 66 years, but there is a
Clear cell tumor is negative for epithelial markers. wide age distribution, ranging from younger than 40
years to older than 80 years. Small cell carcinoma is a
rapidly growing and widely metastatic tumor that is
treated primarily with chemotherapy. NSCLC is treated
DIFFERENTIAL DIAGNOSIS AND PITFALLS with surgery, radiotherapy, and chemotherapy, depend-
ing on clinical stage.
Differential diagnosis includes metastatic renal cell
carcinoma, clear cell variant of carcinoid tumor, granu-
lar cell tumor, and metastatic clear cell sarcoma. It may SQUAMOUS CELL CARCINOMA
be extremely difficult to differentiate these entities
by cytomorphology alone. Ancillary studies are often
needed to reach the correct diagnosis. Clear (‘sugar’) CLINICAL FEATURES
cell tumor of lung and metastatic renal cell carcinoma
are both positive for PAS, but renal cell carcinoma is
positive for epithelial markers and negative for HMB- Squamous cell carcinoma is the most common histo-
45, while clear (‘sugar’) cell tumor has the opposite logic type of lung cancer, with an incidence of 35%. It
staining pattern. Granular cell tumor is distinguished is the most common type of lung cancer in smokers.
by S-100 protein reactivity and negative results with
PAS. Clear cell variant of carcinoid is negative for
HMB-45 and positive for neuroendocrine markers SQUAMOUS CELL CARCINOMA – DISEASE FACT SHEET
(synaptophysin, chromogranin).
Incidence
៉ Most common histologic type of lung cancer (35% of all cases)
៉ Most common histologic type in smokers
PRIMARY MALIGNANT TUMORS
Radiologic Features
៉ Presents commonly as a central, cavitary, submucosal lesion

Lung cancer is the leading type of cancer worldwide Prognosis and Treatment
and is the leading cause of mortality from cancer in
៉ Dependent on stage
both men and women. According to the World Health ៉ Surgery, radiotherapy, and chemotherapy
Organization’s international histologic classification,
there are five major histologic types of lung cancers:

Ch006-F06731.indd 181 10/26/2006 10:27:36 AM


182 FINE NEEDLE ASPIRATION CYTOLOGY

R ADIOLOGIC FEATURES
KERATINIZING (WELL-DIFFERENTIATED) SQUAMOUS CELL
CARCINOMA – PATHOLOGIC FEATURES
Squamous cell carcinoma usually presents as a
central, cavitary, submucosal lesion (two-thirds of Cytopathologic Findings
cases). Rarely, it can present as a peripheral lesion ៉ Cohesive sheets and single cells with evidence of keratinization
(one-third). ៉ N/C ratios range from low to high
៉ Dense, waxy, ‘hard’ orangeophilic cytoplasm with clearly defined
borders
៉ Cells with bizarre forms: fiber, strap, tadpole
៉ Nuclei hyperchromatic with inconspicuous nucleoli or pyknotic
CYTOPATHOLOGIC FEATURES (ink dot-like)
៉ Background: necrosis, debris, and ghost cells

Squamous cell carcinoma is a malignant neoplasm that,


Differential Diagnosis and Pitfalls
by definition, has features of squamous differentiation.
៉ Pulmonary infarct, pneumonia, mycetoma
In cytology, squamous differentiation translates as
៉ Metastatic urothelial carcinoma
keratin pearls, twisted keratin strands (Herxheimer ៉ Metastatic squamous cell carcinoma from other sites (in
spirals), partial or complete cytoplasmic rings, and particular, head and neck areas)
bizarre-shaped cells with bright orangeophilic cyto-
plasm on Papanicolaou stain. In well-differentiated
neoplasms, all or almost all of these features can be
seen, while in poorly differentiated cases, they can be
completely absent. Such cases can be diagnosed as nuclear contours, obscured chromatin details, and often
poorly differentiated non-small cell carcinomas. inconspicuous nucleoli. The nuclei can undergo degen-
eration and become pyknotic (ink dot-like). Frequently,
KERATINIZING (WELL- DIFFERENTIATED) SQUAMOUS squamous cell carcinoma undergoes central necrosis
CELL C ARCINOMA and cavitation; hence, the background can show necro-
sis and debris, ghost cells, multinucleated cells, and
FNA of a well-differentiated squamous cell carci- inflammation.
noma usually yields a cellular aspirate characterized by
cohesive sheets, aggregates, or single malignant cells
NON-KERATINIZING (MODERATELY TO POORLY
with evidence of keratinization (Fig. 6-22). There is DIFFERENTIATED) SQUAMOUS CELL C ARCINOMA
usually little nuclear overlapping. The neoplastic cells
can have bizarre shapes (fiber, strap, tadpole cells) (Fig. FNA of a non-keratinizing, moderately to poorly dif-
6-23). They have variable amounts of dense, waxy, ferentiated squamous cell carcinoma also yields a cellu-
‘hard’ cytoplasm with clearly defined cytoplasmic lar specimen with abundant single, discohesive cells or
borders. The N/C ratios range from low to high. The syncytial groups with no cytologic evidence of keratini-
nuclei are pleomorphic, hyperchromatic, with irregular zation (Fig. 6-24). The neoplastic cells are usually

FIGURE 6-22
Well-differentiated (keratinizing)
squamous cell carcinoma. The FNA
yields a cellular material with
malignant squamous cells with
dark, hyperchromatic, irregular
nuclei and variable amounts of
dense, bright orangeophilic or
cyanophilic cytoplasm. Note the
background containing ghost cells
and pyknotic, degenerating nuclei.
Papanicolaou stain, medium power.

Ch006-F06731.indd 182 10/26/2006 10:27:36 AM


CHAPTER 6 Lung 183

FIGURE 6-23
Well-differentiated (keratinizing)
squamous cell carcinoma. The
malignant squamous cells can have
bizarre shapes (strap cells). Note
the dense eosinophilic cytoplasm.
Papanicolaou stain, high power.

NON-KERATINIZING (MODERATELY TO POORLY DIFFERENTIATED)


SQUAMOUS CELL CARCINOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Single, discohesive groups of cells with no or very little
evidence of keratinization
៉ High N/C ratio
៉ Hyperchromatic nuclei with visible nucleoli
៉ Scant to moderate, often vacuolated, cytoplasm

Differential Diagnosis and Pitfalls


៉ Adenocarcinoma
៉ Large cell carcinoma

adenocarcinoma. More, squamous cell carcinoma may


have a finely vacuolated cytoplasm. This feature com-
FIGURE 6-24 bined with prominent nucleoli may lead to confusion
Poorly differentiated (non-keratinizing) squamous cell carcinoma. The with adenocarcinoma. From a practical standpoint, this
FNA shows syncytial groups of cells with a moderate amount of cyano- distinction is not of critical importance because these
philic cytoplasm. The nuclei are hyperchromatic and pleomorphic, with cases can be classified as moderately to poorly differenti-
small nucleoli. Papanicolaou stain, high power.
ated non-small cell carcinomas.

smaller, with high N/C ratios, moderate to scant cyano-


philic cytoplasm, and irregular, pleomorphic nuclei with DIFFERENTIAL DIAGNOSIS AND PITFALLS
irregular, granular chromatin and visible, prominent
nucleoli (Fig. 6-25) (in contrast to a well-differentiated
squamous cell carcinoma, which has dense nuclear Well-differentiated squamous cell carcinoma needs to be
chromatin, inconspicuous nucleoli or pyknotic nuclei). differentiated primarily from: 1) reactive, inflamma-
Since the cytologic features of keratinization are absent tory and infectious processes; 2) metastatic low-grade
in these cases, it is difficult to distinguish them from urothelial carcinoma; and 3) metastatic squamous cell

Ch006-F06731.indd 183 10/26/2006 10:27:37 AM


184 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 6-25
Poorly differentiated (non-
keratinizing) squamous cell carci-
noma. In this aspirate, the cells
have high N/C ratios, with scant,
hard cytoplasm and large, hyper-
chromatic nuclei with prominent
nucleoli. The presence of nucleoli
may suggest the diagnosis of ade-
nocarcinoma. Such cases can be
diagnosed generically as non-small
cell carcinoma. Diff-Quik stain, high
power.

carcinoma from other sites, particularly head and neck cytologic features are correlated with clinical and radio-
primary. logic findings. Many of these specimens will have limited
Because of central necrosis and cavitation, a well- numbers of cells to evaluate. However, cellularity and
differentiated squamous cell carcinoma can be confused cell debris may vary greatly, depending upon the age of
with reactive, inflammatory and infectious processes the infarction. Recent infarctions may show only hemo-
associated with necrosis, such as granulomatous inflam- siderophages and necrotic cellular debris, a potential
mation (fungal [mycetoma], rheumatoid arthritis), source of diagnostic error if confused with a tumor dia-
pneumonia, and pulmonary infarct. thesis. A diagnosis of malignancy in a low cellularity
Several generalities can be made concerning these specimen should be made with extreme caution.
benign conditions: 1) fewer cells are present when com- Rarely, metastatic urothelial carcinoma may mimic
pared to carcinoma; 2) the nuclear details are not so squamous cell carcinoma, particularly the low-grade
distinct or, if so, they are combined with a relatively urothelial neoplasms. In FNA specimens, metastatic
normal N/C ratio; and 3) degeneration is often greater urothelial carcinoma tends to occur as flat sheets with
in these conditions than in carcinomas. Other helpful accompanying individual cells that look as if they have
features are subtle palisading in granulomatous inflam- been pulled from these sheets. These single cells and the
mation and identification of hyphae. A good rule of cells at the periphery of the fragments have elongated
thumb is to avoid making a diagnosis of malignancy unipolar cytoplasmic processes with bland hyperchro-
in cases with few, degenerated and poorly visualized matic nuclei at the apical cytoplasmic pole. These cells
cells. have been termed cercariform cells because of their
Pulmonary infarct with subsequent repair atypia can resemblance to parasitic cercariform larvae.
be a source of false positive diagnoses. Pulmonary infarct Primary lung keratinizing squamous cell carcinoma
may be asymptomatic. It can mimic a lung neoplasm is morphologically similar to primary head and neck
radiographically. The FNA material shows two- squamous cell carcinoma metastatic to the lung. Com-
dimensional sheets of metaplastic squamous cells and parison with previous histologic or cytologic material,
numerous hemosiderin-laden macrophages. The meta- clinical presentation, and radiologic findings are neces-
plastic squamous cells show features of regeneration and sary in such cases.
atypia characterized by enlarged nuclei with pale, vesicu- Moderately to poorly differentiated squamous cell car-
lar chromatin and a prominent chromocenter or small cinoma may be difficult to distinguish from poorly
nucleolus. The presence of atypical squamous cells on differentiated adenocarcinoma and, to a lesser extent,
aspiration smears may result in overdiagnosis, unless the large cell carcinoma.

Ch006-F06731.indd 184 10/26/2006 10:27:38 AM


CHAPTER 6 Lung 185

ADENOCARCINOMA CYTOPATHOLOGIC FEATURES

CLINICAL FEATURES Adenocarcinoma is a malignant neoplasm with glandu-


lar differentiation. In cytology, this translates as cellu-
lar smears with cohesive clusters of cells and acinar
This is the most common type of lung cancer in women
groups. The cell clusters are tridimensional and exhibit
and non-smokers. Most adenocarcinomas arise at the
depth of focus. There is an increased N/C ratio. The
periphery of the lungs. Central adenocarcinomas,
neoplastic cells have round to oval, enlarged nuclei
though uncommon, may be seen. Many peripheral
with irregular nuclear membranes, granular chromatin
tumors reach a substantial size and may actually meta-
(finely or coarsely distributed), and prominent nucleoli
stasize before they cause any symptoms, because major
(which can be multiple). The cytoplasm is scant to
airways are not affected.

R ADIOLOGIC FEATURES
ADENOCARCINOMA – PATHOLOGIC FEATURES

Adenocarcinoma usually presents as a discrete lung mass Cytopathologic Findings


of variable sizes with infiltrative edges (Fig. 6-26). ៉ Cohesive sheets or clusters of cells with depth of focus (three-
dimensional)
៉ Columnar, cuboidal, or polygonal cells with scant/moderate
ADENOCARCINOMA – DISEASE FACT SHEET cytoplasm with vacuolization
៉ Enlarged nuclei with prominent nucleoli
៉ Mitoses and necrosis may be present
Incidence
៉ Most common lung carcinoma in females and non-smokers
Ancillary Studies
៉ Positive for CK, EMA, CEA, CD15, CK7, and TTF-1; negative for
Radiologic Features
CK20
៉ Discrete lung mass with infiltrative edges

Differential Diagnosis and Pitfalls


Prognosis and Treatment
៉ Metastatic adenocarcinoma (breast, stomach, colon, pancreas,
៉ Prognosis depends on the stage of the tumor
gynecologic tract, etc.)
៉ Resection and/or chemotherapy

FIGURE 6-26
Adenocarcinoma: radiologic findings.
There is an infiltrative mass with
indistinct edges in the right lower
lobe. Surgical follow-up showed this
mass to be an adenocarcinoma.

Ch006-F06731.indd 185 10/26/2006 10:27:38 AM


186 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 6-27
Adenocarcinoma. A, The FNA yields a cellular aspirate with tridimensional clusters of malignant cells with scant cytoplasm and large, hyperchromatic
nuclei with prominent nucleoli. B, Abundant tumor cell necrosis can be seen. Papanicolaou stain, high power.

moderate with coarse to fine vacuolization; occasion-


ally, it may be clear. Mitoses and necrosis may also be
seen (Figs 6-27 & 6-28).

ANCILLARY STUDIES

Immunohistochemical studies are necessary to dis-


tinguish primary lung adenocarcinoma from meta-
static adenocarcinoma. In addition to being positive
for cytokeratin markers, carcinoembryonic antigen
(CEA), epithelial membrane antigen (EMA), and CD15,
primary adenocarcinoma of the lung is usually positive
for cytokeratin 7 (CK7) and thyroid transcription
factor-1 (TTF-1), and negative for cytokeratin 20
(CK20).
FIGURE 6-28
Adenocarcinoma. In this aspirate, note the hyperchromatic nuclei with
irregularly distributed, coarse nuclear chromatin and visible nucleoli.
There is marked nuclear pleomorphism. The cytoplasm is almost absent
and the nuclei are crowded together. Compare with normal bronchial
DIFFERENTIAL DIAGNOSIS AND PITFALLS cells in the upper left. This case can be diagnosed generically as non-
small cell carcinoma. Attention to nuclear details helps in distinguish-
ing non-small cell carcinoma from small cell carcinoma at the time of
Primary lung adenocarcinomas need to be differenti- preliminary interpretation in the radiology suite. Diff-Quik stain,
ated from adenocarcinomas from other sites that medium power.
have metastasized to the lung, i.e. breast, gastro -
intestinal tract (stomach, colon), pancreato-biliary
tract, gynecologic tract, etc. If there is a history of BRONCHIOLOALVEOLAR
a previous neoplasm, comparing the current speci- CARCINOMA (BAC)
men with previous material may help in determin-
ing whether the lung mass is primary or metastatic.
In addition, immunohistochemical stains can be This is an uncommon variant of adenocarcinoma
performed on cell block material or alcohol-fi xed (about 2% of all lung malignancies), which, by defini-
slides. tion, has a pure bronchioloalveolar growth pattern

Ch006-F06731.indd 186 10/26/2006 10:27:39 AM


CHAPTER 6 Lung 187

BRONCHIOLOALVEOLAR CARCINOMA – DISEASE FACT SHEET BRONCHIOLOALVEOLAR CARCINOMA – PATHOLOGIC FEATURES

Incidence Cytopathologic Findings


៉ Uncommon; approximately 2% of all lung malignancies ៉ Non-mucinous and mucinous variants
៉ Highly cellular aspirate with bland cuboidal to columnar
Radiologic Features neoplastic cells in papillary clusters or fl at sheets
៉ Discrete nodule, multiple nodules, or pneumonic infiltrate ៉ Nuclear pseudoinclusions and psammoma bodies
៉ Abundant intracellular/extracellular mucin (in the mucinous
variant)
Prognosis and Treatment
៉ Better prognosis than bronchogenic adenocarcinoma
Histopathologic Findings
៉ Non-mucinous variant has better prognosis that mucinous variant
៉ Treatment: surgical (if discrete nodule), chemotherapy (if multiple ៉ Tumor cells grow along the alveolar septa, with no stromal,
nodules, pneumonic infiltrate) vascular, or pleural invasion

Differential Diagnosis and Pitfalls


៉ Reactive pneumocytes, mesothelial cells, or bronchial cells
៉ Clues: high cellularity, lack of terminal bars and/or cilia
with no evidence of stromal, vascular, or pleural inva-
sion. BAC presents radiologically as a single nodule,
multiple nodules, or as a diffuse ‘pneumonic’
infi ltrate.
material will result in flat sheets, acinar arrays, and/or
papillary fragments with depth of focus. The neoplastic
CYTOPATHOLOGIC FEATURES cells may resemble alveolar pneumocytes, mesothelial
cells, or even bronchial cells (Fig. 6-29). The nuclei are
usually enlarged and there is an increase in N/C ratio,
BAC can have either a mucinous or non-mucinous and the nuclear membrane may be slightly irregular
appearance. The non-mucinous type is more common (Fig. 6-30). Nuclear pseudoinclusions and psammoma
and may be difficult to differentiate from bronchogenic bodies may be seen. The mucinous variant of BAC
adenocarcinoma. Aspirate smears are highly cellular consists of bland columnar cells with basally oriented
and composed of bland neoplastic cells (cuboidal or nuclei and abundant apical cytoplasmic mucin (Fig.
columnar), usually in clusters. No terminal bars or cilia 6-31). Mucinous BAC is often multicentric and usually
are identified on these cells. Since BAC’s predominant has a worse prognosis than the non-mucinous
pattern of growth is along the alveolar septa, the FNA variant.

FIGURE 6-29
Bronchioloalveolar carcinoma. The
FNA yields a highly cellular aspirate
with fl at groups of cytologically
bland cells. These cells can be con-
fused with alveolar pneumocytes or
mesothelial cells. The high cellular-
ity is a very helpful clue for a malig-
nant diagnosis. Avoid making a
diagnosis of malignancy from speci-
mens with scant cellularity. Papani-
colaou stain, medium power.

Ch006-F06731.indd 187 10/26/2006 10:27:40 AM


188 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 6-30
Bronchioloalveolar carcinoma.
Careful evaluation of the nuclei
identifies slight nuclear variability
and irregular nuclear contours with
occasional nuclear grooves. Note
also slight depth of focus of this
group of malignant cells. Papanico-
laou stain, high power.

FIGURE 6-31
Bronchioloalveolar carcinoma. In
the mucinous variant of bronchio-
loalveolar carcinoma, the neoplas-
tic cells have moderate to abundant,
vacuolated, mucinous cytoplasm.
Extracellular mucin can be present
in the background. The nuclei are
round–oval and uniform. Diff-Quik
stain, high power.

Ch006-F06731.indd 188 10/26/2006 10:27:41 AM


CHAPTER 6 Lung 189

DIFFERENTIAL DIAGNOSIS AND PITFALLS


SMALL CELL CARCINOMA – PATHOLOGIC FEATURES

The chief mimics of BAC are reactive pneumocytes, Cytopathologic Findings


mesothelial cells, and bronchial cells. Carcinoma should ៉ Highly cellular aspirate
never be diagnosed in aspirate materials with atypical ៉ High N/C ratio, very little cytoplasm, and large hyperchromatic
cells that have terminal bars and/or cilia. A very impor- nuclei with finely granular chromatin and inconspicuous nucleoli
៉ Spectrum of viability
tant clue to the right diagnosis is high cellularity, with
៉ Nuclear molding, crush artifact
neoplastic cells arranged in flat sheets and/or papillary ៉ Individual cell necrosis and tumor diathesis
fragments. Supportive clinical and radiologic findings
are usually necessary for a definitive diagnosis of Ancillary Studies
BAC. ៉ pan-CK, EMA, CK7, TTF-1 (positive in 90% cases);
neuroendocrine markers (positive in 50% of cases); CK20
(negative)

SMALL CELL CARCINOMA Differential Diagnosis and Pitfalls


៉ Carcinoid tumor
៉ Lymphoma
CYTOPATHOLOGIC FEATURES ៉ Poorly-differentiated non-small cell carcinoma

The aspirate smear is very cellular, composed of viable


and degenerating cells dispersed singly or in loose clus-
ters. These tumor cells are very fragile and as such are
often encountered as single cells or stripped hyperchro- In addition, small cell carcinoma shows a spectrum
matic nuclei. The cells have an increased N/C ratio of viability that is very characteristic: viable cells with
with large, round to oval, hyperchromatic nuclei and intact chromatin, smaller more degenerated cells, pyk-
very scant cytoplasm (Fig. 6-32). The nuclear chroma- notic nuclei and ghost nuclei (individual cell necrosis).
tin is hyperchromatic and finely granular, and nucleoli The process of smearing aspirated material often shears
are inconspicuous. Nuclear molding, crush artifact, the scant, delicate cytoplasm from the tumor cells. This,
and mitotic figures are commonly seen (Fig. 6-33). combined with cell necrosis, creates a very granular
While in effusion specimens the malignant cells are background often with tingible-body macrophages. This
small, 1–1.5 times the size of a lymphocyte, in FNA particular background helps to exclude carcinoid from
material they may be up to three times the size of a the differential diagnosis.
lymphocyte. This rather dramatic size difference is
attributed to the sampling of viable tumor without
degeneration or necrosis. Nuclear detail, including the
presence of small distinct nucleoli, is easier to visualize
in these larger cells. While tumor cells will be round
with slight nuclear irregularities in the majority of cases,
occasionally the cells will be small and oval or spindle-
shaped, hence the term ‘oat’ cell carcinoma.

SMALL CELL CARCINOMA – DISEASE FACT SHEET

Incidence and Distribution


៉ 20% of lung carcinomas
៉ Adult smokers; median age of 66 years
៉ More common in men

Radiologic Features
៉ Discrete central mass
៉ May have extensive pulmonary and extrapulmonary metastases at
FIGURE 6-32
diagnosis Small cell carcinoma. The FNA yields a very cellular aspirate; the malig-
nant cells have large, round–oval nuclei with very scant cytoplasm.
Note the characteristic nuclear features: hyperchromatic, speckled
Prognosis and Treatment
chromatin with inconspicuous nucleoli and prominent nuclear molding.
៉ Prognosis poor Apoptotic cells are noted in the background (range of viability) as well
៉ Chemotherapy and radiotherapy as tingible-body macrophages. The cell size is not an important diag-
nostic criterion, as the small cell carcinoma cells reach quite large sizes
in the FNA material. Diff-Quik stain, medium power.

Ch006-F06731.indd 189 10/26/2006 10:27:42 AM


190 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 6-33
Small cell carcinoma. The Papanico-
laou stain highlights the character-
istic nuclear features. Note the
crush artifact and the apoptotic
cells. Papanicolaou stain, high
power.

ANCILLARY STUDIES LARGE CELL CARCINOMA AND


PLEOMORPHIC CARCINOMA
Small cell carcinomas of the lung are positive for cyto-
keratins (pan-cytokeratin, low molecular weight cyto- These are rare variants of non-small cell carcinoma.
keratin, EMA, CK7) and TTF-1 (90% of cases). CD56 They share similar cytologic features that help to
is positive in more than 90% of cases. Neuroendocrine separate them from small cell carcinoma, and will be
markers (chromogranin A, synaptophysin) are positive discussed together.
in half of cases.

DIFFERENTIAL DIAGNOSIS AND PITFALLS LARGE CELL CARCINOMA AND PLEOMORPHIC CARCINOMA –
DISEASE FACT SHEET
Small cell carcinoma can be confused with carcinoid
Incidence
tumor, lymphoma, or a poorly differentiated non-small
៉ Large cell carcinoma: 9% of all lung cancers
cell carcinoma. Attention to cytologic features, in
particular identifying the spectrum of viability and
Radiologic Features
the characteristic background with necrosis, debris,
៉ Large cell carcinoma: often found at lung periphery; large necrotic
and mitoses, helps in excluding carcinoid tumor. The
tumors
nuclear chromatin quality (hyperchromatic, finely
៉ Pleomorphic carcinoma: large, peripheral tumors that often invade
granular, small or inconspicuous nucleolus) and the the chest wall
presence of nuclear molding are key features to exclude
a poorly differentiated carcinoma. The absence of lym- Prognosis
phoglandular bodies excludes a lymphoid process. In ៉ Poor
difficult cases, immunohistochemical stains on the cell
block can be performed.

Ch006-F06731.indd 190 10/26/2006 10:27:43 AM


CHAPTER 6 Lung 191

an organoid arrangement. However, cytologically, this


LARGE CELL CARCINOMA AND PLEOMORPHIC CARCINOMA – tumor has the features of a non-small cell carcinoma
PATHOLOGIC FEATURES and is not confused with a small cell carcinoma. Specifi-
cally, this tumor has visible nucleoli, a moderate amount
Cytopathologic Findings of cytoplasm, and an abundant necrotic background
៉ Large cell carcinoma: with frequent mitoses.
៉ Undifferentiated malignant tumor Pleomorphic carcinoma is a poorly differentiated
៉ It is a diagnosis of exclusion non-small cell carcinoma that contains giant cells and/
៉ Discohesive, large malignant cells with large nuclei and
or spindle cells. The giant cells are large, often multi-
moderate amount of cytoplasm
៉ Variant: large cell neuroendocrine carcinoma
nucleated, and discohesive with emperipolesis of
៉ Pleomorphic carcinoma: neutrophils (Fig. 6-35).
៉ Poorly differentiated non-small cell carcinoma with giant
cells and/or spindle cells
៉ Giant cells are often multinucleated
៉ Emperipolesis of neutrophils
CARCINOID TUMOR
Histopathologic Findings
៉ Pleomorphic carcinoma: should have at least 10% component of
spindle or giant cells CLINICAL FEATURES

Carcinoid tumor is a rare lung neoplasm, with an inci-


dence of 0.4–3%. It has now been recognized that even
a typical carcinoid has the ability to metastasize. For
this reason, this tumor is classified as a malignant
Large cell carcinoma is an undifferentiated malignant
neoplasm. The median age at presentation is 47 years,
tumor that lacks the characteristic cytologic features of
younger than the median age for lung cancer. There are
squamous cell carcinoma, adenocarcinoma, and small
no known risk factors associated with the development
cell carcinoma. It is a diagnosis of exclusion. The aspi-
of carcinoid tumor. Symptoms at presentation depend
rate is cellular, consisting of discohesive large malignant
on the location. A centrally located carcinoid tumor
cells with large nuclei and a moderate amount of cyto-
will present with hemoptysis, cough, dyspnea, pain, or
plasm. The nuclei have irregular contours, unevenly
recurrent pneumonia. A peripherally located lesion is
distributed, coarse chromatin, and prominent nucleoli
usually asymptomatic, discovered incidentally. This
(Fig. 6-34). A variant of this tumor is large cell neuro-
tumor is treated by surgical excision.
endocrine carcinoma, in which neuroendocrine differ-
entiation can be demonstrated by immunochemistry or
electron microscopy. The cell block material may show

FIGURE 6-35
FIGURE 6-34 Pleomorphic carcinoma. Very cellular aspirate with a discohesive popu-
Large cell carcinoma. Large malignant cells with pleomorphic nuclei lation of large, multinucleated malignant cells. Some of the cells have
with irregular, folded nuclear membranes and irregular chromatin. engulfed neutrophils in their cytoplasm, characteristic finding in pleo-
Mitotic figures are easily identified. Papanicolaou stain, high power. morphic carcinoma. Diff-Quik stain, low power.

Ch006-F06731.indd 191 10/26/2006 10:27:43 AM


192 FINE NEEDLE ASPIRATION CYTOLOGY

CARCINOID TUMOR – DISEASE FACT SHEET CARCINOID TUMOR – PATHOLOGIC FEATURES

Incidence Cytopathologic Findings


៉ 0.4% to 3% ៉ Uniform population of cells with moderate, finely granular
cytoplasm
Clinical Features ៉ Round–oval nuclei, with smooth nuclear contours, stippled
៉ Symptomatic if centrally located (cough, dyspnea, hemoptysis, chromatin, and a small, distinct nucleolus
recurrent pneumonia) ៉ Cells can be spindle-shaped (spindle cell carcinoid)
៉ Asymptomatic if peripheral ៉ Background is clean, with occasional naked nuclei (but no
necrosis or mitotic figures)
៉ Atypical carcinoid (difficult diagnosis to make on FNA material)
Radiologic Features
can be suggested if focal necrosis and rare mitoses are seen
៉ Peripheral mass (25%), central tumor (30%), atelectasis (40%)
៉ Normal chest X-ray (5%)
Histopathologic Findings
៉ Atypical carcinoid: 2–10 mitoses/10 HPF or a focus of necrosis
Treatment
៉ Surgical excision
Ancillary Studies
៉ Positive for neuroendocrine markers
៉ Positive for CK7 and TTF-1; negative for CK20

CYTOPATHOLOGIC FEATURES Differential Diagnosis and Pitfalls


៉ Lymphoma
៉ Adenocarcinoma
The aspirate consists of uniform cells arranged in loose ៉ Other spindle cell lesions
groups, larger tissue fragments, or dispersed as single ៉ Sclerosing hemangioma
cells. The larger tissue fragments have a distinctive
architecture, reminiscent of the trabecular or festoon-
ing patterns seen histologically. When the cells are
dispersed individually, they tend to form small linear have scant to moderate, finely granular cytoplasm with
arrays (similar to the single cell filing seen in lobular poorly defined borders (Fig. 6-36). However, it is not
carcinoma of the breast), rosettes, or acini. It is very uncommon that the cytoplasm is stripped from the
characteristic for carcinoid tumor to have uniform, nuclei, resulting in single, dispersed bare nuclei. At low
small cells with very little pleomorphism. These cells power, these naked, dispersed nuclei can be confused

FIGURE 6-36
Carcinoid tumor. The FNA yields a
cellular aspirate composed of
uniform cells arranged in loose
groups or dispersed as single cells.
They have round nuclei and scant,
fine cytoplasm with indistinct cell
borders. The nuclei have smooth
contours, ‘salt and pepper’ chroma-
tin, and a small nucleolus. Note
the acinar arrangement, which can
be misinterpreted as glandular
differentiation. Papanicolaou stain,
medium power.

Ch006-F06731.indd 192 10/26/2006 10:27:44 AM


CHAPTER 6 Lung 193

ANCILLARY STUDIES

To confirm the diagnosis, immunohistochemical studies


using neuroendocrine markers (chromogranin A, syn-
aptophysin, neuron-specific enolase) can be performed
on the cell block. In addition, a combination of CK7,
CK20, and TTF-1 helps to differentiate between pul-
monary carcinoid, gastrointestinal carcinoid, and pan-
creatic endocrine tumors. Pulmonary carcinoid tumors
are negative for CK20 and have a variable expression
of CK7 and TTF-1. A profi le of CK7+/CK20−/TTF-1+
has high specificity (100%) and moderate sensitivity
(50%) for pulmonary carcinoid tumors. TTF-1 expres-
sion is highly specific for pulmonary carcinoid but is
negative in one-third of cases. Gastrointestinal tract
carcinoid and pancreatic endocrine tumors are negative
for TTF-1. Sixty-seven percent of gastrointestinal car-
cinoid tumors are CK7−/CK20− and 20% of them are
FIGURE 6-37
CK7−/CK20+. Thirty-three percent of pancreatic endo-
Spindle cell carcinoid tumor. Cellular aspirate consisting of uniform, crine tumors are CK7+/CK20−, 33% are CK7−/CK20−,
discohesive spindle-shaped cells with oval nuclei and small amounts of
cytoplasm. Note the finely granular, ‘salt and pepper’ chromatin. Papa- and the rest are equally divided between CK7−/CK20+
nicolaou stain, medium power. and CK7+/CK20+ profi les.

with inflammatory cells. The nuclei are small, round to


oval, with smooth nuclear contours, stippled, ‘salt and DIFFERENTIAL DIAGNOSIS AND PITFALLS
pepper’ chromatin (due to uniform dispersion of the
nuclear chromatin), and a small, distinct nucleolus. Lymphoma, especially a low-grade type, enters in the
Occasionally, variable numbers of small, spindle tumor differential diagnosis in cases with abundant stripped,
cells can be seen. In some cases, the spindle cells are the naked nuclei that at low power are misinterpreted as
only component (spindle cell carcinoid). The cytoplasm lymphocytes. Lack of lymphoglandular bodies, presence
and nuclear characteristics of spindle cell carcinoid are of cohesive groups of epithelial cells with moderate wispy
similar to those of typical carcinoid tumor (Fig. 6-37). cytoplasm, and nuclear characteristics favor a carcinoid
Smears have a clean background because the cyto- tumor. Immunohistochemical stains on the cell block for
plasm of these cells, more abundant than in small cell leukocyte common antigen (CD45) and neuroendocrine
carcinoma, remains intact during the smearing process markers will also help the distinction. In addition, the
and there is no necrosis, apoptotic bodies, or mitoses. rosette formation can be misinterpreted as evidence of
Atypical carcinoid tumor is defined as a tumor with glandular differentiation, resulting in an erroneous diag-
a recognizable carcinoid pattern that has 2–10 mitoses/ nosis of adenocarcinoma. Spindle cell carcinoid can be
10 high-power fields or a focus of necrosis. Since these confused with rare spindle cell lesions (leiomyoma, leio-
features may be focal, sampling is critical in correct clas- myosarcoma, neurofibroma) that occur in the lung.
sification of this tumor. For this reason, a specific dis- Low cellularity specimens may be underdiagnosed as
tinction on FNA between typical and atypical carcinoid ‘negative’ when the typical, uniform carcinoid cells are
may not be always possible. The morphology of atypical confused with benign bronchial cells. The lack of termi-
carcinoid tumor is intermediate between typical car- nal bars and cilia should alert the pathologist that these
cinoid and small cell carcinoma. The tumor cells show bland cells are not bronchial cells.
greater cellular pleomorphism, the nuclei can have Sclerosing hemangioma shows also bland, monoto-
modest membrane irregularities, but the nuclear chro- nous, round to polygonal cells. The cell uniformity and
matin remains finely distributed with small nucleoli. A bland cytology can mimic perfectly a typical carcinoid
diagnosis of atypical carcinoid can be suggested on FNA tumor. Immunohistochemical studies for neuroendo-
material if one sees mitotic activity and necrosis in an crine markers performed on the cell block will help in
otherwise typical carcinoid tumor. difficult cases.

R ADIOLOGIC FEATURES METASTASES

Carcinoid tumor can present as a peripheral mass


(25%), as a centrally located tumor (30%), or as atelec- The lung is a very common site for metastases, because
tasis (40%); in 5% of patients, the chest radiograph is the entire blood supply must circulate through the
normal. lungs. Pulmonary metastases can be diagnosed using

Ch006-F06731.indd 193 10/26/2006 10:27:45 AM


194 FINE NEEDLE ASPIRATION CYTOLOGY

METASTASES – DISEASE FACT SHEET METASTASES – PATHOLOGIC FEATURES

Clinical Features Cytopathologic Findings


៉ Lung is a very common site for metastases ៉ In general, a metastatic adenocarcinoma to the lung has
៉ 14–25% of lung FNAs identify metastases cytologic features similar to those of primary lung
៉ Most common metastases to the lung are adenocarcinomas adenocarcinoma
(breast, colon, kidney) and malignant melanoma ៉ Clues to a specific diagnosis include the following:
៉ Although rare, sarcomas can also metastasize to the lung; most ៉ Cells with abundant vacuolated cytoplasm suggest renal cell
common sarcomas encountered in lung FNA are synovial carcinoma
sarcoma, chondrosarcoma, osteosarcoma, and alveolar soft part ៉ Cells with elongated, hyperchromatic, ‘picket-fence’ nuclei
sarcoma suggest colonic adenocarcinoma
៉ Cercariform cells suggest urothelial carcinoma
៉ Discohesive, epithelioid or spindle cells with prominent
nucleoli, intranuclear inclusions, and pigment suggest
melanoma
FNA; indeed, between 14% and 25% of aspirates iden-
tify metastatic disease. The most common metastases
to the lung identified by FNA are adenocarcinomas
(breast, colon, and kidney) and malignant melanoma,
although different series may vary the order of fre- cytologic features can help in pinpointing towards a
quency for metastasis. specific diagnosis (Figs 6-38–6-43). Identification of car-
The cytomorphology of a metastatic adenocarcinoma cinomas as metastatic is often straightforward if clinical
overlaps significantly with that of a primary lung adeno- information and/or prior surgical or cytologic speci-
carcinoma. However, in certain types of tumors, specific mens are available. In cases in which this correlation is

FIGURE 6-38
Metastatic urothelial carcinoma. The aspirate material is cellular, consisting of discohesive cells with a moderate amount of cytoplasm and centrally
placed, hyperchromatic nuclei. Note in the inset, the cercariform cells, with elongated unipolar cytoplasmic processes and nuclei placed at the
apical pole. Diff-Quik stain, low power (inset: Diff Quik stain, high power).

Ch006-F06731.indd 194 10/26/2006 10:27:46 AM


CHAPTER 6 Lung 195

FIGURE 6-39
Metastatic urothelial carcinoma. Flat sheet of cells with a moderate amount of cytoplasm and hyperchromatic, round, slightly irregular nuclei. This
appearance can be confused with squamous cell carcinoma. Finding cercariform cells (inset) is a clue to the diagnosis of metastatic urothelial
carcinoma. Papanicolaou stain, low power (inset: Papanicolaou stain, high power).

Ch006-F06731.indd 195 10/26/2006 10:27:46 AM


196 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 6-40
Metastatic breast carcinoma. The FNA yields a cellular aspirate with discohesive, tridimensional clusters with depth of focus. The cells have the
typical features of an adenocarcinoma, high N/C ratios, hyperchromatic nuclei, and scant to modest amounts of cytoplasm. On cytomorphology
alone, the distinction between primary lung and metastatic breast carcinoma is very difficult. Comparison with previous material (if available) and
immunohistochemical studies on the cell block material are necessary in the majority of cases. Diff-Quik stain, low power.

FIGURE 6-41
Metastatic renal cell carcinoma. The aspirate material is highly cellular, with clusters and discohesive vacuolated cells. Tumor fragments are associ-
ated with small blood vessels and fibrovascular cores. The cells have mildly increased N/C ratios, round regular nuclei with finely granular chromatin,
and abundant finely vacuolated cytoplasm. Most of the cells retain their cytoplasm during smear preparation. Diff-Quik stain, medium power.

Ch006-F06731.indd 196 10/26/2006 10:27:47 AM


CHAPTER 6 Lung 197

FIGURE 6-42
Metastatic colonic adenocarcinoma. The FNA yields highly cellular smears which show large fragments rather than discohesive cells. Nuclei are fairly
uniform and hyperchromatic, but tend to be elongated and basally oriented, almost in a picket-fence array. The inset shows cell block with a small
fragment of tumor with glandular formation and necrosis, both luminal and in the background. Diff-Quik stain, high power (inset: H&E stain).

FIGURE 6-43
Metastatic malignant melanoma. The aspirated material consists of discohesive, somewhat plasmacytoid, single cells. The cells have enlarged nuclei,
with irregular contours, unevenly distributed chromatin, and prominent nucleoli. Note the intranuclear cytoplasmic inclusion, a non-specific but
common finding in malignant melanoma. The inset shows a binucleated cell with ‘mirror image’ nuclei. Papanicolaou stain, high power (inset:
Papanicolaou stain, high power).

Ch006-F06731.indd 197 10/26/2006 10:27:48 AM


198 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 6-44
Metastatic osteosarcoma. Cellular aspirate with discohesive, bizarre-shaped, multinucleated malignant cells. Note the eosinophilic osteoid matrix
material, a useful clue towards the correct diagnosis. Clinical history is critical in such cases. A, Diff-Quik stain, medium power. B, Papanicolaou
stain, medium power.

not possible, immunohistochemical stains need to be Fritscher-Ravens A, Sriram PV, Topalidis T, et al. Diagnosing sarcoidosis
performed on the cell block material or alcohol-fixed using endosonography-guided fine-needle aspiration. Chest 2000;118:
928–935.
slides. Wildi SM, Judson MA, Fraig M, et al. Is endosonography guided fine needle
Primary pulmonary sarcomas are rare tumors; the aspiration (EUS-FNA) for sarcoidosis as good as we think? Thorax
most common is leiomyosarcoma, which may arise from 2004;59:794–799.
the pulmonary vasculature. Because of the rarity of Wu JJ, Rashcovsky Schiff K. Sarcoidosis. Am Fam Physician 2004;70:
312–322.
primary mesenchymal tumors, metastatic sarcomas
are usually accurately identified (Fig. 6-44). Synovial
sarcoma, chondrosarcoma, alveolar soft part sarcoma, Tuberculosis
and osteosarcoma are just some of the types of sarcomas Maygarden SJ, Flanders E. Mycobacteria can be seen as ‘negative images’
that can metastasize to the lungs. Both primary and in cytology smears from patients with acquired immunodeficiency syn-
secondary sarcomas that are composed of spindle cells drome. Mod Pathol 1989;2:239–243.
Niederman MS, Sarosi GA, Glassroth J. Respiratory Infections, 2nd ed.
will show single, discohesive cells and/or clusters of Philadelphia: Lippincott Williams & Wilkins, 2001.
cells. The individual cells often have fragile cytoplasm Palmer PES. The Imaging of Tuberculosis with Epidemiological, Pathologi-
and granular to coarse chromatin, and there are occa- cal, and Clinical Correlation. Berlin: Springer-Verlag, 2002.
sional bizarre multinucleated cells. The sarcomas can Powers CN. Diagnosis of infectious diseases: a cytopathologist’s perspec-
tive. Clin Microb Rev 1998;11:341–365.
be differentiated from sarcomatoid carcinomas and Silverman JF, Holter JF, Berns LA, et al. Negative images due to clofazimine
melanoma by immunostaining. crystals simulating MAI infection in a bronchoalveolar lavage specimen.
Diagn Cytopathol 1993;9:534–540.

Fungal Infections
SUGGESTED READINGS
Chen KTK. Cytodiagnostic pitfalls in pulmonary coccidioidomycosis. Diagn
Introduction Cytopathol 1995;12:177–180.
Niederman MS, Sarosi GA, Glassroth J. Respiratory Infections, 2nd ed.
Bonfiglio TA. Cytopathologic interpretation of transthoracic fine-needle Philadelphia: Lippincott Williams & Wilkins, 2001.
biopsies. Masson Monographs in Diagnostic Cytopathology. New York: Raab SS, Silverman JF, Zimmerman KG. Fine-needle aspiration biopsy of
Masson Publishing USA, 1983. pulmonary coccidioidomycosis. Spectrum of cytologic findings in 73
Mullan CP, Kelly BE, Ellis PK, et al. CT-guided fine-needle aspiration of patients. Am J Clin Pathol 1993;99:582–587.
lung nodules: effect on outcome of using coaxial technique and imme- Powers CN. Diagnosis of infectious diseases: a cytopathologist's perspec-
diate cytological evaluation. Ulster Med J 2004;73:32–36. tive. Clin Microbiol Rev 1998;11:341–365.
Stewart CJ, Stewart IS. Immediate assessment of fine needle aspiration Sarosi GA, Davies SF. Fungal Diseases of the Lung, 3rd ed. Philadelphia:
cytology of lung. J Clin Pathol 1996;49:839–843. Lippincott Williams &Wilkins, 2000.
Wheat LJ, Goldman M, Sarosi G. State-of-the-art review of pulmonary
fungal infections. Semin Respir Infect 2002;17:158–181.
Sarcoidosis
Hamartoma
Annema JT, Veselic M, Rabe KF. Endoscopic ultrasound-guided fine-needle
aspiration for the diagnosis of sarcoidosis. Eur Respir J 2005;25: Hughes JH, Young NA, Wilbur DC, Renshaw AA, Mody DR. Fine-needle
405–409. aspiration of pulmonary hamartoma: a common source of false-positive

Ch006-F06731.indd 198 10/26/2006 10:27:49 AM


CHAPTER 6 Lung 199

diagnoses in the College of American Pathologists Interlaboratory Com- Carcinoid Tumor


parison Program in Nongynecologic Cytology. Arch Pathol Lab Med
2005;129:19–22. Anderson C, Ludwig ME, O’Donnell M, Garcia N. Fine needle aspiration
cytology of pulmonary carcinoid tumors. Acta Cytol 1990;34:505–510.
Cai YC, Banner B, Glickman J, Odze RD. Cytokeratin 7 and 20 and thyroid
Clear Cell (Sugar) Tumor transcription factor 1 can help distinguish pulmonary from gastrointes-
tinal carcinoid and pancreatic endocrine tumors. Hum Pathol 2001;
Nguyen GK. Aspiration biopsy cytology of benign clear cell (‘sugar’) tumor 32:1087–1093.
of the lung. Acta Cytol 1989;33:511–515. Fekete PS, Cohen C, DeRose PB. Pulmonary spindle cell carcinoid. Needle
aspiration biopsy, histologic and immunohistochemical findings. Acta
Cytol 1990;34:50–56.
Primary Malignant Tumors Gal AA, Nassar VH, Miller JI. Cytopathologic diagnosis of pulmonary
sclerosing hemangioma. Diagn Cytopathol 2002;26:163–166.
Detterbeck FC, Rivera MP, Socinski MA, Rosenman JG. Diagnosis and Treat- Nicholson SA, Ryan MR. A review of cytologic findings in neuroendocrine
ment of Lung Cancer. An Evidence-Based Guide for the Practicing Clini- carcinomas including carcinoid tumors with histologic correlation.
cian. Philadelphia: WB Saunders, 2001. Cancer (Cancer Cytopathol) 2000;90:148–161.
Linder J. Lung cancer cytology. Something old, something new. Am J Clin
Pathol 2000;114:169–171.
Liu J, Farhood A. Immunostaining for thyroid transcription factor-1 on Metastases
fine-needle aspiration specimens of lung tumors: a comparison of direct
smears and cell block preparations. Cancer (Cancer Cytopathol) Flint A, Lloyd RV. Colonic carcinoma metastatic to the lung: cytologic
2004;102:109–114. manifestations and distinction from primary pulmonary adenocarci-
Silverman JF, Weaver MD, Shaw R, Newman WJ. Fine needle aspiration noma. Acta Cytol 1992;36:230–235.
cytology of pulmonary infarct. Acta Cytol 1985;29:162–166. Hummel P, Cangiarella JF, Cohen JM, et al. Transthoracic fine-needle
Travis WD, Colby TV, Corrin B, et al. World Health Organization Interna- aspiration biopsy of pulmonary spindle cell and mesenchymal lesions.
tional Classification of Tumours. Histological Typing of Lung and Pleural A study of 61 cases. Cancer (Cancer Cytopathol) 2001;93:187–198.
Tumours, 3rd ed. Berlin: Springer Verlag, 1999.
Travis WD. Pathology of lung cancer. Clin Chest Med 2002;23:65–81.

Ch006-F06731.indd 199 10/26/2006 10:27:50 AM


7 Mediastinum
Maureen F Zakowski • Anil V Parwani

ganglioneuroblastoma, lymphoma, germ cell tumors,


INTRODUCTION and mesenchymal tumors.
In adults, metastatic lesions are the most common
tumors aspirated, most often small cell carcinoma of the
Fine needle aspiration (FNA) cytology is commonly lung, followed by neurogenic tumors, nerve sheath
used to investigate space-occupying lesions of the medi- tumors, paragangliomas, cysts, thymomas, germ cell
astinum. As with many other body sites sampled by tumors, and endocrine tumors such as thyroid, parathy-
FNA, experience on the part of the cytopathologist roid, and carcinoid. Mesechymal tumors occur infre-
is essential in the correct identification of lesions in quently in the adult mediastinum and one-half of these
this area. Knowledge of the anatomy of the mediasti- are endothelial in origin.
num and familiarity with the normal structures that The FNA method for sampling the mediastinum is
it contains is necessary for the correct interpreta- the same as that for the lung, with the same contraindi-
tion of pathologic conditions. Although a variety of cations to the procedure and a lower complication rate.
lesions, reactive and neoplastic, benign and malignant, The highest rate of complications is found in the middle
occur, some of which are cystic (foregut cysts, bron- mediastinum, probably due to the presence of the great
chogenic cysts, neuroenteric cysts, and pericardial vessels. Complications include pneumothorax, hemo-
cysts), this discussion will not focus on the predomi- thorax, local hemorrhage, and pain. Mortality is rare.
nantly benign cystic lesions of the mediastinum. Sensitivity and specificity are good, but vary with the
Additionally, tumors originating from the thyroid and series reported. The overall sensitivity and specificity of
parathyroid which may be encountered in the medias- FNA versus core biopsy are nearly identical, but some
tinum are not discussed here, but are the subject of investigators prefer core biopsy in cases of suspected
Chapter 2. mesenchymal lesions because of increased accuracy of
tumor typing and availability of more tissue for ancil-
lary studies.
FNA can distinguish benign from malignant lesions
and most often accurately subtype the malignancy. Par-
MEDIASTINUM ticular diagnostic challenges are seen with spindle cell
lesions, perhaps due to their rarity and lack of experi-
ence on the part of pathologists. Other areas of challenge
The mediastinum is the middle compartment between include the subclassification of small cell malignancies,
the right and left thoracic cavities; it extends from the including the misdiagnosis of small cell carcinoma for
thoracic inlet cranially to the diaphragm caudally. It lymphoma and the separation of the germ cell tumors
extends from the sternum anteriorly to the spine pos- from adenocarcinoma. False positive diagnoses are rare
teriorly, and between the pleural cavities laterally and and false negative rates here, as in other sites, are due
from the diaphragm up to the thoracic inlet (Table 7-1). to sampling errors and fibrotic or necrotic conditions.
The majority of lesions of the mediastinum, both benign The unsatisfactory rate as presented in the literature
and malignant, present as asymptomatic masses found is approximately 10%. Studies have found that a single
on chest radiography. When symptomatic, these lesions needle pass is sufficient in approximately half the
are a result of impingement of vital structures. Pain, patients, but that as many as three biopsies may be
dyspnea, cough, or superior vena cava syndrome may needed in the remaining half.
result from involvement of these structures. Metastases Age is not a factor in performing the FNA of the
are more common than primary tumors in the adult mediastinum; so pediatric lesions can be easily aspirated
population. as well. Most aspirations are done under radiologic
Age plays an important role in the correct diagnosis guidance, with computerized tomography (CT) pre-
of tumors of this area because very different lesions are ferred over fluoroscopy. Ultrasound-guidance is often
expected in childhood as compared to adulthood (Table employed in the anterior mediastinum. Adults, for
7-2). The majority of mediastinal lesions in childhood example, have a much lower incidence of neuroblas-
are primary malignant tumors. Frequent here are the toma, ganglioneuroblastoma, and mesenchymal lesions
neurogenic tumors, specifically neuroblastoma and than do children. Metastases, while they do occur, are
201

Ch007-F06731.indd 201 10/26/2006 10:28:49 AM


202 FINE NEEDLE ASPIRATION CYTOLOGY

TABLE 7-1 TABLE 7-3


The Mediastinum Location of Mediastinal Lesions by
Compartment in Adults
• The middle space between the right and left thoracic
cavities Superior
• Boundaries:
• From thoracic inlet above to diaphragm below Metastases (in all compartments)
• From sternum in front to spine in back Thymoma
• Majority of space-occupying lesions are asymptomatic Thymic cysts
• FNA is an excellent method for sampling the Thyroid lesions
mediastinum Parathyroid neoplasms
Lymphoma

Anterior Middle Posterior

Thymoma Pericardial cysts Neurogenic tumor


TABLE 7-2 Thymic cysts Bronchogenic Schwannoma
Thyroid lesion cysts Neurofibroma
Differential Diagnosis of Mediastinal Masses Parathyroid Lymphoma Ganglioneuroma
neoplasm Metastases Ganglioneuroblastoma
Pediatric Adults Lymphoma Malignant peripheral
Paraganglioma nerve sheath
Neurogenic: Metastases Angioma tumor
Neuroblastoma Neurogenic: Lipoma Neuroblastoma
Ganglioneuroblastoma Nerve sheath Paraganglioma
Lymphoma Paraganglioma Gastroenteric cysts
Cysts Cysts Lymphoma
Germ cell Thymoma Metastases
Mesenchymal Germ cell
Thymoma (unlikely) Mesenchymal
Carcinoid (unlikely) Endocrine:
Thyroid
Parathyroid
Carcinoid
15% of the cases were unsatisfactory and a 6% discor-
dance rate between cytology and histology was found.
This was mostly due to the misclassification of carcino-
mas. They also encountered difficulty in separating
infrequent in the pediatric population, and almost Hodgkin from non-Hodgkin lymphoma.
always follow a known primary. Geisinger (1995) divided mediastinal tumors into
Dividing the mediastinum into compartments is categories by morphologic pattern. As expected, small
helpful in determining which lesions may be encoun- cell patterns were usually seen in lymphomas, carci-
tered (Table 7-3). The mediastinum can be considered noid, and small cell carcinoma. Polygonal or epithelioid
to contain several compartments: the superior medias- patterns were seen in thymomas, germinomas, embryo-
tinum, the inferior mediastinum, and the middle medi- nal carcinoma, and metastatic carcinoma. Powers et al
astinum. The inferior mediastinum is further divided (1996) found that 71% of their 189 cases were neo-
into the anterior and posterior compartments. plastic. Interestingly, most of the primary tumors were
The superior mediastinum contains the thymus, thymoma and lymphoma, and the majority of the metas-
lymph nodes, thyroid, and parathyroid; therefore, tases were small cell carcinoma from the lung. The
thymoma, carcinoid tumor, lymphoma, goiter, thyroid remainder of the cases equally divided between un-
and parathyroid adenomas, and various cysts can be satisfactory and non-neoplastic. The sensitivity in this
found. The anterior mediastinum is the site for tumors series was 87%; specificity, 88% for neoplasms and
and cysts of the thymus as well as germ cell tumors. 82% for distinguishing benign from malignant. The
Lymphoid tissue is present in all compartments and positive predictive value for tumors was 97%. There
lymphoma may be encountered in any part of the medi- were three false positive diagnoses in this group.
astinum. The middle mediastinum is composed mainly As the majority of mediastinal FNAs are done under
of the heart and great vessels, and atrial myxomas and radiologic guidance, the benefit of immediate assess-
associated cardiac lesions may be found here. Neuro- ment and feedback to the operator by an experienced
genic tumors, both benign and malignant, are most com- cytopathologist or cytotechnologist is obvious. It is also
monly located in the posterior mediastinum. especially important when considering mediastinal
Singh et al (1997) examined and reported on 189 pathology, because of the frequency of lymphomas
cases of mediastinal FNAs. In their series, less than half here. On-site assessment can assure material is sent for
of the patients had surgical follow-up. Approximately flow cytometry or additional molecular studies such as

Ch007-F06731.indd 202 10/26/2006 10:28:50 AM


CHAPTER 7 Mediastinum 203

needed for germ cell tumor analysis. Careful teamwork


involving cytology and interventional radiology can THYMOMA – DISEASE FACT SHEET
make the difference between a satisfactory specimen
and the need to re-biopsy the patient. Treatment can Definition
be undertaken when the results of the FNA are un- ៉ A primary neoplasm of thymic epithelial cells which is associated
equivocal, and can eliminate the need for surgery in with lymphocytes, predominantly immature T cells
cases of inoperable disease. As always, ancillary studies
on cytology material can be done to support diagnoses, Incidence
and material such as cell blocks and liquid-based prepa- ៉ Most common primary tumor of the anterior mediastinum
rations lend themselves well to immunoperoxidase, ៉ Incidence: 0.15 cases per 100,000
៉ 25% of all primary mediastinal tumors
molecular, and genetic analyses.
The cytomorphologic appearances of cysts and
Gender and Age Distribution
inflammation of the mediastinal compartments are
៉ Males = females
similar to these conditions at any other site and will not
៉ Most occur in adults, with a mean age of 50 years.
be further discussed. Chronic sclerosing mediastinitis
៉ Rare in children
can be confused with tumors having a significant scle-
rotic or collagenous component, such as nodular scleros- Clinical Features
ing Hodgkin disease. This condition, however, would
៉ Most are asymptomatic
probably not yield much material from an aspirate, and ៉ Some patients present with chest pain, cough, dyspnea, or
would likely need surgical biopsy for confirmation. superior vena cava syndrome
៉ Myasthenia gravis is associated with thymoma and is more
common in women
៉ Often large (5–10 cm) at presentation

THYMOMA AND THYMIC LESIONS


Prognosis and Treatment
៉ Mixed: associated with aggressive behavior
៉ Spindle: associated with better prognosis
THYMOMA ៉ Cytologic appearance does not predict behavior
៉ Prognosis dependent upon capsular invasion
៉ Generally responsive to chemotherapy
CLINICAL FEATURES

Thymoma – a primary neoplasm of thymic epithelial


cells, with secondary reactive mature T lymphocytes –
is the most common neoplasm of the anterior mediasti-
num in adults, with a median age of 50 years. Thymomas
can also be found in the superior middle mediastinum,
THYMOMA – PATHOLOGIC FEATURES
but rarely in the posterior. The various classification
systems depend on the proportion of epithelial cells and Cytopathologic Findings
lymphocytes, but it is difficult to apply a classification
៉ Usually well encapsulated with fibrous septations
system to these tumors using only FNA material. Most ៉ Cystic change common
often these masses are asymptomatic, and are discov- ៉ Some have components that are necrotic and hemorrhagic
ered on incidental chest films. Some patients may expe- ៉ Dual population of epithelial and lymphoid components in
rience symptoms from mass lesions such as cough, variable proportions
dyspnea, and chest pain. A subset of patients may come ៉ Epithelial cells: bland nuclei, smooth contour; nucleoli
to medical attention because of associated paraneoplas- prominent but not macro; intranuclear invaginations can occur;
tic syndromes such as myasthenia gravis. few mitoses; +/− rosettes and Hassall’s corpuscles; spindle and
clear cell variants
៉ Lymphocytes: small and mature-appearing; polymorphic pattern
possible with immature lymphocytes, clefting, chromocenters,
and nucleoli
CYTOPATHOLOGIC FEATURES
Ancillary Studies
Since thymoma is a neoplasm with thymic epithelial ៉ Pancytokeratin, cytokeratin 5/6
៉ CD5 is usually positive in thymic carcinomas
cells and secondary reactive mature T lymphocytes, a
correct diagnosis of thymomas is dependent on the
Differential Diagnosis and Pitfalls
recognition of the two cell types, epithelial and lym-
៉ If the lymphocytic component predominates, the lesion may be
phocytic. Either component can dominate. Multiple
mistaken for a lymphoproliferative disorder
classification systems have been used to classify thymo-
៉ If the epithelial component predominates, the lesion may be
mas. A common classification used is that of Bernatz mistaken for a metastatic carcinoma
et al., which divides thymoma into four categories: 1)
epithelial predominant, 2) lymphocytic predominant,

Ch007-F06731.indd 203 10/26/2006 10:28:50 AM


204 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 7-1
Thymoma. The classic bimodal pop-
ulation of neoplastic epithelial and
non-neoplastic lymphoid elements.
The cluster of epithelial cells is
cohesive in the background of scat-
tered lymphocytes, with occasional
single cells. The neoplastic cells are
variable in size and shape, with
usually scant to moderate amounts
of cytoplasm. Nuclei are bland with
often-conspicuous nucleoli. Diff-
Quik stain, low magnification.

3) mixed lymphoepithelial, and 4) spindle cell types.


Suster and Moran (1999) have proposed a classification
system with three categories (thymoma, atypical
thymoma, and thymic carcinoma). A well-character-
ized prognostic factor for predicting the behavior of a
thymoma is if it invades into or through the capsule.
More recently, the World Health Organization has pro-
posed that the classification of thymomas and thymic
carcinomas should be as type A thymoma (spindle cell,
medullary), type B thymoma (B1 thymoma [lympho-
cyte-rich, lymphocytic, predominantly cortical, organ-
oid], B2 thymoma [cortical], B3 thymoma [epithelial,
atypical, squamoid, well-differentiated thymic carci-
noma]), type AB thymoma (mixed), and type C
thymoma (thymic carcinoma).
The smears obtained from a thymoma are often cel-
FIGURE 7-2
lular with a characteristic dual (bimodal) population of
Thymoma. Split screen showing cohesive fragments of epithelial cells
cells: the classic neoplastic epithelial cells and the non- on the left with cystic spaces similar to what is seen in histology. Few
neoplastic lymphoid cells (thymocytes) (Fig. 7-1). The lymphocytes are visible among epithelial cells on the right. Papanico-
epithelial cells of thymoma are variable in size and laou stain, low and high magnification.
shape, with usually scant to moderate amounts of
cytoplasm. Nuclei are bland with conspicuous nucleoli.
Mitoses are often not observed (Fig. 7-2). Spindle cell A predominance of lymphocytes or epithelial cells
morphology is common in the epithelial component of may sometimes be obvious. Because malignancy here is
thymomas, and, when plentiful, usually signifies a good dependent on the demonstration of vascular or capsular
prognosis. invasion, cytology is not useful in making the diagnosis
The epithelial cells, even in cases of lymphocyte- of malignant thymoma (Fig. 7-3). Only when the cyto-
predominant thymoma, will demonstrate significant logic atypia is sufficient to categorize a tumor here as
cohesion. Clusters of epithelial cells can be seen with thymic carcinoma, should the word malignant be used
interspersed loosely arranged lymphocytes surrounded in the diagnosis. ‘Suspicious for malignancy’, ‘malig-
by vascular structures (Fig. 7-1). This arrangement of nancy cannot be ruled out’, and ‘satisfactory for evalu-
vessels is similar to that seen in hepatocellular carci- ation consistent with thymoma’ are some of the phrases
noma. The epithelial cells are often arranged in such a that are useful when diagnosing a thymoma using cyto-
way to suggest cystic structures (Fig. 7-2). logic material.

Ch007-F06731.indd 204 10/26/2006 10:28:50 AM


CHAPTER 7 Mediastinum 205

FIGURE 7-3
Malignant thymoma. Cellular smear
from a patient with a malignant
thymoma. There is a predominance
of epithelial cells. Because malig-
nancy here is dependent on the
demonstration of vascular or capsu-
lar invasion, cytology alone is not
useful in making the diagnosis of
malignant thymoma. Thymoma with
a predominant epithelial component
may be mistaken for a metastatic
carcinoma. Diff-Quik stain, low
magnification.

ANCILLARY STUDIES

The thymoma lymphocytes are mostly small mature T


cells in a polymorphous pattern with some immature
cells and mitotic figures. Immunocytochemical stains for
keratin can demonstrate the epithelial cells when they
are not obvious and thymoma is suspected. Many of the
thymic carcinomas are positive for CD5, while germ cell
tumors and metastatic carcinomas are negative.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

For an accurate diagnosis of thymoma, the characteris- FIGURE 7-4


tic dual population of epithelial and lymphoid elements Thymic carcinoma. Note the presence of large obviously malignant cells
must be recognized. If the lymphocytic component is with pleomorphic and hyperchromatic nuclei. Differential diagnosis
predominant but, due to sampling the epithelial com- here must include metastatic disease. Papanicolaou stain, low
magnification.
ponent, is not recognized, the thymic lesion may be
mistaken for a lymphoproliferative disorder. Similarly,
if the epithelial component is predominant, thymoma THYMIC CARCINOMAS
may be mistaken for a metastatic non-small cell carci-
noma (Fig. 7-3). Metastatic thymomas are cytologically
similar to the primary tumor. Hassall’s corpuscles When necrosis, marked atypia, or epithelial mitosis is
(squamous pearls) can be present, but are rarely seen significant, thymic carcinomas must be considered.
and are not necessary for the diagnosis. Care must be Thymic carcinomas are aggressive epithelial malignan-
taken not to mistake Hassall’s corpuscles for metastatic cies. They are exceedingly rare, and are most frequent
squamous cell carcinoma. in middle-aged men. Patients often present with cough,
Unlike in Hodgkin disease, eosinophils are not a dyspnea, and chest pain and weight loss.
feature of this tumor. When the number of lymphocytes In contrast to thymomas, thymic carcinomas are
is large, lymphoma must be included in the differential cytologically atypical with features of malignancy (Figs
diagnosis and excluded by ancillary studies. 7-4 & 7-5). The most common type of thymic carcinoma

Ch007-F06731.indd 205 10/26/2006 10:28:51 AM


206 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 7-5
Well-differentiated thymic carcinoma. Cohesive fragments of neoplastic cells with few lymphocytes in the background. Occasional cells have a
spindled morphology. The nuclei are moderately sized, with predominantly coarse, granular chromatin. A, Diff-Quik stain, low magnification.
B, H&E stain, high magnification.

is squamous cell carcinoma, but almost any subtype of


carcinoma, including small cell, clear cell, carcinosar- MEDIASTINAL GERM CELL TUMORS – DISEASE FACT SHEET
coma, and lymphoepithelioma-like, can be seen. Epithe-
lial markers are useful to confirm the epithelial origin Definition
of the tumors when the differential diagnosis includes ៉ The mediastinum is a common site of extragonadal germ cell
melanoma and lymphoma. The differential diagnosis of tumors, which are thought to arise from primitive germ cells
thymic carcinomas includes embryonal carcinoma and that failed to migrate completely during embryonic
metastatic carcinomas. development
Ruling out metastasis must prove primary origin in
Incidence
the thymus from a distant site. Germ cell tumors can
៉ The mediastinum is a common site of extragonadal germ cell
have a mixture of epithelial cells and lymphocytes, but
tumors
the epithelial component here is outright malignant.
៉ 25% of all primary mediastinal tumors in children are germ cell
Treatment is dependent on tumor stage and grade. Com- tumors
plete surgical resection is the primary treatment. ៉ 15% of all primary mediastinal tumors in adults are germ cell
tumors
៉ Mature cystic teratoma is the most common mediastinal germ cell
tumor
THYMIC FOLLICULAR HYPERPLASIA
Gender and Age Distribution
៉ Men more than women, except for mature cystic teratoma, which
Thymic follicular hyperplasia with an increased has equal incidence in men and women
number of follicles is associated with myasthenia gravis
and other autoimmune diseases. True thymic hyper- Clinical Features
plasia is a marked increase in the size of the thymus, ៉ Rapid growth and invasion of the tumor result in symptoms
both epithelial and lymphocytic components. This con- ៉ Patients may present with chest pain, dyspnea, superior
dition usually occurs in children and can be difficult to vena cava syndrome, cough, weight loss, night sweats, and
separate from normal thymic tissue on cytology. dysphagia.
៉ Elevated β-HCG (choriocarcinoma, embryonal carcinoma, some
seminomas) and AFP (yolk sac tumors and embryonal carcinoma)
in serum

GERM CELL TUMORS Prognosis


៉ Non-seminomatous mediastinal germ cell tumors are more
aggressive and metastasize earlier than mediastinal
CLINICAL FEATURES seminomas

Only a small percentage of primary germ cell tumors


arise in the mediastinum, but this site accounts for
the majority of all extragonadal germ cell tumors

Ch007-F06731.indd 206 10/26/2006 10:28:52 AM


CHAPTER 7 Mediastinum 207

(50% to 70%). They are more common in children


than in adults. The anterior mediastinum is the pre- MEDIASTINAL GERM CELL TUMORS – PATHOLOGIC FEATURES
ferred site for these tumors and a metastasis to the
mediastinum from the gonads is far more common than Cytopathologic Findings
a primary tumor here. Mediastinal germ cell tumors ៉ Teratoma:
account for 1–15% of adult anterior mediastinal tumors. ៉ Tumor is large, calcified, cystic, and filled with grumous
A gonadal primary must always be excluded when granular material
៉ An FNA smear may show these components and skin
making the diagnosis of primary mediastinal germ cell
structures, gastrointestinal, respiratory, and cartilaginous
tumor. Males are far more likely to have mediastinal
material
germ cell tumors than are females. The germ cell ៉ Foreign body granulomas are common in response to the
tumors that occur in the mediastinum include semi- keratin found in teratomas
noma, endodermal sinus tumor (yolk sac tumors), ៉ Hair is not often seen in a cytology specimen, but short
embryonal carcinoma, and teratomas. Mature terato- strands can sometimes be noted
mas represent the majority of mediastinal germ cell ៉ Seminoma:
tumors (60% to 70%). The remaining mediastinal ៉ Tumor cells are infiltrated by reactive lymphocytes
germ cell tumors are divided between seminomas ៉ Plasma cells are common
(40%) and non-seminomatous germ cell tumors (60%). ៉ Poorly cohesive, and appears malignant at first glance
៉ Hyperchromatic enlarged nuclei are seen with very consistent,
Most of the malignant germ cell tumors are symptom-
prominent nucleoli; cytoplasm is clear
atic, but only about half of the teratomas produce
៉ An often-reported feature is the striped or ‘tigroid’ pattern in
symptoms. the background seen in air-dried smears
៉ Granulomas and multinucleated giant cells are common
៉ Yolk sac tumor:
៉ Irregular, large, cohesive, 3-D balls and papillae, few single
CYTOPATHOLOGIC FEATURES cells
៉ Large cells with abundant vacuolated cytoplasm
៉ Schiller-Duval bodies: glomeruloid structures formed by the
M ATURE CYSTIC TERATOMA invagination of malignant cells into an empty space
៉ Hyaline globules: large, round, PAS-positive intracytoplasmic
Mature cystic teratoma is the most common medias- inclusions containing AFP (also free in background)
tinal germ cell tumor and is found equally in men ៉ Embryonal carcinoma:
and women, unlike the other germ cell tumors. This ៉ Smears show large clusters and papillae of poorly
tumor is large, calcified, cystic, and filled with grum- differentiated cells with vacuolated cytoplasm
៉ Nuclei are large with coarsely granular chromatin and
ous granular material identical to its ovarian counter-
prominent nucleoli
part. FNA can show these components and skin adnexal ៉ Close resemblance to poorly differentiated adenocarcinoma
structures, gastrointestinal, respiratory, and cartilagi-
nous material (Fig. 7-6). Foreign body granulomas Ancillary Studies
are common in response to the keratin found in terato- ៉ Seminoma: positive for immunostaining with c-kit and PLAP
mas. Hair is not often seen in a cytology specimen, but ៉ Embryonal carcinoma: positive for CD30
short strands can sometimes be observed. Immature ៉ Yolk sac tumor: positive for AFP
teratomas are more common in men and show the ៉ Choriocarcinoma: positive for HCG
presence of immature epithelial, mesenchymal, and ៉ The most common karyotype abnormality is i(12p), present in
neural material. It is an uncommon tumor in the about 40% of patients
mediastinum.
Differential Diagnosis and Pitfalls
៉ Metastatic disease must be excluded
SEMINOMA

Germinomas or seminomas occur exclusively in men.


This is the most frequent mediastinal germ cell tumor YOLK SAC (ENDODERMAL SINUS) TUMOR
in males. The tumor cells here are infiltrated by reactive
lymphocytes, and because of this dual population, Yolk sac tumor is similar to embryonal carcinoma in
thymoma enters the differential; however, unlike in its vacuolated cytoplasm and malignant-appearing cells.
thymoma, plasma cells are common. Seminoma is poorly The cytoplasm of these tumor cells contains large red-
cohesive and appears malignant at first glance. Hyper- staining hyaline globules, which are also found free
chromatic enlarged nuclei are seen with very consistent, in the background. These globules contain alpha-
prominent nucleoli. Cytoplasm is clear. An often- fetoprotein (AFP), which can be also elevated in the
reported feature is the striped or ‘tigroid’ pattern in the serum of these patients. The background is bloody with
background seen in air-dried smears. We find this tumor cell fragments and a ‘dirty’ appearance. An
feature more often mentioned in the literature than inflammatory component may be present (Figs 7-7 &
actually seen on slides. Granulomas and multinucleated 7-8). The presence of glomeruloid or Schiller-Duval
giant cells are common. These giant cells are of the bodies, which are the invagination of cells into a sinus
syncytotrophoblast type, and not the cytotrophoblast or small space, signify the yolk sac nature of this tumor
type seen in choriocarcinoma. (Fig. 7-9).

Ch007-F06731.indd 207 10/26/2006 10:28:53 AM


208 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 7-6
Benign cystic teratoma. This cell
block shows benign epithelial
glandular elements. The corre-
sponding smear consisted of anucle-
ated squames and benign squamous
cells, proteinaceous debris, and
mixed infl ammatory infiltrate. H&E
stain, high magnification.

FIGURE 7-7 FIGURE 7-8


Yolk sac tumor. Papillary-like structure with large cells and prominent Yolk sac tumor. The tumor shows large cells with prominent nucleoli
nucleoli. Infl ammation and mucoid material are present. Diff-Quik stain, and intracytoplasmic hyaline globules. Multiple bizarre cell shapes are
high magnification. present. Papanicolaou stain, high magnification.

EMBRYONAL CARCINOMA ANCILLARY STUDIES


Embryonal carcinoma is seen as large clusters and
papillae of poorly differentiated cells with vacuolated
cytoplasm. The nuclei are large with coarsely granular Immunohistochemistry may be helpful in confirming
chromatin and prominent nucleoli (Fig. 7-10). The the diagnosis of germ cell tumors. Seminoma is high-
resemblance to poorly differentiated adenocarcinoma is lighted by immunostain for c-kit and placental-like
significant, and tumor and/or serum markers may be alkaline phosphatase (PLAP), while CD30 stains the
necessary to identify these tumors as germ cell in embryonal component. AFP stains for yolk sac tumor.
origin. Molecular analysis of i(12p), which is characteristic of

Ch007-F06731.indd 208 10/26/2006 10:28:53 AM


CHAPTER 7 Mediastinum 209

or respiratory tract carcinoma with a defining t(15;19).


This tumor occurs in young people and can involve
the area of the thymus. It is a poorly differentiated
carcinoma showing varying degrees of squamous
differentiation.

THYMIC NEUROENDOCRINE
(CARCINOID) TUMORS

CLINICAL FEATURES

Thymic neuroendocrine (carcinoid) tumors are a rare


FIGURE 7-9 neoplasm of the anterior mediastinum. These tumors
Yolk sac tumor. A cell block of yolk sac tumor showing a Schiller-Duval frequently exhibit a wide spectrum of histology and
body invaginating into a vascular space. Papanicolaou stain, high
magnification.
appear to follow a more aggressive behavior than their
non-thymic counterparts. Primary neuroendocrine
tumors may occur in the mediastinum and are charac-
terized as atypical or malignant carcinoid tumors.
Rarely, primary small cell carcinomas of the thymus
may also occur. Carcinoid tumors that occur in the
mediastinum, like pulmonary carcinoid, are thought
to be of foregut origin. This disease of adults can
have paraneoplastic symptoms and a minority of these
tumors are associated with multiple endocrine neopla-
sia (MEN) type 1. Thymic carcinoids are more aggres-
sive and have a poorer clinical outcome than do their

THYMIC NEUROENDOCRINE (CARCINOID) TUMORS –


DISEASE FACT SHEET

Definition
៉ A malignant tumor, cytologically similar to carcinoid tumors
FIGURE 7-10
found at other sites
Embryonal carcinoma. The smear shows large, undifferentiated epithe-
lial cells with enlarged nuclei and conspicuous nucleoli. Papanicolaou
Incidence
stain, high magnification.
៉ Highest incidence is in the 4th to 5th decades of life

germ cell tumors, can be done on fresh tissue taken at Gender and Age Distribution
the time of the FNA, either as a needle core or as an ៉ Three out of four patients are men
additional pass for cytologic material.
Clinical Features
៉ Associated with Cushing syndrome and multiple endocrine
neoplasia (MEN) syndromes
DIFFERENTIAL DIAGNOSIS AND PITFALLS ៉ Presents as a large and lobulated mass in the anterior
mediastinum
៉ Patients may experience cough, dyspnea, superior vena cava
A feature common to germ cell tumors in cytology is syndrome, fatigue, fever, and night sweats
the resemblance to poorly differentiated carcinoma,
such as seen in the lung. Care must be taken to accu- Prognosis and Treatment
rately diagnose germ cell tumors, which generally have ៉ Metastasis is common (one-third with bone metastases)
៉ Thymic carcinoid associated with MEN syndrome is more
a good prognosis with appropriate treatment, from
malignant in behavior
poorly differentiated/undifferentiated midline carci-
៉ Treatment is complete surgical resection
noma, where the prognosis is not favorable. ៉ Radiation and chemotherapy may have limited roles
Germ cell tumor and other midline tumors must be
differentiated from the recently described lethal midline

Ch007-F06731.indd 209 10/26/2006 10:28:55 AM


210 FINE NEEDLE ASPIRATION CYTOLOGY

pulmonary counterparts, even when morphology is


similar.

CYTOPATHOLOGIC FEATURES

This tumor is composed of a single population of poorly


cohesive, small, round to oval, bland-appearing cells
with a distinctive ‘salt and pepper’ chromatin pattern
(Fig. 7-11). Inconspicuous nucleoli and scant cytoplasm
characterize carcinoid of the mediastinum and of any
site (Figs 7-11 & 7-12). Rosettes and spindle cells may
be present. The mitotic count is the primary tool for
distinguishing typical from atypical carcinoid, but
mitotic figures are often difficult to identify in cytologic
material, even when they are multiple and obvious in FIGURE 7-11
the corresponding tissue. Additionally, it is not practi- Thymic carcinoid tumor. Bland and loosely cohesive fragment of
cal to count mitotic figures per high-power fields in spindled cells, with fine chromatin. Nucleoli are not conspicuous. The
cytology; therefore, we recommend the diagnosis of car- chromatin pattern is distinctively ‘salt and pepper’. A vessel is seen in
cinoid on FNA material without an attempt to classify the mid to upper field. Diff-Quik stain, low magnification.
it into typical or atypical. The differential diagnosis of
thymic or mediastinal carcinoids includes small cell
carcinoma metastatic from, usually, lung. Parathyroid tumors may occur in this location and can
be distinguished by their staining with antibody to
parathormone. Lymphomas will be immunopositive for
leukocyte common antigen, and germ cell tumors will
ANCILLARY STUDIES react with immunostain for PLAP.

Immunohistochemical markers such as chromogranin


and synaptophysin must confirm the neuroendocrine DIFFERENTIAL DIAGNOSIS AND PITFALLS
nature of the cells. These tumors react with antibodies
to cytokeratin and chromogranin A, as well as other
neuroendocrine markers, including synaptophysin. Carcinoid tumors may be distinguished from thymo-
mas by the lack of background lymphocytes, rosette
formation, granularity of cytoplasm, and ‘salt and
pepper’ chromatin (Fig. 7-12).
THYMIC NEUROENDOCRINE (CARCINOID) TUMORS –
PATHOLOGIC FEATURES

Cytopathologic Findings MEDIASTINAL LYMPHOMAS


៉ Composed of single population of poorly cohesive, small, round
to oval, bland-appearing cells with a distinctive ‘salt and
pepper’ chromatin pattern
៉ Inconspicuous nucleoli and scant cytoplasm characterize
CLINICAL FEATURES
carcinoid of the mediastinum and of any site
៉ Rosettes and spindle cells may be present
Malignant lymphoma is the most common primary
middle mediastinal tumor, but occurs more frequently
Ancillary Studies
in the anterior and superior mediastinum overall.
៉ Tumors react with antibodies to cytokeratin and chromogranin
Most mediastinal lymphomas are Hodgkin lymphomas,
A, as well as other neuroendocrine markers, including
synaptophysin
usually the nodular sclerosing subtype.

Differential Diagnosis and Pitfalls


៉ The differential diagnosis of thymic or mediastinal carcinoids
includes small cell carcinoma metastatic from, usually, lung,
CYTOPATHOLOGIC FEATURES
and lymphomas
៉ Lymphomas will be immunopositive for leukocyte common
In Hodgkin lymphoma, the FNA may be scant due to
antigen, and germ cell tumors will react with immunostains for
PLAP
sampling of the collagen and it may be difficult to iden-
tify the necessary Reed-Sternberg (R-S) cells. Nodular
lymphocyte-predominant Hodgkin disease (NLPHD)

Ch007-F06731.indd 210 10/26/2006 10:28:56 AM


CHAPTER 7 Mediastinum 211

FIGURE 7-12
Thymic carcinoid tumor. A cellular
smear with cohesive fragments of
monotonous neoplastic cells with
round to ovoid nuclei, and fine
chromatin. Nucleoli are not con-
spicuous. Papanicolaou stain, low
magnification.

THYMIC LYMPHOMAS (HODGKIN AND NON-HODGKIN TYPES) – has somewhat pleomorphic, lobated, ‘popcorn-like’ R-S
DISEASE FACT SHEET cell variants (CD30- and CD15-negative), while classi-
cal Hodgkin lymphoma contains the highly pleomor-
Definition phic classic R-S cells (CD30- and CD15-positive).
៉ The three most common types of mediastinal lymphomas include Smears may show large abnormal lymphoid cells, crush
nodular sclerosing Hodgkin disease, large B-cell lymphoma, and artifact, and several R-S cells (Fig. 7-13).
lymphoblastic lymphoma The background present in the smears is similar to
៉ Hodgkin disease (HD) is a lymphoma with large abnormal cells that in Hodgkin lymphoma from any other site, with a
termed Reed-Sternberg (R-S) cells and Hodgkin cells present in a heterogeneous population of lymphocytes with plasma
mixed background of lymphocytes and mixed inflammatory cells
cells and eosinophils (Fig. 7-14). The presence of lym-
៉ Lymphoblastic lymphoma is the most common lymphoma of
children and most arise in the thymus
phoglandular bodies is often reported in the background
of air-dried smears in lymphoma. These pinched-off bits
Incidence of cytoplasm can be found with all stains and indicate
៉ Malignant lymphoma is the most common primary neoplasm of
a heavy lymphoid infiltrate. They make no comment on
the middle mediastinum, but in absolute numbers it is more benign or malignant conditions. We do not find them of
common in the anterior and superior mediastinum particular help in classifying lymphomas or even in
៉ HD is the most common mediastinal lymphoma (70%) and nodular recognizing lymphoid populations, as cytoplasm frag-
sclerosing is the most common subtype ments from any other cell can mimic lymphoglandular
៉ HD has an incidence of 2–4 per 100,000 bodies.
Large cell, B-cell lymphomas also occur, and may be
Gender and Age Distribution extranodal or primary in the thymus. All cell types
៉ HD: slightly more common in males; peak age, 15 to 35 years present are lymphoid, in contrast to the picture of lym-
៉ Non-Hodgkin lymphoma: highest incidence is in white men with a phoid and epithelial cells in thymoma. Large cell lym-
mean age of 55 years phoma has large, vesicular nuclei and often-irregular
៉ Lymphoblastic lymphoma: mean age, 28 years nuclear membranes (Figs 7-15–7-17). These large lym-
phoid cells may be mixed with smaller reactive cells.
Clinical Features
Lymphomas on cytology are rarely of a single popula-
៉ Subclassification of HD is less important now than earlier (age tion of cells and the presence of reactive cells and tingi-
and stage most important)
ble-body macrophages should not distract the observer
៉ Systemic symptoms (fever, night sweats, weight loss) carry worse
prognosis
from the presence of the diagnostic large malignant lym-
phocytes (Figs 7-15 & 7-16).
Prognosis Lymphoblastic lymphoma is the most common lym-
៉ HD is curable in >90% with stage I–II disease
phoma of children and most arise in the thymus. These
៉ Lymphoblastic lymphoma is highly aggressive
tumors are of pre-T-cell lineage, present with respira-
tory distress, and are often fatal. The diagnostic feature
of these lymphomas is lymphoid blasts, which are

Ch007-F06731.indd 211 10/26/2006 10:28:56 AM


212 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 7-13 FIGURE 7-14


Hodgkin lymphoma. Note the large abnormal lymphoid cells, crush Hodgkin lymphoma. This smear shows a classic Reed-Sternberg cell (left
artifact, and several Reed-Sternberg cells. Diff-Quik stain, low of center) and polymorphous background. Diff-Quik stain, low
magnification. magnification.

THYMIC LYMPHOMA (HODGKIN AND NON-HODGKIN TYPES) –


PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Hodgkin disease (HD):
៉ Arises in thymus or lymph nodes, almost never extranodal
៉ Reed-Sternberg (R-S) cells contain bilobed nuclei with
prominent eosinophilic nuclei
៉ R-S cells may be rare; they are necessary but insufficient for
diagnosis
៉ Background important: lymphocytes, plasma cells,
eosinophils, histiocytic
៉ +/− cellular (sparse aspirate from a large node is suspicious
in itself)
៉ Inverse relationship between number of R-S cells and
lymphocytes
៉ Metachromatic material present in smears FIGURE 7-15
៉ Non-Hodgkin lymphoma: Non-Hodgkin lymphoma. Monotonous population of malignant lympho-
៉ Large vesicular nuclei cytes displaying occasional conspicuous nucleoli. Papanicolaou stain,
៉ Nucleoli prominent but not macro low magnification.
៉ Irregular (cleaved) or smooth (non-cleaved) membranes
៉ Large cells may be mixed with smaller cells
៉ Cellularity may be quite limited when sclerotic characterized by a delicate chromatin pattern and
irregular nuclear outlines. Nucleoli are not prominent.
Ancillary Studies Necrosis, mitosis, eosinophils, granulomas, and macro-
៉ Classical HD: R-S cells are positive for CD15 and CD30, and phages may accompany the blasts. Since these lympho-
negative for CD20 mas infiltrate thymic tissue, normal thymic components
៉ Flow cytometry is helpful for the classification of non-Hodgkin
may be present in the aspirates.
lymphoma

Differential Diagnosis and Pitfalls


៉ Differential diagnosis includes thymoma, and germinoma ANCILLARY STUDIES
៉ Cleaved lymphocytes may mimic cells in thymoma but all cell
types here are lymphoid
៉ Sclerotic large cell lymphomas are common in the mediastinum Flow cytometric studies can be done on cytologic mate-
and may mimic HD rial; therefore the importance of on-site cytopathologic
៉ Chronic sclerosing mediastinitis can be confused with tumors
evaluation is critical. Cell blocks may be prepared and
having a significant sclerotic or collagenous component such as
nodular sclerosing HD
appropriate immunostain may be used to further
classify and subtype the lymphomas. These details are
discussed in greater length in Chapter 3.

Ch007-F06731.indd 212 10/26/2006 10:28:57 AM


CHAPTER 7 Mediastinum 213

NEUROGENIC TUMORS – DISEASE FACT SHEET

Definition and Incidence


៉ Neurogenic tumors account for the majority of tumors arising in
the posterior mediastinum
៉ Neurogenic tumors include nerve root tumors such as schwannoma
and neurofibroma, and sympathetic ganglion tumors such as
neuroblastoma, ganglioneuroblastoma, and ganglioneuroma
៉ Paragangliomas may rarely present as posterior mediastinal
masses
៉ Schwannoma is the most common neurogenic tumor of the
mediastinum; it is generally solitary and large, with cystic change
or calcification common

Gender and Age Distribution


៉ 20% of all adult mediastinal neoplasms
៉ 40% of all pediatric mediastinal neoplasms
FIGURE 7-16
Non-Hodgkin lymphoma. High-power view of monotonous population of Clinical Features
large malignant lymphocytes with prominent nucleoli. Papanicolaou
៉ 95% of these tumors in adults are benign and are usually
stain, high magnification.
asymptomatic
៉ In children, most neurogenic tumors are malignant

Prognosis
៉ 80% of these tumors are benign

Ganglioneuroblastoma occurs in children and is a


neuroblastoma with the presence of ganglion cell dif-
ferentiation; ganglioneuroma is a tumor of adults and is
composed of mature ganglion cells. Ganglia have large
prominent nucleoli and can be bi- or multinucleated. On
a cursory inspection, they can be mistaken for malig-
nant cells.

FIGURE 7-17 CYTOPATHOLOGIC FEATURES


Non-Hodgkin lymphoma. High-power view of malignant lymphoma cells.
Note mitoses and cleaved nuclei. Diff-Quik stain, high magnification.
Neurofibroma and schwannoma both have bland
spindle cells loosely arranged and may be calcified.
Schwannomas are more common, and when there is
degeneration or atypia present, they can be mistaken
NEUROGENIC TUMORS for sarcoma or melanoma. The smears may show an
extensively degenerative background, fragile cytoplasm,
and occasional large atypical cells (Fig. 7-18). Antoni
CLINICAL FEATURES A and B growth patterns can often be seen in cellular
cytology preparations of schwannomas (Fig. 7-19).
However, malignant schwannoma and malignant
Neurogenic tumors are most frequently located in the peripheral nerve sheath tumors do occur in the poste-
posterior mediastinum and most are malignant sympa- rior mediastinum and they can be recognized by necro-
thetic tumors of childhood. Nerve sheath tumors and sis, mitoses, and nuclear atypia (Figs 7-20–7-22).
gangliomas are more commonly found in the adult Paragangliomas can arise in the superior or anterior
population. When benign, these neurogenic tumors are mediastinum and are very vascular tumors of the sym-
often paucicellular, making FNA diagnosis difficult. pathetic nerve chain. The tumors are composed of cells
Neuroblastoma appears as atypical small round blue grouped into ball-like arrangements, the ‘Zellballen’.
cell tumor and has the same differential diagnosis and The tumor has large cells with generous, pale, some-
work-up as elsewhere in the body, remembering that what granular eosinophilic cytoplasm. The nuclei
small cell carcinoma is not expected in children. of these benign cells may show marked atypia and

Ch007-F06731.indd 213 10/26/2006 10:28:58 AM


214 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 7-20
FIGURE 7-18
Malignant peripheral nerve sheath tumor. Note the bizarre pleomorphic
Schwannoma. Smear showing degenerative background, spindled cells cells. This tumor was located in the posterior mediastinum of an adult.
with fragile cytoplasm, and occasional large pleomorphic cells. Diff- The differential diagnosis includes sarcoma NOS and sarcomatoid carci-
Quik stain, high magnification. noma. Diff-Quik stain, high magnification.

A B

FIGURE 7-19
Schwannoma. A, Large tissue fragment from an FNA of a schwannoma, showing Antoni A and B areas. B, Higher power of the spindled cells with
uniform oval nuclei. Papanicolaou, low and high magnification.

FIGURE 7-22
FIGURE 7-21 Malignant peripheral nerve sheath tumor. Note the bizarre multinucle-
ate giant cell on the left and sarcomatoid appearance on the right.
Malignant peripheral nerve sheath tumor. Note the spindle and large
Papanicolaou stain, low magnification (left); H&E stain (right).
atypical cells. The appearance is that of a malignant spindle cell neo-
plasm, and clinical history and ancillary studies are vital for an accurate
diagnosis. Papanicolaou stain, low magnification.

Ch007-F06731.indd 214 10/26/2006 10:28:59 AM


CHAPTER 7 Mediastinum 215

NEUROGENIC TUMORS – PATHOLOGIC FEATURES METASTATIC TUMORS – DISEASE FACT SHEET

Cytopathologic Findings Definition


៉ Schwannomas: ៉ Metastatic lesions are the common cause of a mediastinal mass in
៉ Tumor has cellular (Antoni A) and degenerated/myxoid adults
(Antoni B) areas ៉ FNA can establish the diagnosis and subtype metastatic lesions to
៉ Variable cellularity with large thick fragments, smaller the mediastinum
clusters, and single cells ៉ This may be a secondary lesion in a patient with a previous
៉ Cells are elongated and spindle-shaped and may palisade history of cancer or a new presentation
៉ Uniform, round to plump to oval nuclei with fine chromatin
and inconspicuous nucleoli Incidence
៉ Regressive or degenerative changes and increased cellularity ៉ Dependent on the tumor type
possible ៉ Small cell carcinoma is the most common type
៉ Intranuclear cytoplasmic invaginations can be seen
៉ Cytoplasm is fragile and sometimes strips away Gender and Age Distribution
៉ Ground substance is fibrillar
៉ Dependent on the tumor type
៉ No mitoses or necrosis

Clinical Features
Ancillary Studies
៉ Most common source of mediastinal metastasis is small cell
៉ Immunohistochemistry for S-100 may be helpful when
carcinoma of lung
confirming the neurogenic origin of these tumors
៉ Associated with multiple endocrine neoplasia type 1

Differential Diagnosis and Pitfalls


Prognosis and Treatment
៉ Cytologic atypia, mitosis, necrosis – think malignancy
៉ Metastatic small cell carcinoma is aggressive with extensive
metastases
៉ Treatment for small cell carcinoma: radiation therapy and
chemotherapy

enlargement. Small amounts of focal necrosis can be


present even in benign tumors, and the tumor cells can
resemble the cells of carcinoid, but carcinoids lack the
usual atypia of paragangliomas. Paragangliomas have
been misdiagnosed on cytology material as anaplastic METASTATIC TUMORS – PATHOLOGIC FEATURES
carcinoma with giant cells and that was thought to be
Cytopathologic Findings
due to the presence of vascular-rich papillary structures,
៉ Variable, but usually overtly malignant features are needed to
acinar-glandular structures, cellular pleomorphism, the
document malignancy
prominent nucleoli, and the ‘squamoid’ appearance of
the cytoplasm.
Ancillary Studies
Mesenchymal tumors are rare in the mediastinum
៉ Immunohistochemical stains for defining the origin of the lesion,
and resemble their counterparts in other body sites.
such as TTF (lung), CK7/CK20 profile, melanoma markers, S-100,
germ cell markers (PLAP, CD30), lymphoma markers

Differential Diagnosis and Pitfalls


METASTATIC LESIONS ៉ FNA can establish the diagnosis and subtype metastatic lesions to
the mediastinum
៉ Other common metastatic lesions include breast, thyroid, head and
neck, kidney, prostate, and testicular tumors, and melanoma
FNA is highly reliable for establishing a diagnosis of
metastatic lesions in the mediastinum. The aspirate
and the material collected concurrently, may be helpful
in establishing a new diagnosis or used to confirm that
a secondary lesion is metastatic from a previously
known primary. The material may also be useful for
molecular diagnostic testing, such as the evaluation of features of small cell carcinoma seen in the primary
the recently discovered epidermal growth factor recep- lesion are present here, including crush artifact and
tor (EGFR) mutation, to ascertain therapeutic efficacy nuclear molding (Figs 7-23 & 7-24). Squamous cell car-
of tyrosine kinase inhibitors cinoma from the head and neck or a lung primary may
Studies have highlighted that most metastatic lesions present as a metastatic lesion in the mediastinum (Fig.
to mediastinal lymph nodes are derived from the lung 7-25). Other common metastatic lesions include breast,
and that small cell carcinoma represents one of the most thyroid, kidney, prostate, and testicular tumors, and
common metastatic lesions to the mediastinum. Typical melanoma.

Ch007-F06731.indd 215 10/26/2006 10:29:00 AM


216 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 7-23
Metastatic small cell carcinoma.
Smear depicts cells with prominent
nuclear molding, scant cytoplasm,
mitoses, and nuclear crush artifact.
This is indistinguishable from a
primary mediastinal small cell
carcinoma. Diff-Quik stain, low
magnification.

FIGURE 7-24
Metastatic small cell carcinoma. Characteristic features of small cell carcinoma, with fine nuclear membranes, powdery chromatin, and inconspicuous
nucleoli. Papanicolaou stain, high magnification.

Ch007-F06731.indd 216 10/26/2006 10:29:00 AM


CHAPTER 7 Mediastinum 217

FIGURE 7-25
Metastatic squamous cell carci-
noma. Smear showing bizarre,
pleomorphic cells with dark nuclear
chromatin, thick opaque cytoplasm,
and conspicuous nucleoli. Papanico-
laou stain, high magnification.

SUGGESTED READINGS Bangerter M, Behnisch W, Griesshammer M. Mediastinal masses diagnosed


as thymus hyperplasia by fine needle aspiration cytology. Acta Cytol
Introduction/Mediastinal Masses 2000;44:743–747.
Chhieng DC, Rose D, Ludwig ME, Zakowski MF. Cytology of thymomas:
Belfiore G, Camera L, Moggio G, Vetrani A, Fraioli G, Salvatore M. Middle
emphasis on morphology and correlation with histologic subtypes.
mediastinum lesions: preliminary experience with CT-guided fine-needle
Cancer 2000;90:24–32.
aspiration biopsy with a suprasternal approach. Radiology 1997;202:
Dadmanesh F, Sekihara T, Rosai J. Histologic typing of thymoma according
870–873.
to the new World Health Organization classification. Chest Surg Clin N
Blegvad S, Lippert H, Simper LB, Dybdahl H. Mediastinal tumours. A report
Am 2001;11:407–420.
of 129 cases. Scand J Thorac Cardiovasc Surg 1990;24:39–42.
Dahlgren S, Sandstedt B, Sundstrom C. Fine needle aspiration cytology of
Duwe BV, Sterman DH, Musani AI. Tumors of the mediastinum. Chest
thymic tumors. Acta Cytol 1983;27:1–6.
2005;128:2893–2909.
Morgenthaler TI, Brown LR, Colby TV, Harper CM Jr, Coles DT. Thymoma.
Geisinger KR. Differential diagnostic considerations and potential pitfalls
Mayo Clin Proc 1993;68:1110–1123.
in fine-needle aspiration biopsies of the mediastinum. Diagn Cytopathol
Okumura M, Ohta M, Miyoshi S, et al. Oncological significance of WHO
1995;13:436–442.
histological thymoma classification. A clinical study based on 286
Panelli F, Erickson RA, Prasad VM. Evaluation of mediastinal masses by
patients. Jpn J Thorac Cardiovasc Surg 2002;50:189–194.
endoscopic ultrasound and endoscopic ultrasound-guided fine needle
Ritter JH, Wick MR. Primary carcinomas of the thymus gland. Semin Diagn
aspiration. Am J Gastroenterol 2001;96:401–408.
Pathol 1999;16:18–31.
Powers CN, Silverman JF, Geisinger KR, Frable WJ. Fine-needle aspiration
Sherman ME, Black-Schaffer S. Diagnosis of thymoma by needle biopsy.
biopsy of the mediastinum. A multi-institutional analysis. Am J Clin
Acta Cytol 1990;34:63–68.
Pathol 1996;105:168–173.
Shin HJ, Katz RL. Thymic neoplasia as represented by fine needle aspira-
Shabb NS, Fahl M, Shabb B, Haswani P, Zaatari G. Fine-needle aspiration
tion biopsy of anterior mediastinal masses. A practical approach to the
of the mediastinum: a clinical, radiologic, cytologic, and histologic
differential diagnosis. Acta Cytol 1998;42:855–864.
study of 42 cases. Diagn Cytopathol 1998;19:428–436.
Suster S, Moran CA. Primary thymic epithelial neoplasms showing com-
Singh HK, Silverman JF, Powers CN, Geisinger KR, Frable WJ. Diagnostic
bined features of thymoma and thymic carcinoma. A clinicopathologic
pitfalls in fine-needle aspiration biopsy of the mediastinum. Diagn
study of 22 cases. Am J Surg Pathol 1996;20:1469–1480.
Cytopathol 1997;17:121–126.
Tao LC, Pearson FG, Cooper JD, Sanders DE, Weisbrod G, Donat EE. Cyto-
Sterrett G, Whitaker D, Shilkin KB, Walters MN. The fine needle aspiration
pathology of thymoma. Acta Cytol 1984;28:165–170.
cytology of mediastinal lesions. Cancer 1983;51:127–135.
Wakely PE Jr. Cytopathology-histopathology of the mediastinum: epithe-
lial, lymphoproliferative, and germ cell neoplasms. Ann Diagn Pathol Germ Cell Tumors
2002;6:30–43.
Wakely PE Jr. Cytopathology-histopathology of the mediastinum II. Chhieng DC, Lin O, Moran CA, et al. Fine-needle aspiration biopsy of
Mesenchymal, neural, and neuroendocrine neoplasms. Ann Diagn Pathol nonteratomatous germ cell tumors of the mediastinum. Am J Clin Pathol
2005;9:24–32. 2002;118:418–424.
Kiffer JD, Sandeman TF. (Primary malignant mediastinal germ cell tumours:
a literature review and a study of 18 cases. Australas Radiol 1999;
Thymic Lesions 43:58–68.
Kwon MS. Aspiration cytology of mediastinal seminoma: report of a case
Ali SZ, Erozan YS. Thymoma. Cytopathologic features and differential with emphasis on the diagnostic role of aspiration cytology, cell block
diagnosis on fine needle aspiration. Acta Cytol 1998;42:845–854. and immunocytochemistry. Acta Cytol 2005;49:669–672.

Ch007-F06731.indd 217 10/26/2006 10:29:01 AM


218 FINE NEEDLE ASPIRATION CYTOLOGY

Moran CA, Suster S, Koss MN. Primary germ cell tumors of the mediasti- Neurogenic Tumors
num: III. Yolk sac tumor, embryonal carcinoma, choriocarcinoma, and
combined nonteratomatous germ cell tumors of the mediastinum – a de Montpreville VT, Mussot S, Gharbi N, Dartevelle P, Dulmet E. Paragan-
clinicopathologic and immunohistochemical study of 64 cases. Cancer glioma with ganglioneuromatous component located in the posterior
1997;80:699–707. mediastinum. Ann Diagn Pathol 2005;9:110–114.
Moran CA, Suster S, Przygodzki RM, Koss MN. Primary germ cell tumors Geisinger KR. Differential diagnostic considerations and potential pitfalls
of the mediastinum: II. Mediastinal seminomas – a clinicopathologic in fine-needle aspiration biopsies of the mediastinum. Diagn Cytopathol
and immunohistochemical study of 120 cases. Cancer 1997;80:691– 1995;13:436–442.
698. Khanlou H, Khanlou N, Eiger G. Schwannoma of posterior mediastinum: a
Motoyama T, Yamamoto O, Iwamoto H, Watanabe H. Fine needle aspiration case report and concise review. Heart Lung 1998;27:344–347.
cytology of primary mediastinal germ cell tumors. Acta Cytol 1995;39: Marchevsky AM. Mediastinal tumors of peripheral nervous system origin.
725–732. Semin Diagn Pathol 1999;16:65–78.
Weidner N. Germ-cell tumors of the mediastinum. Semin Diagn Pathol Slagel DD, Powers CN, Melaragno MJ, Geisinger KR, Frable WJ, Silverman
1999;16:42–50. JF. Spindle-cell lesions of the mediastinum: diagnosis by fine-needle
aspiration biopsy. Diagn Cytopathol 1997;17:167–176.
Strollo DC, Rosado-de-Christenson ML, Jett JR. Primary mediastinal
Thymic Neuroendocrine (Carcinoid) Tumors tumors: part II. Tumors of the middle and posterior mediastinum. Chest
1997;112:1344–1357.
Caceres W, Baldizon C, Sanchez J. Carcinoid tumor of the thymus: a unique Topcu S, Alper A, Gulhan E, Kocyigit O, Tastepe I, Cetin G. Neurogenic
neoplasm of the mediastinum. Am J Clin Oncol 1998;21:82–83. tumours of the mediastinum: a report of 60 cases. Can Respir J
Moran CA, Suster S. Spindle-cell neuroendocrine carcinomas of the thymus 2000;7:261–265.
(spindle-cell thymic carcinoid): a clinicopathologic and immunohisto- Wakely PE Jr. Cytopathology-histopathology of the mediastinum II. Mes-
chemical study of seven cases. Mod Pathol 1999;12:587–591. enchymal, neural, and neuroendocrine neoplasms. Ann Diagn Pathol
Nichols GL Jr, Hopkins MB 3rd, Geisinger KR. Thymic carcinoid. Report of 2005;9:24–32.
a case with diagnosis by fine needle aspiration biopsy. Acta Cytol
1997;41:1839–1844.
Slagel DD, Powers CN, Melaragno MJ, Geisinger KR, Frable WJ, Silverman Metastatic Lesions
JF. Spindle-cell lesions of the mediastinum: diagnosis by fine-needle
aspiration biopsy. Diagn Cytopathol 1997;17:167–176. Al-Haddad M, Wallace MB. Molecular diagnostics of non-small cell lung
Wick MR, Scheithauer BW. Thymic carcinoid. A histologic, immunohisto- cancer using mediastinal lymph nodes sampled by endoscopic ultra-
chemical, and ultrastructural study of 12 cases. Cancer 1984;53: sound-guided needle aspiration. Cytopathology 2006;17:3–9.
475–484. Bavi P, Shet T, Gujral S. Malignant melanoma of mediastinum misdiag-
nosed as a spindle cell thymoma in a fine needle aspirate: a case report.
Acta Cytol 2005;49:424–426.
Mediastinal Lymphomas Blanco N, Kirgan DM, Little AG. Metastatic squamous cell carcinoma of
the mediastinum with unknown primary tumor. Chest 1998;114:938–
Friedman HD, Hutchison RE, Kohman LJ, Powers CN. Thymoma mimicking 940.
lymphoblastic lymphoma: a pitfall in fine-needle aspiration biopsy Duwe BV, Sterman DH, Musani AI. Tumors of the mediastinum. Chest
interpretation. Diagn Cytopathol 1996;14:165–169; discussion 169– 2005;128:2893–2909.
171. Lau CL, Bentley RC, Gockerman JP, Que LG, D’Amico TA. Malignant mela-
Sheibani K, Nathwani BN, Winberg CD, et al. Antigenically defined sub- noma presenting as a mediastinal mass. Ann Thorac Surg 1999;67:
groups of lymphoblastic lymphoma. Relationship to clinical presenta- 851–852.
tion and biologic behavior. Cancer 1987;60:183–190. Saad RS, Landreneau RJ, Liu Y, Silverman JF. Utility of immunohisto-
Silverman JF, Raab SS, Park HK. Fine-needle aspiration cytology of primary chemistry in separating thymic neoplasms from germ cell tumors and
large-cell lymphoma of the mediastinum: cytomorphologic findings with metastatic lung cancer involving the anterior mediastinum. Appl Immu-
potential pitfalls in diagnosis. Diagn Cytopathol 1993;9:209–214; dis- nohistochem Mol Morphol 2003;11:107–112.
cussion 214–215. Singh HK, Silverman JF, Powers CN, Geisinger KR, Frable WJ. Diagnostic
Strickler JG, Kurtin PJ. Mediastinal lymphoma. Semin Diagn Pathol pitfalls in fine-needle aspiration biopsy of the mediastinum. Diagn
1991;8:2–13. Cytopathol 1997;17:121–126.
Yu GH, Salhany KE, Gokaslan ST, Cajulis RS, De Frias DV. Thymic epithelial Sinner WN. Directed fine needle biopsy of anterior and middle mediastinal
cells as a diagnostic pitfall in the fine-needle aspiration diagnosis masses. Oncology 1985;42:92–96.
of primary mediastinal lymphoma. Diagn Cytopathol 1997;16:460– Sterrett G, Whitaker D, Shilkin KB, Walters MN. The fine needle aspiration
465. cytology of mediastinal lesions. Cancer 1983;51:127–135.

Ch007-F06731.indd 218 10/26/2006 10:29:02 AM


8 Liver
Syed Z Ali • Natasha Rekhtman

Along with the pancreas, the liver is one of the most In our experience, benign hepatic entities are often
common visceral organs targeted by fine needle aspira- more taxing to diagnose on FNA, perhaps because they
tion (FNA). The usual clinical scenario is that of an are rarer than malignancies in the liver and also because
asymptomatic patient presenting with a solitary or mul- of a relative lack of characteristic findings on cytopatho-
tiple liver lesions. In a significant number of cases, there logic evaluation (Table 8-5).
is no prior history of malignancy and the liver lesion is
a totally incidental finding, therefore adding to the diag-
nostic challenge when such lesion is aspirated.
Liver is approached through a percutaneous trans- NON-NEOPLASTIC DISEASES OF THE LIVER
abdominal route with radiologic guidance. At Johns
Hopkins, almost all liver lesions are aspirated via ultra-
sound guidance, which is a much faster and safer pro-
cedure than computed tomography (CT) guidance and MACROREGENERATIVE NODULE
offers real-time visualization of the needle positioning
and aspiration. All FNAs are evaluated on-site by a
cytopathologist for adequacy. Material is triaged for CLINICAL FEATURES
ancillary studies (flow cytometry, microbial culture,
etc.) and, if needed and technically feasible, the radio- Macroregenerative nodules (MRNs), also referred to as
logist is asked for tissue core biopsy as well. As in other adenomatous hyperplasia, are, in essence, large regen-
body sites, it is extremely important to be familiar with erative nodules arising in cirrhotic livers. Some authors
the appearances of normal cells comprising a liver FNA have applied the size cut-off of 0.8 cm to distinguish
(Table 8-1; Figs 8-1 & 8-2). these lesions from typical nodules of macronodular cir-
It is imperative to be aware of the exact location of rhosis. MRNs are found in up to 50% of cirrhotic livers.
the lesion in the liver since unusual cell types (repre- MRNs are usually asymptomatic and are discovered
senting needle contaminants; Table 8-2) can be seen in incidentally during the radiographic surveillance of
addition to hepatic tissue; for example, gastric-type epi- patients with cirrhosis. Serum alpha-fetoprotein (AFP)
thelium may be observed in lesions of the left hepatic is not increased.
lobe if the needle traverses the stomach wall, or respira- MRNs are present in the population with cirrhosis:
tory-type epithelium may be seen in lesions located deep incidence increases with advancing age, and men are
in the hepatic dome (Fig. 8-3). Another uncommon cell
type which may add to diagnostic confusion to an inex-
perienced interpreter is mesothelium, often seen as a
flat, monolayered sheet with characteristic spacing in
between individual cells. MACROREGENERATIVE NODULE – DISEASE FACT SHEET
Liver FNA is an extremely safe procedure in experi-
Incidence
enced hands, with rare morbidity (Table 8-3). Mortality
៉ Present in up to 50% of cirrhotic livers
is also extremely rare, having been reported in the
literature as isolated case reports. In our experience,
Gender and Age Distribution
we have seen only two cases of hepatic FNA that led
៉ Reflects the demographic of cirrhosis
to significant intraperitoneal bleeding requiring some
៉ After age 60 in the USA; younger age in Asia and Africa
amount of intensive care. Both cases had occurred in
៉ Men > women (worldwide)
patients with large hepatocellular carcinomas. A recent
review of the literature on this subject revealed a mor- Prognosis
tality rate less than 0.1%.
៉ Premalignant potential is controversial
There are relatively rare contraindications for liver ៉ Up to 25% of lesions regress
FNA in view of an overall excellent safety profile of this
procedure (Table 8-4).
219

Ch008-F06731.indd 219 10/26/2006 10:30:03 AM


220 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 8-1 FIGURE 8-3


Normal liver. Small fragment of hepatocytes with polygonal cell shapes, Needle contamination. Diaphragmatic skeletal muscle in an FNA of a
granular cytoplasm, and small nucleoli. Focal bile staining is evident. hepatic dome lesion. Diff Quik stain, intermediate power.
Papanicolaou stain, high power.

TABLE 8-2
Normal Non-liver Cell Component
(Needle Contaminants)

• Mesothelium
• Respiratory epithelium
• Gastrointestinal tract epithelium
• Skeletal muscle and skin

TABLE 8-3
Complications of Hepatic FNA (n = number of cases
FIGURE 8-2 reported in the English-language literature)
Normal liver. Biliary epithelium with a more cohesive appearance of the
cells, high N/C ratio, and glandular formations (middle of the field). • Tumor seeding of the needle tract (n = 12):
Numerous hepatocytes are present in the background. Diff Quik stain, • Hepatocellular carcinoma [HCC] (n = 9)
high power. • Metastatic colonic adenocarcinoma (n = 3)
• Bleeding (n = 8):
• HCC (n = 5)
• Hemangioma (n = 2)
• Angiosarcoma (n = 1)
TABLE 8-1 • Bile peritonitis (n = 1)
Liver – Normal Cytologic Constituents • Carcinoid crisis (n = 1)
• Lymphorrhea (n = 1)
• Biliary venous fistula (n = 1)
• Hepatocytes
• Sudden hypoglycemia – clear cell HCC (n = 1)
• Biliary epithelium
• Endothelium
• Kupffer’s cells
• Pigments:
• Bile
• Hemosiderin
• Lipofuscin

Ch008-F06731.indd 220 10/26/2006 10:30:04 AM


CHAPTER 8 Liver 221

TABLE 8-4
Contraindications of Hepatic FNA

• Bleeding diathesis
• Difficult anatomic location of the lesion (such as close
proximity to vessels)
• Hydatid disease

TABLE 8-5
Benign Hepatic Diseases on FNA

• Non-neoplastic: FIGURE 8-4


• Granulomatous disease Nodular regenerative hyperplasia. Disorganized fragment of liver tissue.
• Infection: Cells display prominent nucleoli. However, there is lack of vascular
• Hydatid disease proliferation and single cells. Papanicolaou stain, low power.
• Other (amebic, schistosomiasis)
• Congenital cysts
• Focal nodular hyperplasia (FNH)
• Cirrhosis
• Benign neoplasm:
MACROREGENERATIVE NODULE – PATHOLOGIC FEATURES
• Hepatic adenoma
• Hemangioma
Cytopathologic Findings
៉ Reactive hepatocytes (high N/C ratio, often prominent nucleoli)
៉ Higher than normal amount of biliary epithelium
៉ Varying amount of fibroconnective tissue fragments
៉ Marked focal atypia of hepatocytic or biliary epithelium
affected more frequently than women. The premalig- ៉ Varying amount of lympho-mononuclear cells
nant potential of MRNs is a controversial subject. It is ៉ Cholestasis
generally believed that MRNs are not associated with ៉ Focal necrosis (rare)
an increased risk of hepatocellular carcinoma (HCC),
and up to 25% of lesions regress on radiographic follow- Histopathologic Findings
up. MRNs may pose a challenge differentiating them ៉ Gross:
clinically and pathologically from HCC. Patients with ៉ Usually multiple lesions
៉ Can measure over 5 cm
MRNs are followed with more frequent radiographic
៉ Background cirrhotic liver
studies.
៉ Microscopic:
៉ Indistinguishable from surrounding liver
៉ Normal cell plates (<2 cells thick)
៉ Reticulin network intact
PATHOLOGIC FEATURES ៉ Cytologic atypia absent or minimal
៉ Portal tracts present but decreased and distorted

Grossly, MRNs are similar in appearance to the sur- Ancillary Studies


rounding liver, although more pronounced bile staining ៉ CD34: no endothelial cell wrapping (unlike hepatocellular
may be present. Lesions are commonly multiple, and carcinoma [HCC])
can measure over 5 cm. MRNs are circumscribed by a ៉ Reticulin stain: reticulin framework intact (unlike HCC)
rim of fibrous tissue, analogous to the usual smaller
nodules in macronodular cirrhosis. Differential Diagnosis and Pitfalls
Histologically, MRNs are indistinguishable from the ៉ Well-differentiated HCC: like MRN, arises in cirrhotic liver;
surrounding liver: cell plates are of normal thickness increased cell plate thickness (>2 cells thick); disrupted
(less than two cells), reticulin network is intact, and reticulin framework; cytologic atypia present, but may be
cytologic atypia is absent or minimal. Portal tracts are minimal
present, although they may be decreased in number and ៉ Focal nodular hyperplasia (FNH) and hepatocellular adenoma
usually show architectural distortion. Cytopathologic (HA): distinction impossible based on cytology alone; arise in
non-cirrhotic liver; FNH – central scar, fibrous septae with bile
findings are non-specific and may require intact tissue
ducts and infl ammation; HA – history of oral contraceptive use,
architecture for a definitive diagnosis. Therefore, in absent bile ducts
this setting, a core biopsy accompanying the FNA is
extremely helpful (Figs 8-4 & 8-5).

Ch008-F06731.indd 221 10/26/2006 10:30:05 AM


222 FINE NEEDLE ASPIRATION CYTOLOGY

ducts are absent in HA, whereas they are present in


MRN.

FOCAL NODULAR HYPERPLASIA

CLINICAL FEATURES

An uncommon benign lesion, focal nodular hyperpla-


sia (FNH) is characterized by hyperplastic hepatocytes
and associated stellate scar. The lesion occurs predomi-
nantly in adult women (3rd to 5th decades of life; M : F
= 8 : 1); however, cases have been reported in children.
It has been recently suggested that FNH results from
a vascular malformation which causes a hyperplastic
hepatocellular reaction. A histopathogenetic relation-
FIGURE 8-5 ship with oral contraceptives (OCPs) has been sug-
Nodular regenerative hyperplasia. Trabecular arrangement of hepato- gested. In contrast to hepatocellular adenoma, OCPs
cytes displaying atypia. Although the N/C ratio is low, the cells have
macronucleoli. Papanicolaou stain, high power.
are not directly related to the initial development of
FNH. However, it appears that OCPs cause subsequent
increase in vascularity and enlargement of FNH, which
leads to an increased risk of rupture. Approximately
80% of FNH are solitary, and 80% are clinically asymp-
ANCILLARY STUDIES tomatic. In symptomatic cases, the clinical presentation
includes palpable mass, pain or discomfort, and, rarely,
manifestations of portal hypertension. Unlike hepato-
Immunoperoxidase staining has a limited role. CD34 cellular adenoma, FNH rarely if ever results in hemo-
may be used to help differentiate MRN from HCC: peritoneum. Liver function tests (including serum
CD34 highlights endothelial cell wrapping in HCC, AFP) are within normal limits.
whereas this feature is absent in MRNs. Some studies FNH is treated by local or segmental liver resection.
(including our own) have demonstrated that expression Small lesions or lesions that are deep seated often do not
of proliferation markers (Ki67 and PCNA) and p53 is require surgical intervention. Prognosis is excellent and
higher in HCC as compared to MRN. risk of complications (rupture or hemorrhage) for unre-
Special stain for reticulin is helpful in distinguishing sected lesions is extremely low. Radiologic studies may
MRN from HCC: reticulin stain highlights a disrupted show the central scar in 65% of the cases. Enhanced
reticulin network in HCC, whereas reticulin network in magnetic resonance imaging (MRI) is considered the
MRN is preserved. imaging modality of choice for FNH, with a high sensi-
tivity and specificity, often making a tissue diagnosis
unnecessary in these cases.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

FOCAL NODULAR HYPERPLASIA – DISEASE FACT SHEET


The most important differential diagnosis for MRN is
a well-differentiated hepatocellular carcinoma (WD-
Incidence
HCC). Both MRN and WD-HCC arise in a cirrhotic
៉ Uncommon
liver and may look identical radiographically. The
distinction is based primarily on an increased cell-
Gender and Age Distribution
plate thickness and accompanying loss of reticulin
៉ Male to female ratio is 8 : 1
framework in HCC. Cytologic atypia, if present, favors
៉ Adults (usually 30–50 years old)
HCC, but atypical features may be subtle in well-
differentiated cases. Clinical Features
MRN is difficult or impossible to definitively distin-
៉ Normal AFP
guish from hepatocellular adenoma (HA) and focal
nodular hyperplasia (FNH) based on cytologic examina- Prognosis
tion alone. Clinical features may be helpful: in contrast
៉ Benign; no known association with hepatocellular carcinoma
to MRN, both FNH and HA arise in non-cirrhotic liver. ៉ Hemoperitoneum rare (unlike hepatocellular adenoma)
In addition, FNH frequently displays a central scar,
which may be apparent radiographically. Finally, bile

Ch008-F06731.indd 222 10/26/2006 10:30:05 AM


CHAPTER 8 Liver 223

PATHOLOGIC FEATURES

Grossly, FNH has a very characteristic appearance:


circumscribed, bulging, lobulated, with a firm to
rubbery consistency and multinodular cut surface due
to multiple stellate scars. Most lesions present as a soli-
tary mass and measure less than 5 cm.
Histologic examination shows thick fibrous bands
dividing hyperplastic but normal-appearing hepatocytes
into nodules. Fibrous septae contain prominent bile
ductule proliferations and inflammatory infiltrate.
Hepatocyte plates are not thickened (one to two cells
thick). No portal tracts or central veins are present
within the hepatocellular nodules.
Cytopathologic evaluation does not show any specific
characteristics. When FNH is suspected on radiologic
and clinical examination, the cytopathologic analysis
can only be supportive of such lesion. In our experience, FIGURE 8-6
Focal nodular hyperplasia. Abundant fibrosis representing sampling of
the central stellate scar and fragments of reactive hepatocytes. Papa-
nicolaou stain, low power.

FOCAL NODULAR HYPERPLASIA – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Normal-appearing hepatocytes (most often large fragments)
៉ Biliary epithelium (slightly increased in amount)
៉ Fragments of fibrous tissue
៉ Absence of endothelial/vascular proliferation
៉ Lack of atypia (as opposed to cirrhotic liver)
៉ Cell block/tissue core may be extremely helpful

Histopathologic Findings
៉ Gross:
៉ Focal lesion in the background of non-cirrhotic liver
៉ Central stellate scar is often present
៉ Microscopic:
៉ Bands of fibrosis that separate bland hepatocytes into
nodules
៉ Abnormally thick-walled vessels often in the center of the
lesion FIGURE 8-7
៉ Infl ammation and bile ductule proliferation within the fibrous
Focal nodular hyperplasia. Smear shows abundance of biliary epithelium
septae
and unremarkable hepatocytes. The findings are non-specific. Diff Quik
៉ Cell plates are not thickened (1–2 cells)
stain, intermediate power.
៉ No portal tracts or central veins

Ancillary Studies
៉ CD34: no endothelial cell wrapping (unlike hepatocellular
carcinoma [HCC]) rarely if ever is a definitive diagnosis of FNH is made
៉ Reticulin stain: reticulin framework intact (unlike HCC)
on cytology alone. Cytomorphologic changes, therefore,
are non-specific (Figs 8-6 & 8-7).
Differential Diagnosis and Pitfalls
៉ Well-differentiated HCC: arises in cirrhotic liver; increased cell
plate thickness (>2 cells); disrupted reticulin framework;
cytologic atypical present, but may be minimal
ANCILLARY STUDIES
៉ Macroregenerative nodule: arises in cirrhotic liver; cannot be
distinguished from focal nodular hyperplasia based on cytology
alone
As for MRN, CD34 may help differentiate FNH from
៉ Hepatocellular adenoma: like FNH, arises in non-cirrhotic liver;
strong association with oral contraceptive use; cannot be
well-differentiated hepatocellular carcinoma (WD-
distinguished from FNH based on cytology alone HCC). As is the case for MRN, reticulin may be helpful
in differentiating FNH (reticulin framework intact)
from WD-HCC (reticulin framework disrupted).

Ch008-F06731.indd 223 10/26/2006 10:30:06 AM


224 FINE NEEDLE ASPIRATION CYTOLOGY

DIFFERENTIAL DIAGNOSIS AND PITFALLS


TABLE 8-6
Cystic Lesions of the Liver
Similarly to MRN, the most important differential
diagnosis for FNH is WD-HCC. The distinction is
• Congenital/simple cyst
based predominantly on cytologic atypia in HCC. In • Ciliated foregut cyst
addition, unlike WD-HCC, FNH has an intact reticulin • Hydatid cyst
framework and a normal cell-plate thickness. • Abscess
Based on cytologic parameters alone, FNH cannot be • Granulomatous disease
distinguished from hepatocellular adenoma (HA) and • Cystic neoplasms
MRN in cirrhosis. Clinical features, such as background
cirrhosis (favors MRN), history of OCP use (favors
HA), or presence of stellate scar (favors FNH), may be
helpful in this differential diagnosis.

TABLE 8-7
CYSTIC LESIONS OF THE LIVER Parasitic Cysts of the Liver

• Hydatid cyst (Echinococcus granulosus)


These are distinctly rare compared to more common • Schistosoma mansoni
• Entamoeba histolytica
solid mass-forming diseases and may include non-
neoplastic diseases, i.e. pseudotumors (e.g. parasitic
cysts) and, rarely, cystic neoplasms (Tables 8-6 & 8-7).

disease can involve all ages and there is no gender


HYDATID DISEASE predilection. Cysts are asymptomatic until they enlarge
to the size of at least 10 cm. Clinical manifestations of
hepatic cysts include hepatomegaly, obstructive jaun-
CLINICAL FEATURES
dice, and cholangitis. If untreated, infection can be
fatal. Treatment is surgical removal of cysts or percu-
Hydatid disease is caused by the flat tapeworm Echi- tanous drainage and treatment of the cyst with scoli-
nococcus granulosus. It is the most common cause of cidal agents such as hypertonic saline or alcohol.
hepatic cysts worldwide, particularly in the Middle The treatment is usually in parallel with the use of
East. In the United States, the disease primarily affects antihelminthic medication to prevent recurrence of
immigrants. Approximately 75% of infected patients disease.
develop hepatic cysts, which may be multiple and pre- Ultrasonography of the liver lesions demonstrates
dominantly located in the right hepatic lobe. Hydatid cystic membranes, septa, and hydatid sand. CT and MRI
scan show cyst wall calcification and cyst wall defects,
as well as the passage of contents through a defect.

HYDATID DISEASE – DISEASE FACT SHEET PATHOLOGIC FEATURES


Incidence
៉ Most common hepatic cysts worldwide Grossly, the cysts are usually unilocular and solitary.
៉ A common disease in the Middle East Circular ‘egg-shell’ calcification of the outer layer of the
៉ Primarily affects immigrant population in the United States cyst is present in 25% of cases.
៉ Disease is caused by Echinococcus granulosus Histologically, the cyst contains a laminated mem-
brane and is filled with colorless fluid with acellular
Gender and Age Distribution
debris and occasional protoscolices (future heads of the
៉ No gender predilection
adult tapeworm) and loose hooklets. Dense fibroinflam-
៉ Any age can be affected
matory tissue with prominent eosinophils surrounds
the cyst. Cytopathologic findings on FNA include frag-
Treatment
ments of scolices or hooklets (Figs 8-8 & 8-9). The
៉ Surgical removal or percutanous drainage of cyst
hooklets stain positive for modified acid-fast bacilli
៉ Treatment of the cyst with scolicidal agents
៉ Systemic therapy with antihelminthic medication
(AFB) stain. Cyst wall may be highlighted by periodic
acid–Schiff (PAS) and Gomori methenamine silver
(GMS).

Ch008-F06731.indd 224 10/26/2006 10:30:07 AM


CHAPTER 8 Liver 225

FIGURE 8-8 FIGURE 8-9


Hydatid disease. ‘Shark tooth’ appearance of a loose hooklet in a dirty Hydatid disease. Dense granular debris with an embedded protoscolex.
smear background. Diff Quik stain, high power. Papanicolaou stain, high power.

CILIATED HEPATIC FOREGUT CYST

HYDATID DISEASE – PATHOLOGIC FEATURES


CLINICAL FEATURES
Cytopathologic Findings
៉ Scolices or hooklets Ciliated foregut cysts are extremely rare. Only several
៉ Hooklets stain positive with modified AFB stain dozen case reports were reviewed by the Armed Forces
Institute of Pathology (AFIP). The average age is 48
Histopathologic Findings years, with an equal incidence in males and females.
៉ Gross: Malignant transformation is exceptionally rare. Only
៉ Usually solitary, unilocular
one case of a squamous cell carcinoma arising in a
៉ ’Egg-shell’ calcifications of the cyst wall (25%)
ciliated foregut cyst has been reported.
៉ Microscopic:
៉ Laminated cyst membrane
៉ Cyst fl uid with acellular debris and occasional protoscolices
and loose hooklets
PATHOLOGIC FEATURES
Ancillary Studies
៉ PAS and GMS: highlight cyst wall
Ciliated foregut cyst represents a developmental
៉ AFB: highlights hooklets
anomaly. It is thought to arise from displaced embry-
onic foregut differentiating toward bronchial epithe-
Differential Diagnosis and Pitfalls
lium. Ciliated foregut cyst is usually subcapsular,
៉ Other parasitic diseases: Schistosoma, Clonorchis, Entamoeba
solitary, and unilocular. It is lined by respiratory-type
៉ Non-parasitic solitary cysts: simple unilocular cyst, ciliated
foregut cyst, pseudocyst, neoplastic cysts (bile duct
epithelium consisting of ciliated columnar and mucous
cystadenoma and bile duct cystadenocarcinoma) cells. Cyst fluid is clear or, less commonly, mucinous.

CILIATED HEPATIC FOREGUT CYST – DISEASE FACT SHEET

Incidence
DIFFERENTIAL DIAGNOSIS AND PITFALLS ៉ Extremely rare

Gender and Age Distribution


The differential diagnosis includes other parasitic dis-
៉ Equal gender distribution
eases that present as hepatic mass lesions, including ៉ Average age 48 years
Schistosoma, Clonorchis, and Entamoeba. Non-parasitic
solitary cysts are also considered in the differential Prognosis
diagnosis. These include simple unilocular cyst, ciliated ៉ Malignant transformation is exceedingly rare
foregut cyst, pseudocysts, and neoplastic cysts (bile duct
cystadenoma and bile duct cystadenocarcinoma).

Ch008-F06731.indd 225 10/26/2006 10:30:07 AM


226 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 8-11
FIGURE 8-10 Ciliated foregut cyst. Cell block section shows the partially intact cyst
Ciliated foregut cyst. Fragments of glandular epithelium with focal wall with fibrous capsule overlying hepatic tissue. Cells are pseudo-
ciliated cell differentiation (arrow). Possibility of bronchial epithelial stratified with ciliated lining. H&E stain, low power.
contamination should be considered in such cases before rendering a
definitive diagnosis. Papanicolaou stain, high power.
tamination by bronchial tissue (in subdiaphragmatic
lesions) should also be kept in mind.
CILIATED HEPATIC FOREGUT CYST – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Clear to viscid material
BENIGN TUMORS OF THE LIVER
៉ Clusters and fragments of tall columnar cells, often with well-
defined ciliated borders
៉ Numerous foamy macrophages HEMANGIOMA (CAVERNOUS HEMANGIOMA)
៉ Mucinous background

Histopathologic Findings CLINICAL FEATURES


៉ Gross:
៉ Subcapsular, solitary, and unilocular
៉ Microscopic: Hemangioma is the most common benign hepatic tumor
៉ Lined by respiratory-type epithelium (ciliated columnar and and the liver is the most common visceral site of
mucous cells) hemangioma. The lesion is usually small and asymp-
៉ Clear, less commonly mucinous, cyst fl uid
tomatic, discovered incidentally during radiographic
evaluation for other diseases. Hemangiomas are found
Differential Diagnosis and Pitfalls
in 1–7% of autopsies. Hemangioma may present at any
៉ Other solitary hepatic cysts: simple unilocular cyst, pseudocyst,
age, with the mean age at diagnosis being 46 years.
bile duct cystadenoma and bile duct cystadenocarcinoma,
parasitic cysts (e.g. echinococcal)
They are more commonly found in women than in

HEMANGIOMA (CAVERNOUS HEMANGIOMA) –


On FNA, the aspirate reveals clear to viscid material. DISEASE FACT SHEET
Clusters of tall columnar cells, often with well-defined
ciliated borders, are noted, as well as numerous foamy Incidence
macrophages, often in a mucinous background (Figs ៉ Most common benign hepatic tumor
8-10 & 8-11).
Gender and Age Distribution
៉ More common in women
៉ Any age (mean age at diagnosis, 46 years)
DIFFERENTIAL DIAGNOSIS AND PITFALLS
Prognosis and Treatment
៉ Usually asymptomatic, discovered incidentally
The differential diagnosis for ciliated foregut cyst
៉ Spontaneous rupture is very uncommon
includes other solitary hepatic cysts: simple unilocular ៉ No resection required for asymptomatic hemangioma
cyst, pseudocyst, bile duct cystadenoma, and bile duct
cystadenocarcinoma. The possibility of needle con-

Ch008-F06731.indd 226 10/26/2006 10:30:08 AM


CHAPTER 8 Liver 227

men. Spontaneous rupture is a very uncommon com-


plication of hemangiomas, occurring predominantly
with giant hemangiomas (>4 cm in size). Asymptom-
atic hemangiomas do not require surgical resection.

PATHOLOGIC FEATURES

Hemangiomas are typically solitary, less often multiple.


Most commonly they are located in a subcapsular
region. Lesions are grossly dark red with a spongy
consistency.
Histologically, hemangiomas are characterized by
anastomosing dilated thin-walled vessels, which may
have intramural thrombi. Vessels are lined by flat bland-
appearing endothelial cells. Areas of central scarring
and hyalinization are typical. Large ‘feeder vessels’ are FIGURE 8-12
occasionally present at the periphery.
Hemangioma. Cohesive fragment of stromal tissue with uniform fusi-
Cytopathologic features include: extremely bloody form nuclei and background fresh blood. Vascular endothelium is not
smears (fresh blood), scant cellularity (‘unyielding usually evident. Papanicolaou stain, intermediate power.
FNAs’), fragments of acellular fibrous tissue, occasional
small, loosely arranged fragments of vascular endothe-

HEMANGIOMA (CAVERNOUS HEMANGIOMA) –


PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Extremely bloody smears (fresh blood)
៉ Scant cellularity (‘unyielding FNAs’)
៉ Fragments of fibrous stromal tissue
៉ Occasional small fragments of vascular endothelium with
slender to plump, fusiform, often grooved nuclei, loosely
arranged
៉ Well-formed vascular channels rarely observed
៉ Cell blocks usually diagnostic

Histopathologic Findings
៉ Gross: FIGURE 8-13
៉ Usually solitary, but may be multiple
Hemangioma. Single, bland-appearing, spindled nuclei with long taper-
៉ Usually subcapsular ing cytoplasmic processes representing stromal cells of the tumor. Diff
៉ Appear dark red with a spongy consistency Quik stain, high power.
៉ Microscopic:
៉ Anastomosing dilated thin-walled vessels
៉ Bland fl at endothelial cells
៉ Intramural thrombi
lium with slender to plump, fusiform, often grooved
Ancillary Studies
nuclei. Well-formed vascular channels are rarely
observed. Cell blocks are usually diagnostic and depict
៉ Endothelial markers: CD34, CD31, Factor VIII, and Ulex
europaeus
tissue fragments with interconnecting vascular chan-
៉ Electron microscopy: Weibel-Palade bodies nels (Figs 8-12–8-14).

Differential Diagnosis and Pitfalls


៉ Other vascular lesions: angiosarcoma, epithelioid
hemangioendothelioma
ANCILLARY STUDIES
៉ Non-vascular spindle cell lesions: leiomyosarcoma,
granulomatous hepatitis, spindle cell-predominant
angiomyolipoma
The vascular nature of the lesion may be confirmed by
immunohistochemical stains including CD34, CD31,
and Factor VIII.

Ch008-F06731.indd 227 10/26/2006 10:30:09 AM


228 FINE NEEDLE ASPIRATION CYTOLOGY

reproductive age who have a history of OCP use. The


majority of these women are under 30 years of age. In
some cases, OCP use may be remote. Save for OCP use,
hepatic adenomas are seen in only a few other clinical
settings, including patients with androgen use and
patients with glycogen storage diseases (types I and
III).
Unlike focal nodular hyperplasia, HAs are more
often symptomatic. Complication of HA includes intra-
tumoral hemorrhage, which may present with acute
abdominal pain. In addition, HA may rupture through
the liver capsule, causing intraperitoneal hemorrhage
and shock.
Malignant transformation to HCC has traditionally
been considered highly unusual. However, more recent
studies estimate that about 10% of unresected HAs
evolve into HCC. Resection is generally recommended.
FIGURE 8-14
Hemangioma. Core biopsy accompanying one of the aspiration samples
clearly displays interconnecting vascular lumina with attenuated endo-
thelial lining supported by a sclerotic stroma. H&E stain, low power.
PATHOLOGIC FEATURES

DIFFERENTIAL DIAGNOSIS AND PITFALLS HAs arise in non-cirrhotic livers and typically present
as solitary lesions, but occasionally patients may present
with multiple adenomas. Histopathologically, HAs
The differential diagnosis includes other spindle cell
lesions of the liver, most common of which are metastatic
leiomyosarcoma, granulomatous hepatitis, and spindle
cell-predominant angiomyolipoma. Other vascular
lesions, such as angiosarcoma and epithelioid heman- HEPATOCELLULAR ADENOMA – PATHOLOGIC FEATURES
gioendothelioma, are also in the differential diagnosis.
Cytopathologic Findings
៉ Could be entirely non-specific
៉ Abundant normal-appearing hepatocytes
HEPATOCELLULAR ADENOMA ៉ Lack of biliary epithelium, fibrous tissue fragments
៉ Lack of endothelial/vascular proliferation

CLINICAL FEATURES Histopathologic Findings


៉ Gross:
៉ Solitary, rarely multiple
Overall, hepatocellular adenomas (HAs) are rare. The ៉ Background non-cirrhotic liver
estimated yearly incidence is 3 to 4 cases per 100,000 ៉ Microscopic:
women with a history of long-term oral contraceptive ៉ Well-circumscribed, majority encapsulated
៉ Hepatocytes lack normal acinar structure: no portal tracts
(OCP) use. Over 90% of HAs develop in women of
៉ Atypia is absent or minimal
៉ Normal cell-plate thickness (<2 cells)
៉ Reticulin framework intact

HEPATOCELLULAR ADENOMA – DISEASE FACT SHEET Ancillary Studies


៉ CD34: no endothelial cell wrapping (unlike hepatocellular
Incidence
carcinoma [HCC])
៉ Rare: per year, 3–4 cases per 100,000 women with a history of ៉ Reticulin stain: reticulin framework intact (unlike HCC)
long-term oral contraceptive (OCP) use
Differential Diagnosis and Pitfalls
Gender and Age Distribution
៉ Well-differentiated HCC: arises in cirrhotic liver; increased cell
៉ 90% of cases are women with a history of OCP use plate thickness (>2 cells); disrupted reticulin framework;
៉ Most patients are under 30 years of age cytologic atypia present, but may be minimal
៉ Focal nodular hyperplasia: cannot be distinguished from
Prognosis and Treatment adenoma based on cytology alone; stellate scar; bile ducts
៉ May cause intratumoral and intraperitoneal hemorrhage present in fibrous septae
៉ 10% association with hepatocellular carcinoma ៉ Macroregenerative nodule: arises in cirrhotic liver; cannot be
៉ Resection generally recommended distinguished from adenoma based on cytology alone

Ch008-F06731.indd 228 10/26/2006 10:30:09 AM


CHAPTER 8 Liver 229

macroregenerative nodule is generally impossible based


on cytologic parameters alone.

BILE DUCT ADENOMA

CLINICAL FEATURES

Bile duct adenoma is found incidentally in approxi-


mately 30% of autopsies. These lesions are also fre-
quently discovered during intra-abdominal surgery for
other disease (70% in one study). Bile duct adenoma
can occur at any age (mean age, 55 years), with a similar
rate in men and women. Bile duct adenoma is always
discovered incidentally. The behavior of this lesion is
benign.
FIGURE 8-15
Hepatic adenoma. Bland-appearing hepatocytes with a slightly disor-
ganized architecture and lacking the second cell component, i.e. biliary
epithelium. Diff Quik stain, low power.
PATHOLOGIC FEATURES

Bile duct adenomas are solitary in the vast majority of


cases. Most of the lesions are subcapsular and measure
less than 0.5 cm, not exceeding 2 cm.
consist entirely of hepatocytes and lack the normal Histologically, bile duct adenomas are well circum-
acinar architecture (bile ducts and central veins are scribed, but are not encapsulated. They are composed
absent). HAs are well-circumscribed lesions, 75% are of numerous angulated tubules with little or no lumen.
encapsulated. Background liver is always non-cirrhotic. They are lined by a single layer of bland biliary epithe-
HAs are composed of bland hepatocytes, indistinguish- lium. No atypia or mitotic figures are present. Stroma
able from normal hepatocytes. Hepatocyte plates are is generally fibrotic and hyalinized, often containing
not thickened (one to two cells thick). Increased steato- numerous inflammatory cells. Cytomorphology reveals
sis is a common feature. abundant, normal-appearing biliary epithelium, often
Cytomorphologic features are often entirely non- admixed with benign hepatocytes (needle pick-up). Cell
specific. Abundant normal-appearing hepatocytes are blocks may be helpful.
seen. There is a total lack of biliary epithelium, fibrous
tissue fragments, and endothelial/vascular proliferation
(Fig. 8-15).
ANCILLARY STUDIES

ANCILLARY STUDIES These have a limited role in the diagnosis.

As described above, CD34 may be useful to distinguish


HA from well-differentiated hepatocellular carcinoma
(WD-HCC): CD34 highlights endothelial cell wrapping
in WD-HCC, whereas this feature is absent in HA. BILE DUCT ADENOMA – DISEASE FACT SHEET
Preserved reticulin framework is helpful for distinguish-
ing HA from WD-HCC. Incidence
៉ Very common
៉ Always discovered incidentally
៉ Found incidentally in ~30% of autopsies
DIFFERENTIAL DIAGNOSIS AND PITFALLS
Gender and Age Distribution
៉ No gender predilection
The main differential diagnosis for HA is WD-HCC. ៉ Occurs at any age (mean age, 55 years)
The majority of WD-HCCs in the United States arise
in the background of cirrhosis, whereas HAs arise in Prognosis
non-cirrhotic livers. In addition, cytologic atypia is a ៉ Behavior is benign
feature of HCC but not HA. As discussed above, dis-
tinction of HA from focal nodular hyperplasia and

Ch008-F06731.indd 229 10/26/2006 10:30:10 AM


230 FINE NEEDLE ASPIRATION CYTOLOGY

BILE DUCT ADENOMA – PATHOLOGIC FEATURES HEPATOBILIARY CYSTADENOMA – DISEASE FACT SHEET

Cytopathologic Findings Incidence


៉ Abundant, normal-appearing biliary epithelium ៉ Accounts for 5% of solitary hepatic cysts
៉ Often admixed with benign hepatocytes (‘needle pick-up’)
៉ Cell blocks may be helpful Gender and Age Distribution
៉ 95% develop in women
Histopathologic Findings ៉ Adults (mean age, 45 years)
៉ Gross:
៉ Majority are solitary Prognosis and Treatment
៉ Usually subcapsular ៉ May develop into a malignancy
៉ Usually <0.5 cm; always <2 cm ៉ May show delayed recurrence
៉ Microscopic: ៉ Complete surgical resection is recommended
៉ Well circumscribed, but unencapsulated
៉ Angulated tubules with little or no lumen
៉ Lined by a single layer of bland biliary epithelium
៉ No atypia or mitotic figures
៉ Stroma fibrotic and hyalinized PATHOLOGIC FEATURES
៉ Prominent stromal infl ammation
៉ No intraluminal bile
Hepatobiliary cystadenomas are generally solitary,
Differential Diagnosis and Pitfalls ranging in size from 2.5 to 28 cm. The cysts are
៉ Cholangiocarcinoma and metastatic adenocarcinoma: cytologic grossly multilocular and generally contain mucinous
atypia present; may be larger than 2 cm secretions.
៉ Von Meyenburg complexes: not a solitary lesion (present Benign hepatobiliary cystadenomas are lined by a
diffusely); lumina dilated; contain intraluminal bile single layer of cuboidal or tall columnar mucinous
epithelium, similar to bile duct or gastric foveolar
epithelium. In women, the surrounding stroma shows
distinctive dense spindle cells, resembling ovarian
stroma. These stromal cells are immunoreactive for
DIFFERENTIAL DIAGNOSIS AND PITFALLS estrogen receptors (ER) and progesterone receptors
(PR). These lesions are histologically identical to muci-
nous cystadenomas of other organs.
The main differential diagnoses for bile duct adenoma
are intrahepatic cholangiocarcinoma and metastatic
adenocarcinoma. The distinction is based on the pre-
sence of cytologic atypia in the carcinomas, but not bile HEPATOBILIARY CYSTADENOMA – PATHOLOGIC FEATURES
duct adenoma.
Von Meyenburg complexes may look histologically Cytopathologic Findings
and cytologically similar to bile duct adenoma. However, ៉ Single population of benign-appearing ductal epithelium
unlike the solitary bile duct adenoma, von Meyenburg ៉ Monotonous with uniform round nuclei devoid of nucleoli
complexes are present diffusely throughout the liver. In ៉ Mucinous differentiation with goblet cells
៉ Background of extracellular mucin
addition, ductal lumina of von Meyenburg complexes
are generally dilated and contain bile plugs, whereas
Histopathologic Findings
these features are not present in bile duct adenoma.
៉ Gross:
៉ Solitary
៉ Size range 2.5 to 28 cm
៉ Multilocular
HEPATOBILIARY CYSTADENOMA ៉ Contain mucinous secretions
៉ Microscopic:
៉ Mucinous epithelial lining (bile duct or gastric foveolar-type)
CLINICAL FEATURES ៉ Ovarian-type spindle cell stroma
៉ Epithelial cells are bland (no dysplasia)

Hepatobiliary cystadenoma accounts for 5% of solitary


Ancillary Studies
hepatic cysts, with almost 95% developing in women.
៉ Ovarian-type stroma: ER- and PR-reactive
Virtually all cases are from adults, with a mean age at
diagnosis of 45 years. Patients usually present with
Differential Diagnosis and Pitfalls
abdominal pain and, occasionally, jaundice. Develop-
៉ Hepatobiliary cystadenocarcinoma: epithelial dysplasia; stromal
ment of malignancy is a recognized complication of
invasion; must be differentiated from metastases (pancreas,
cystadenoma. Complete surgical resection is the treat- ovary, appendix)
ment of choice, which is generally curative. Delayed
recurrence has been reported.

Ch008-F06731.indd 230 10/26/2006 10:30:10 AM


CHAPTER 8 Liver 231

DIFFERENTIAL DIAGNOSIS AND PITFALLS

Benign hepatobiliary cystadenoma should be distin-


guished from hepatobiliary cystadenocarcinoma. This
distinction is based on the presence of high-grade dys-
plasia and areas of overt stromal invasion in the carci-
noma. Metastases from other sites, such as pancreas,
ovary, or appendix, should be excluded.

INFLAMMATORY MYOFIBROBLASTIC TUMOR


(INFLAMMATORY PSEUDOTUMOR)

CLINICAL FEATURES
FIGURE 8-16
Hepatobiliary cystadenoma. A tight, three-dimensional fragment of
ductal-type epithelium. Cells have high N/C ratios, some appearing as Inflammatory myofibroblastic tumor (IMT) is a lesion
naked nuclei. Nuclei are round, uniform, and lack nucleoli. No back- of unclear etiology, currently thought to represent a
ground hepatocytes are seen. Papanicolaou stain, high power. neoplastic process in most instances. IMT affects
various organs, most commonly lung. Liver is an
unusual site of involvement by IMT. IMT presents in
adults (mean age, 37 years; range, 10 months to 83
years), with a male to female ratio of 3 : 1. Patients may
present with symptoms of vague abdominal pain,
weight loss, and low-grade fever. Surgical resection is
curative. There are case reports of subsequent recur-
rence as malignant sarcoma.

PATHOLOGIC FEATURES

IMTs are generally solitary and well circumscribed,


varying in size from 1 to 25 cm. The tumor is composed
of plump spindle-shaped myofibroblasts, interspersed
with a prominent inflammatory infi ltrate consisting of
plasma cells, histiocytes, lymphocytes, and granulo-
cytes. Mitotic rate may be brisk, but no atypical mitotic
figures should be present.
On FNA, IMT shows hypercellular smears with an
FIGURE 8-17 admixture of various cell types (Figs 8-18 & 8-19),
Hepatobiliary cystadenoma. Cell block section displays well-formed including inflammatory cells with predominance of
glandular epithelium with mucinous differentiation. Numerous goblet
cells are seen. H&E stain, intermediate power.
plasma cells, fibroblastic proliferation, granulation tissue

INFLAMMATORY MYOFIBROBLASTIC TUMOR (INFLAMMATORY


On FNA, the smears show a single population of PSEUDOTUMOR) – DISEASE FACT SHEET
benign-appearing ductal-type glandular epithelium.
Cells are monotonous with uniform round nuclei devoid Incidence
of nucleoli (Figs 8-16 & 8-17). Mucinous differentiation ៉ Extremely rare (less than 90 cases reported)
is evident in the form of well-developed goblet cells.
Occasionally, the smears show extracellular background Gender and Age Distribution
mucin as well. Cellular necrosis is not observed. ៉ Male to female ratio is 3 : 1
៉ Adults (mean age, 37 years; range, 10 months to 83 years)

Prognosis and Treatment


ANCILLARY STUDIES ៉ Surgical resection is curative
៉ Case reports of recurrence as malignant sarcoma

ER and PR are reactive in the ovarian-type stroma.

Ch008-F06731.indd 231 10/26/2006 10:30:10 AM


232 FINE NEEDLE ASPIRATION CYTOLOGY

INFLAMMATORY MYOFIBROBLASTIC TUMOR (INFLAMMATORY


PSEUDOTUMOR) – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Hypercellular smears with an admixture of various cell types
៉ Infl ammatory cells with predominance of plasma cells
៉ Fibroblastic proliferation, granulation tissue formation
៉ Atypical-appearing histiocytes with enlarged nuclei, occasional
nuclear grooves, and intranuclear inclusions

Histopathologic Findings
៉ Gross:
៉ Solitary
៉ Well circumscribed
៉ Size 1 to 25 cm
៉ Microscopic:
៉ Plump spindle cells
៉ Interspersed infl ammatory infiltrate (plasma cells, histiocytes,
FIGURE 8-18 lymphocytes, granulocytes)
Infl ammatory pseudotumor. Hypercellular smear with predominantly ៉ Mitotic figures allowed
single cells, comprised of a mixture of infl ammatory cells, macrophages, ៉ Should have no atypical mitoses
endothelial cells, and fibroblasts. Diff Quik stain, intermediate power.
Ancillary Studies
៉ ALK-negative (unlike IMT of some other organs, e.g. bladder)
៉ Aberrant cytokeratin reactivity

Differential Diagnosis and Pitfalls


៉ Spindle cell sarcomas (e.g. leiomyosarcoma): frank
pleomorphism; atypical mitotic figures

DIFFERENTIAL DIAGNOSIS AND PITFALLS

Many hepatic masses of various types may be associ-


ated with a brisk inflammatory response and these
lesions should be differentiated from IMT. Particularly,
IMT should be distinguished from spindle-cell sarco-
mas (e.g. leiomyosarcoma). Presence of frank pleomor-
phism and atypical mitotic features favors the latter.
FIGURE 8-19
Infl ammatory pseudotumor. Endothelial cells, macrophages with grooved
nuclei, and infl ammatory cells with predominance of plasma cells.
Papanicolaou stain, high power. MALIGNANT TUMORS OF THE LIVER
formation, and atypical-appearing histiocytes with enlar-
ged nuclei, occasional nuclear grooves, and intranuclear
Primary malignant and metastatic/secondary malig-
inclusions. Fibroblastic proliferation with mitoses may
nant tumors of the liver are listed in Table 8-8 and
mimic mesenchymal neoplasms. Cytomorphology is non-
Table 8-9, respectively.
specific and IMT is usually a diagnosis of exclusion.

PRIMARY MALIGNANT EPITHELIAL TUMORS


ANCILLARY STUDIES
HEPATOCELLULAR CARCINOMA
Anaplastic lymphoma kinase (ALK) expression has
been found in some IMTs, such as IMT involving the CLINICAL FEATURES
bladder. However, expression of ALK has not been found
in the hepatic IMT. Aberrant cytokeratin expression
has been reported in IMT of various sites and should not The most important risk factors for HCC are infection
be misinterpreted as evidence of carcinoma. with hepatitis B virus (HBV) and hepatitis C virus

Ch008-F06731.indd 232 10/26/2006 10:30:11 AM


CHAPTER 8 Liver 233

common. The estimated annual incidence of HCC in the


TABLE 8-8 United States is 4 cases per 100,000. HCC accounts for
more than 80% of primary liver carcinomas. In addition
Malignant Tumors – Primary to hepatitis viruses and alcohol, a number of chemical
carcinogens have been linked to HCC, including
• Epithelial: aflatoxin B1 (a toxic metabolite of Aspergillus flavus). A
• Hepatocellular carcinoma high risk of HCC is seen in patients with hereditary
• Cholangiocarcinoma hemochromatosis and hereditary tyrosinemia.
• Neuroendocrine neoplasms (extremely rare)
Elevated AFP is present in 50–75% of patients, with
• Non-epithelial:
• Angiosarcoma/other sarcomas values usually over 1000 ng/mL. Levels greater than
• Non-Hodgkin lymphoma/Hodgkin lymphoma 4000 ng/mL are highly suggestive of HCC. Elevated AFP
is, however, not present in all cases of HCC, and AFP
elevation may also be seen with yolk-sac tumors and
several non-neoplastic conditions, including massive
liver necrosis and fetal neural tube defects.
Epidemiology of HCC shows stark geographic vari-
TABLE 8-9 ability. In Asia and Africa, where HBV predominates,
HCC develops at a young age (20–40 years); cirrhosis is
Malignant Tumors – Metastatic/Secondary present in less than 50% of cases. In contrast, in the
United States, where HCV predominates, HCC rarely
• Epithelial: develops before the age of 60 and cirrhosis is present in
• Adenocarcinoma 75–90% of cases. In the United States, the average age
• Squamous cell carcinoma at the time of HCC diagnosis is 61 years. Worldwide,
• Small cell carcinoma HCC is more common in men than in women (M : F
• Other (adenoid cystic carcinoma, renal cell carcinoma,
etc.)
ratio is 2 : 1 to 5 : 1).
• Non-epithelial: The prognosis is extremely poor, with mean survival
• Malignant melanoma of 11 months. The factors that influence survival are
• Granulosa cell tumor age (older patients do worse), gender (men do worse
• Sarcomas than women), tumor stage and grade, and presence of
• Non-Hodgkin lymphoma/Hodgkin lymphoma cirrhosis (worse prognosis if cirrhosis is present).
• Other

HEPATOCELLULAR CARCINOMA – DISEASE FACT SHEET


PATHOLOGIC FEATURES

Incidence
Grossly, HCC may be solitary, multinodular, or diffuse.
៉ >80% of primary hepatic malignancies In cirrhotic liver, HCC tends to present as a solitary
៉ United States: 4 cases per 100,000
mass with multiple satellite nodules. The tumor ranges
៉ Asia, Africa: higher rates (80% of cases arise in Asia)
in size from those at the threshold of radiographic
Gender and Age Distribution
detection to those replacing the entire liver.
Histologically, HCC is characterized by the loss of
៉ Male to female ratio is between 2 : 1 and 5 : 1
៉ Asia and Africa: 20–40 years (HBV; cirrhosis in <50%)
normal liver architecture: hepatocyte plates are thick-
៉ USA: 61 years (HCV; cirrhosis in 75–90%)
ened (greater than two cells thick) and the normal
acinar structure, including portal tracts and central
Clinical Features veins, is absent. The neoplastic cells show crowding and
៉ Elevated AFP (>4000 ng/mL highly suggestive of HCC)
cytologic atypia, which increases in severity in higher-
grade lesions. Vascular invasion is present in two of
Prognosis three cases. Presence of cytoplasmic bile pigment and
៉ Extremely poor (mean survival, 11 months)
lipid vacuoles (steatosis) is helpful for differentiating
primary HCC from metastatic tumors.
Multiple architectural patterns may be present,
including trabecular, glandular, and solid. Cytologic
variants include pleomorphic (giant cell), clear cell,
(HCV). The most common cause of HCC worldwide is oncocyte-like, and sarcomatoid HCC (Table 8-10).
HBV, particularly in the areas where HBV infection is Cytologic grading of HCC is helpful for a number of
endemic, including Asia and Africa. Nearly 80% of all reasons (Table 8-11). A three-tier system of FNA grading
HCCs develop in Asian countries. is employed at Johns Hopkins: well-differentiated (Figs
In the United States, the most important risk factors 8-20–8-22), moderately differentiated (Figs 8-23 & 8-
for HCC are HCV and chronic alcoholism; HBV is less 24), and poorly differentiated (Figs 8-25–8-28).

Ch008-F06731.indd 233 10/26/2006 10:30:12 AM


234 FINE NEEDLE ASPIRATION CYTOLOGY

HEPATOCELLULAR CARCINOMA – PATHOLOGIC FEATURES TABLE 8-10


Cytopathologic Findings Hepatocellular Carcinoma – Uncommon Variants
៉ Well-differentiated HCC:
៉ Hypercellularity • Clear cell
៉ Bland-appearing hepatocytes, minimally pleomorphic, mildly • Mixed hepatocellular–cholangiocarcinoma
increased N/C ratio, small nucleoli • Acinar or adenoid cell
៉ Predominantly cohesive fragments, trabecular growth pattern, • Fibrolamellar
often smooth round fragment outline • Giant cell
៉ Prominent bile staining • Sarcomatoid
៉ Vascular proliferation associated with loosely hanging nests • Osteoclastic
of hepatocytes • Sclerotic
៉ Moderately differentiated HCC:
៉ Hypercellular smears, cohesive nests or trabeculae
៉ Fragments with irregular edges, numerous single cells
៉ High N/C ratio, irregular nuclear membrane
៉ Numerous atypical naked nuclei
៉ Prominent nucleoli, intranuclear inclusions
៉ Vascular/endothelial proliferation present (less obvious than
in well-differentiated HCC)
៉ Poorly differentiated HCC:
៉ Extremely hypercellular
៉ Loose nests, occasional 3-D fragments (gland-like)
៉ Marked pleomophism, high N/C ratio, macronucleoli
៉ Endothelial wrap-arounds ‘packeting’
៉ Large naked nuclear population
៉ Tumor giant cells
៉ Vascular/endothelial proliferation rarely seen
៉ Numerous mitoses, necrosis

Histopathologic Findings
៉ Gross:
៉ Solitary, multinodular, or diffuse
៉ Size ranges from small to massive
៉ Microscopic:
៉ Thickened hepatocyte plates (>2 cells thick) FIGURE 8-20
៉ Absence of normal acinar structure (no portal tracts)
Hepatocellular carcinoma. Well-differentiated tumor with irregular
៉ Cellular crowding and cytologic atypia nests of malignant hepatocytes loosely arranged around arborizing
៉ Cytoplasmic bile and lipid vacuoles (steatosis) may be present capillaries. Few single cells are seen as well. Papanicolaou stain, low
power.
Ancillary Studies
៉ HepPar-1, AFP, canalicular polyclonal CEA and CD10
៉ Reticulin highlights the loss of reticulin framework and the
expansion of cell plates
៉ Mucicarmine-negative

Differential Diagnosis and Pitfalls


៉ Well-differentiated HCC:
៉ Normal liver
៉ Hepatocellular adenoma (HA), focal nodular hyperplasia
(FNH), macroregenerative nodule (MRN): hepatocyte plates
not thickened (<2 cells thick); reticulin framework intact;
for HA and FNH, background liver non-cirrhotic
៉ Metastatic tumors: renal cell carcinoma, adrenal cortical
carcinoma
៉ Moderately differentiated HCC:
៉ Metastatic tumors: renal cell carcinoma, melanoma,
adenocarcinoma
៉ Poorly-differentiated HCC:
៉ Cholangiocarcinoma: no bile, no lipid (steatosis); prominent
desmoplasia; mucin; HepPar-1-negative, usually CK7 and CK20
double-positive FIGURE 8-21
៉ Metastatic carcinoma, melanoma, sarcoma, lymphoma: no Hepatocellular carcinoma. Well-differentiated tumor cells show a deli-
bile, no lipid (steatosis); mucin in some adenocarcinomas; cate attachment to fine branching capillaries. The diagnosis is often
melanin pigment in some melanomas made at low magnification, as individual cells may not appear too dif-
ferent from benign hepatocytes. Diff Quik, low power.

Ch008-F06731.indd 234 10/26/2006 10:30:12 AM


CHAPTER 8 Liver 235

TABLE 8-11
Cytologic Grading of Hepatocellular Carcinoma:
Why Do We Grade?

• Difference in clinical prognosis and survival


• Treatment options
• Differential diagnosis

FIGURE 8-24
Hepatocellular carcinoma. Moderately differentiated tumor shows a
diffuse population of naked nuclei with macronucleoli in a very granular
smear background. Papanicolaou stain, intermediate power.

FIGURE 8-22
Hepatocellular carcinoma. Well-differentiated neoplasm with large
polygonal hepatocytes anchored to vascular endothelium. Cells have a
low N/C ratio and lack macronucleoli. Papanicolaou stain, high power.

FIGURE 8-25
Hepatocellular carcinoma. Poorly differentiated tumor with a large
syncytial-like fragment of malignant cells. N/C ratio is extremely high,
with all the cells displaying macronucleoli. Morphologic kinship to
hepatocytes is lost and distinction of this lesion from other primary
and metastatic carcinomas may require immunostaining. Papanicolaou
stain, intermediate power.

nodule. In addition, increased p53 expression has


recently been shown to favor HCC.
A number of immunohistochemical stains may be
FIGURE 8-23
applied to help distinguish primary HCC from cholan-
Hepatocellular carcinoma. Moderately differentiated tumor displays giocarcinoma or metastatic carcinomas (Table 8-12).
greater pleomorphism, macronucleoli, and a sprinkling of naked nuclei
in the background. Diff Quik stain, high power. The markers that favor HCC include HepPar-1 (highly
specific), AFP (non-specific), and a canalicular pattern
of polyclonal carcinoembryonic antigen (pCEA) and
ANCILLARY STUDIES (Figs 8-29–8-32) CD10. Also, HCC is unique in its cytokeratin profile:
HCC reacts with low molecular weight cytokeratin anti-
bodies (such as CAM 5.2), but is generally non-reactive
CD34 highlights endothelial cell wrapping in HCC, and with pancytokeratin antibodies (such as AE1/AE3) or
may be helpful for distinguishing HCC from benign high molecular weight cytokeratin antibodies (such
hepatocellular lesions, including focal nodular hyper- as K903). HCC is most commonly CK7 and CK20
plasia, hepatocellular adenoma, and macroregenerative double-negative.

Ch008-F06731.indd 235 10/26/2006 10:30:13 AM


236 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 8-26 FIGURE 8-27


Hepatocellular carcinoma. Poorly differentiated tumor shows large frag- Hepatocellular carcinoma. Poorly differentiated tumor shows a three-
ment with tightly packed malignant cells reflecting a markedly expanded dimensional gland-like fragment (middle of the field) with numerous
hepatic plate. Cells display macronucleoli and are wrapped around by single naked nuclei in the background admixed with non-neoplastic
endothelium ‘cell packets’. Papanicolaou stain, intermediate power. liver. It is not possible to differentiate this from an adenocarcinoma
without immunostaining. Diff Quik stain, high power.

FIGURE 8-28 FIGURE 8-29


Hepatocellular carcinoma. Poorly differentiated tumor with thick tissue Hepatocellular carcinoma. Polyclonal CEA (pCEA) immunostain displays
fragments and abundant necrosis. Necrosis is not a usual feature and the characteristic one-sided ‘canalicular’ staining pattern of the cell
is often noted focally in high-grade HCC. Diff Quik stain, low power. surface. Intermediate power.

FIGURE 8-30
FIGURE 8-31
Cholangiocarcinoma. In contrast to hepatocellular carcinoma, the
malignant cells display diffuse immunostaining of the cytoplasm with Hepatocellular carcinoma. Immunostaining with CD31 shows the
pCEA. Intermediate power. characteristic endothelial reactivity creating ‘packets’ of malignant
hepatocytes. High power.

Ch008-F06731.indd 236 10/26/2006 10:30:14 AM


CHAPTER 8 Liver 237

Reticulin stain highlights the loss of reticulin frame-


work and the expansion of cell plates in HCC. Mucin
reactivity is vanishingly rare in HCC; mucin staining
(with mucicarmine or PAS) strongly favors cholangio-
carcinoma or metastatic adenocarcinoma over HCC.
In a recent (as yet unpublished) study, we found that
in a few cases of well-differentiated HCC where the
exclusion of macroregenerative nodule, focal nodular
hyperplasia, or hepatocellular adenoma could be diffi-
cult, p53 immunostaining can play an important role.
However, p53, although quite specific, suffers from rela-
tively low sensitivity (55% for HCC in our experience).
Results are summarized in Table 8-13.

FIGURE 8-32 DIFFERENTIAL DIAGNOSIS AND PITFALLS


Hepatocellular carcinoma. p53 immunostaining shows focal nuclear
positivity in the core biopsy of a difficult case of well-differentiated
tumor where hepatocellular adenoma was difficult to exclude based on The differential diagnosis for well-differentiated HCC
morphology alone. Intermediate power. includes other hepatocellular lesions: hepatocellular
adenoma, focal nodular hyperplasia, and macroregen-
erative nodule. The differential diagnosis for poorly
differentiated HCC includes cholangiocarcinoma as
well as metastatic neoplasms, including carcinoma,
melanoma, sarcoma, and lymphoma.

TABLE 8-12
FIBROLAMELLAR HEPATOCELLULAR
Hepatocellular Carcinoma – Immunoperoxidase Staining CARCINOMA
• Conventional markers:
• CAM 5.2 CLINICAL FEATURES
• Polyclonal CEA
• CD34/CD31
• HepPar-1 Fibrolamellar carcinoma (FLC) represents approxi-
• Vimentin mately 5% of all hepatocellular carcinomas. FLC arises
• Alpha-fetoprotein (AFP) in younger patients, with a mean age of 25 years, and
• Alpha-1-antitrypsin (AAT) has a slight female predominance. Traditionally, it has
• More recent markers: been suggested that the prognosis of FLC is better than
• TTF-1 for typical HCC. However, it appears that after correct-
• Glut-1
• MOC-31
ing for age, stage, and other factors, the difference in
• CA 15-3 prognosis may not be significant. FLC has a stronger
propensity for lymph node metastasis than has typical
HCC.

TABLE 8-13
p53 Immunoreactivity in Hepatic FNA

Hepatocellular Carcinoma Focal Nodular Hyperplasia Regenerative Nodule Hepatic Adenoma

p53 6/11 (55%) 0/5 (0%) 0/5 (0%) 0/5 (0%)


Beta-catenin 7/11 (64%) 4/6 (67%) 4/6 (67%) 5/5 (100%)
Ki-67(moderate-high) 4/11 (36%) 0/6 (0%) 0/6 (0%) 0/5 (0%)

Ch008-F06731.indd 237 10/26/2006 10:30:16 AM


238 FINE NEEDLE ASPIRATION CYTOLOGY

FIBROLAMELLAR HEPATOCELLULAR CARCINOMA –


DISEASE FACT SHEET

Incidence
៉ Represents 5% of all HCC

Gender and Age Distribution


៉ Slight female predominance
៉ Younger patients (mean age, 25 years)

Prognosis
៉ Thought to have a better prognosis than typical HCC

PATHOLOGIC FEATURES FIGURE 8-33


Hepatocellular carcinoma, fibrolamellar variant. Large pleomorphic
hepatocytes loosely arranged around branching capillaries. Distinction
Histopathologically, in contrast to the usual-type from a conventional well-differentiated hepatocellular carcinoma can
be difficult, and clinical information is crucial for accurate subtyping.
HCC, FLC arises in the background of non-cirrhotic Papanicolaou stain, low power.
liver. The tumor is characterized by dense bands of
fibrosis that tend to run in parallel arrays (lamellar
architecture).
On FNA, the smears show predominantly single
discohesive cells; trabeculae are distinctly uncommon.
The neoplastic cells are large and polygonal with
prominent nucleoli, densely granular eosinophilic
‘oncocytic-appearing’ cytoplasm, and prominent intra-
nuclear inclusions. Cytoplasmic hyaline inclusion
bodies as well as occasional cytoplasmic ‘pale bodies’
are characteristically seen. Lymphocytes and frag-
ments of loose fibroconnective tissue may also be seen
(Figs 8-33–8-35).

FIBROLAMELLAR HEPATOCELLULAR CARCINOMA –


PATHOLOGIC FEATURES

Cytopathologic Findings FIGURE 8-34


៉ Discohesive cells, trabeculae distinctly uncommon Hepatocellular carcinoma, fibrolamellar variant. Pleomorphic hepato-
៉ Large polygonal cells, prominent nucleoli, granular eosinophilic cytes, often with bizarre shapes, densely granular ‘oncocytic’ cytoplasm
cytoplasm and macronucleoli. Papanicolaou stain, high power.
៉ Prominent intranuclear inclusions
៉ Cytoplasmic hyaline inclusion bodies, cytoplasmic ‘pale bodies’
ANCILLARY STUDIES
Histopathologic Findings
៉ Background of non-cirrhotic liver
៉ Similar features as HCC, but with dense bands of lamellar The immunohistochemical profi le of FLC is similar to
fibrosis that of HCC, except that FLC is usually CK7-positive,
whereas HCC is generally CK7-negative. In addition,
Ancillary Studies AFP is generally non-reactive.
៉ Immunostaining pattern similar to HCC, except CK7 usually
positive and AFP usually non-reactive

Differential Diagnosis and Pitfalls DIFFERENTIAL DIAGNOSIS AND PITFALLS


៉ Typical HCC: background cirrhotic liver; older patients; absence
of lamellar fibrosis
៉ Cholangiocarcinoma and metastatic malignancies (see HCC)
FLC is distinguished from typical HCC based on the
above morphologic features (lamellar fibrosis) and
demographics (non-cirrhotic liver in young patients).

Ch008-F06731.indd 238 10/26/2006 10:30:17 AM


CHAPTER 8 Liver 239

setting of primary biliary cirrhosis. Patients usually


present with abdominal pain and weight loss. Unlike its
extrahepatic counterpart, intrahepatic cholangiocarci-
noma rarely presents with obstructive jaundice. Patients
with cholangiocarcinoma generally have a dismal prog-
nosis, with most patients succumbing to disease within
1 year of diagnosis. Patients die as a result of extensive
tumor burden, liver failure, gastrointestinal bleeding, or
infection. At autopsy, 75% of patients have metastases.
Treatment options include resection, but 80% of tumors
are deemed unresectable at diagnosis. Transplantation
is no longer performed, since recurrence of carcinoma
is nearly universal.

PATHOLOGIC FEATURES
FIGURE 8-35
Hepatocellular carcinoma, fibrolamellar variant. Fragment of sclerotic Cholangiocarcinoma may present either as a solitary
tissue, possibly arising from lamellar fibrosis in the tumor, accompany-
ing bizarre malignant hepatocytes. Papanicoalou stain, high power. mass or as multiple masses. Tumors are typically
firm and gritty, owing to the exuberant desmoplastic
reaction.
INTRAHEPATIC CHOLANGIOCARCINOMA Histologically, the tumor may be well circumscribed
but is never encapsulated. Tumors are usually well to

CLINICAL FEATURES
INTRAHEPATIC CHOLANGIOCARCINOMA – PATHOLOGIC FEATURES
Cholangiocarcinoma is the second most common
primary hepatic tumor after HCC; however, it is much Cytopathologic Findings
less frequent. Cirrhosis is not a risk factor. Risk factors ៉ Usually appears well differentiated
for cholangiocarcinoma include the diseases that cause ៉ Hypercellular, predominantly crowded fragments
៉ Monolayered or 3-D fragments with acinar formations
chronic inflammation of the biliary tree: primary scle-
៉ High N/C ratio, prominent nucleoli
rosing cholangitis, Caroli’s disease, hepatolithiasis, and
៉ Occasionally, pleomorphic cells, giant cells
parasitic infestation (Clonorchis sinesis or Opisthorchis ៉ Rarely, mitoses and necrosis
viverrini). Other risk factors are exposure to Thorotrast
and anabolic steroids. However, no identifiable risk Histopathologic Findings
factor is present in the majority of cases (80–90%). ៉ Gross: solitary mass or multinodular
Patients typically present with obstructive jaundice. ៉ Microscopic:
Serum elevation of CA 19-9 and CEA is typical, and ៉ Malignant glandular proliferation
AFP is not increased. ៉ Usually well to moderately differentiated
Most cases of cholangiocarcinoma occur after age 60. ៉ Prominent desmoplastic reaction
Men and women are affected similarly; however, there ៉ No cytoplasmic bile
is a strong male predominance in cases arising in the ៉ Cytoplasmic mucin nearly always present

Ancillary Studies
៉ Usually CK7 and CK20 double-positive (unlike HCC, which is CK7
and CK20 double-negative)
INTRAHEPATIC CHOLANGIOCARCINOMA – DISEASE FACT SHEET
៉ High molecular weight cytokeratin and pancytokeratin positive
(unlike HCC)
Incidence
៉ HepPar-1 and canalicular CEA and CD10 negative (unlike HCC)
៉ Second most common primary hepatic tumor
៉ Much less frequent than HCC
Differential Diagnosis and Pitfalls
៉ Hepatocellular carcinoma with a glandular pattern: bile and
Gender and Age Distribution
steatosis are hints when present; immunostains are helpful
៉ No gender predilection (see above)
៉ Majority after age 60 ៉ Metastatic adenocarcinomas: distinction generally requires
immunostains
Prognosis and Treatment ៉ Benign and reactive bile duct proliferations (e.g. bile duct
៉ Dismal prognosis (survival <1 year) adenoma): no cytologic atypia; no perineural or vascular
៉ 80% unresectable at diagnosis invasion

Ch008-F06731.indd 239 10/26/2006 10:30:17 AM


240 FINE NEEDLE ASPIRATION CYTOLOGY

moderately differentiated and a prominent desmoplastic cholangiocarcinomas show high nuclear to cytoplasmic
response is typical. In contrast to HCC, bile is not pro- (N/C) ratio cells, often in smaller fragments or single
duced. Intracytoplasmic mucin is nearly always present. cells with prominent nucleoli. Pleomorphic cells, giant
FNA shows a usually well-differentiated gland- cells, and, rarely, mitoses and necrosis are seen as well.
forming tumor (Figs 8-36–8-39). Smears are hypercel-
lular and there is a predominance of crowded cellular
fragments with a three-dimensional architecture with
acinar formations. A significant number of cases display ANCILLARY STUDIES
flat monolayered sheets with minimal nuclear enlarge-
ment and mild anisonucleosis; therefore, exclusion of
reactive biliary epithelium with atypia could become a There are no markers that are entirely specific for chol-
tricky issue. Few, if any, benign hepatocytes are present angiocarcinoma. The pattern of cytokeratin expres-
in the background, possibly representing needle sam- sion may be used to differentiate cholangiocarcinoma
pling outside the main lesion. Poorly differentiated from poorly differentiated HCC: cholangiocarcinoma is

FIGURE 8-38
Cholangiocarcinoma. Poorly differentiated tumor with large pleomor-
FIGURE 8-36 phic nuclei, some naked, with macronucleoli. Focal gland-like architec-
Cholangiocarcinoma. Well-differentiated tumor shows a single crowded ture is evident. This case needed immunostaining to prove its glandular
fragment of biliary epithelium. Nuclei are small, hyperchromatic, and origin and distinction from hepatocellular carcinoma. Diff Quik stain,
tightly packed with a vague three-dimensional architecture. Few back- high power.
ground benign hepatocytes are seen. Diff Quik stain, high power.

FIGURE 8-37
Cholangiocarcinoma. Well-differentiated tumor with a somewhat fl at
monolayered sheet of ductal-type epithelium. Cells show enlarged FIGURE 8-39
nuclei with inconspicuous nucleoli. Differentiation from reactive biliary Cholangiocarcinoma. Poorly differentiated tumor with high N/C ratio
epithelium can become a challenge in limited samples. Papanicolaou cells, macronucleoli, and three-dimensional architecture. Papanicolaou
stain, high power. stain, high power.

Ch008-F06731.indd 240 10/26/2006 10:30:18 AM


CHAPTER 8 Liver 241

typically CK7 and CK20 double-positive, whereas HCC


is usually CK7 and CK20 double-negative. Unlike HCC, HEPATOBLASTOMA – DISEASE FACT SHEET
cholangiocarcinoma reacts with pancytokeratins (e.g.
AE1/AE3) and high molecular weight cytokeratins Incidence
(e.g. K903). Unlike HCC, cholangiocarcinoma does not ៉ Most common primary hepatic tumor in young children
react with HepPar-1 and does not display canalicular ៉ Rare overall (0.5% of all pediatric neoplasms)
staining with polyclonal CEA and CD10.
Cholangiocarcinoma may be distinguished from meta- Gender and Age Distribution
static adenocarcinoma by virtue of negative staining for ៉ Male to female ratio is 2 : 1
various organ-specific markers, such as ER and PR ៉ Children <5 years of age
(breast and ovary), TTF-1 (lung and thyroid), and those
used for colon (CDX-2, CK7-negative, CK20-positive). Clinical Features
Distinction from pancreatic adenocarcinoma may be ៉ Elevated serum AFP
៉ No association with cirrhosis
aided by the loss of DPC4 expression in 40% of cases.
Prognosis and Treatment
៉ Surgical resection and chemotherapy
៉ Long-term survival is 60–70% after resection
DIFFERENTIAL DIAGNOSIS AND PITFALLS

Cholangiocarcinoma should be differentiated from


HCC with a glandular pattern. Clinically, history of The majority of hepatoblastomas occur in children
cirrhosis and elevated AFP strongly favor HCC over under 5 years of age. Few cases have been diagnosed at
cholangiocarcinoma. Bile and steatosis, when present, birth and it rarely occurs after age 15. There is a male
are helpful hints for the diagnosis of HCC over cho- predominance (M : F = 2 : 1). Patients usually present
langiocarcinoma, but frequently immunohistochemical with an abdominal mass. Patients with familial adeno-
markers are required to make a definitive diagnosis. matous polyposis have a significantly increased risk of
With the exception of colon cancer, cholangiocarci- hepatoblastoma. Serum AFP is elevated with high titers.
noma is difficult or impossible to distinguish from There is no association with cirrhosis.
metastatic adenocarcinoma from other sites based on Hepatoblastoma is managed by resection and chemo-
morphologic grounds alone. As described above, immu- therapy. With current therapy, nearly 90% of tumors
nomarkers may be helpful in distinguishing cholangio- are considered resectable, with 60–70% having long-
carcinoma from metastases from other organs. In term survival.
addition, well-differentiated cholangiocarcinoma should
be differentiated from benign and reactive bile duct
proliferations, including bile duct adenoma and bile
duct hamartoma. PATHOLOGIC FEATURES

Hepatoblastoma presents as a solitary mass, more com-


HEPATOBLASTOMA monly involving the right lobe of the liver. It measures
from 3 to 20 cm. Prominent calcifications are often
evident on gross examination.
CLINICAL FEATURES Histologically, hepatoblastomas are classified as either
epithelial or mixed epithelial–mesenchymal. Epithelial
cells resemble hepatocytes (fetal pattern) or may consist
Hepatoblastoma, although rare as such, represents the
of more poorly differentiated cells (embryonal pattern).
most common primary hepatic tumor in infants and
Osteoid production may be present prominently in the
young children (Table 8-14). Hepatoblastoma accounts
mixed epithelial–mesenchymal type of hepatoblastoma,
for about 50% of all primary hepatic malignancies in
particularly following chemotherapy. Foci of admixed
children and about 0.5% of all pediatric neoplasms.
extramedullary hematopoiesis may be found, which may
help differentiate hepatoblastoma from HCC.
Cytopathologic characteristics (Fig. 8-40) depend on
the subtype of hepatoblastoma.
TABLE 8-14
Pediatric Liver Tumors
ANCILLARY STUDIES
• Hepatoblastoma
• Embryonal sarcoma
• Fibrolamellar hepatocellular carcinoma Similar to HCC, hepatoblastoma reacts with HepPar-1
and AFP and shows canalicular staining with poly-
clonal CEA and CD10.

Ch008-F06731.indd 241 10/26/2006 10:30:19 AM


242 FINE NEEDLE ASPIRATION CYTOLOGY

DIFFERENTIAL DIAGNOSIS AND PITFALLS

In children, hepatoblastoma should be primarily dif-


ferentiated from metastases from other pediatric
malignancies, including neuroblastoma, lymphoblastic
lymphoma, Wilms tumor, and rhabdomyosarcoma.
Primary hepatic neoplasms that enter the differential
diagnosis for hepatoblastoma include undifferentiated
(embryonal) sarcoma and HCC. If an epithelial compo-
nent is present, embryonal sarcoma can be excluded.
HCC is uncommon in children under 5 years of age.
Presence of foci of extramedullary hematopoiesis may
help differentiate hepatoblastoma from HCC.

FIGURE 8-40
PRIMARY MALIGNANT MESENCHYMAL
Hepatoblastoma. Primitive-appearing small round blue cells with a high
N/C ratio, speckled chromatin, and densely pink cytoplasm. No hepato- TUMORS OF THE LIVER
cytic differentiation is evident. Diagnosis is usually straightforward due
to the young age of the patient and a markedly elevated serum AFP.
H&E stain, high power. EPITHELIOID HEMANGIOENDOTHELIOMA

CLINICAL FEATURES
HEPATOBLASTOMA – PATHOLOGIC FEATURES
Epithelioid hemangioendothelioma (EHE) is an uncom-
Cytopathologic Findings mon tumor arising in the liver and various other organs.
៉ Depend on the subtype The risk factors for EHE are not well characterized.
៉ Epithelial (fetal, embryonal): resemble HCC – hypercellular Oral contraceptive use and vinyl chloride exposure
smears with cords, nests, or trabeculae of small crowded have been implicated in isolated cases. EHE most com-
monotonous hepatocytes; cells often oval to spindle-shaped, monly develops in middle-aged women.
high N/C ratio, frequent mitosis Patients generally present with abdominal pain,
៉ Mixed epithelial–mesenchymal: more pleomorphic; also contains
weight loss, and, occasionally, hepatic rupture. In approx-
a neoplastic mesenchymal component (cartilage, bone, or
imately 40% of cases, EHE is found incidentally. Clinical
skeletal muscle)
៉ Anaplastic/small cell undifferentiated: hypercellular smears with
course is unpredictable, although overall survival rates
uniform primitive-appearing small blue cells, high mitotic are better than for angiosarcoma. Survival ranges from
index, karyorrhexis, necrosis several months to 27 years. Treatment of EHE is a con-
troversial subject. Resection is generally the treatment of
Histopathologic Findings choice. Extrahepatic involvement of other organs, such
៉ Gross: as lung, has been found in nearly 50% of cases.
៉ Usually solitary
៉ Prominent calcifications frequent
៉ Microscopic:
៉ Epithelial or epithelial-mesenchymal types
៉ Prominent osteoid production, particularly after EPITHELIOID HEMANGIOENDOTHELIOMA – DISEASE FACT SHEET
chemotherapy
៉ Foci of extramedullary hematopoiesis Incidence
៉ Rare
Ancillary Studies
៉ Similar to HCC: positive for HepPar-1, AFP, canalicular polyclonal Gender and Age Distribution
CEA and CD10
៉ Most common in middle-aged women

Differential Diagnosis and Pitfalls


Prognosis and Treatment
៉ Metastases from other pediatric malignancies: neuroblastoma,
៉ Unpredictable clinical course
lymphoblastic lymphoma, Wilms tumor, rhabdomyosarcoma
៉ Resection generally the treatment of choice
៉ Other primary liver neoplasms:
៉ HCC: rare in children less than age 5 years; no foci of
extramedullary hematopoiesis
៉ Undifferentiated (embryonal) sarcoma: no epithelial
component; cytokeratin-negative

Ch008-F06731.indd 242 10/26/2006 10:30:19 AM


CHAPTER 8 Liver 243

PATHOLOGIC FEATURES

EHE presents are multiple liver nodules, involving both


lobes of the liver in 80% of cases. Nodule sizes vary
from 1 to 12 cm. The tumor cells form single-fi le cords,
which course through the myxohyaline matrix. A diag-
nostic clue to the vascular nature of this neoplasm is
the presence of ‘blister cells’, characterized by cytoplas-
mic vacuoles representing incipient vascular lumina.
These lumina may contain erythrocytes.
FNA shows a paucicellular aspirate with discohesive,
large polygonal-round epithelioid cells, rarely with
gland-like formations (Fig. 8-41). Cells have large nuclei,
often with vesicular chromatin and prominent nucleoli,
and granular-pale, abundant eosinophilic cytoplasm.
Also observed are intracytoplasmic lumina/vacuoles as
well as intracytoplasmic hemosiderin pigment.
FIGURE 8-41
Epithelioid hemangioendothelioma. Epithelioid cells with round to oval
ANCILLARY STUDIES uniform nuclei resembling a non-small cell carcinoma. An intranuclear
inclusion is also seen. Diagnosis is dependent on immunostaining. Diff
Quik stain, high power.
Electron microscopy may be used to identify Weibel-
Palade bodies, which are the components of endothelial DIFFERENTIAL DIAGNOSIS AND PITFALLS
cells.
EHE reacts with endothelial markers (CD34, CD31,
Factor VIII, Ulex europaeus). Up to 15% of EHE react Vacuoles in blister cells may be mistaken for signet-ring
with cytokeratin stains, which should not be misinter- cells. Unlike signet-ring cells, the blister cells do not
preted as evidence of carcinoma. react with mucicarmine. EHE should also be differenti-
ated from angiosarcoma.

EPITHELIOID HEMANGIOENDOTHELIOMA – ANGIOSARCOMA


PATHOLOGIC FEATURES

Cytopathologic Findings CLINICAL FEATURES


៉ Paucicellular aspirate
៉ Discohesive, large polygonal–round epithelioid cells, rarely
gland-like formations Although rare as such, angiosarcoma is considered the
៉ Large nuclei, often vesicular chromatin, prominent nucleoli most common primary hepatic sarcoma. It represents
៉ Granular–pale, abundant eosinophilic cytoplasm,
intracytoplasmic lumina/vacuoles, hemosiderin

ANGIOSARCOMA – DISEASE FACT SHEET


Histopathologic Findings
៉ Gross:
Incidence
៉ Multiple liver nodules (1 to 12 cm)
៉ Microscopic: ៉ Most common primary hepatic sarcoma
៉ Single-file cords of cells ៉ Overall rare (<1% of primary hepatic neoplasms)
៉ Myxohyaline matrix
៉ ’Blister cells’: cytoplasmic vacuoles with erythrocytes Gender and Age Distribution
(incipient vascular lumina) ៉ Male to female ratio is 3 : 1
៉ Age 60 to 70 years
Ancillary Studies
៉ Endothelial markers: CD34, CD31, Factor VIII, Ulex europaeus Clinical Features
៉ Cytokeratin reactivity in up to 15% of cases ៉ Associated with cirrhosis in 30% of cases
៉ Electron microscopy: Weibel-Palade bodies
Prognosis and Treatment
Differential Diagnosis and Pitfalls ៉ Prognosis extremely poor, most patients dying of disease within 6
៉ Angiosarcoma months of diagnosis
៉ Signet-ring cell carcinoma ៉ Recommended treatment is surgical resection and radiation

Ch008-F06731.indd 243 10/26/2006 10:30:20 AM


244 FINE NEEDLE ASPIRATION CYTOLOGY

less than 1% of primary hepatic neoplasms. Most


patients are in their sixth or seventh decade of life,
with a male to female ratio of 3 : 1. Similar to HCC,
angiosarcoma is associated with cirrhosis (30%). Other
etiologic factors include exposure to vinyl chloride,
Thorotrast, and arsenic. Most patients present with
hepatomegaly, abdominal pain, ascites, and, uncom-
monly, acute abdomen due to spontaneous rupture of
the tumor. Most cases are treated with surgery and
radiation. Prognosis is extremely poor, with most
patients dying of disease within 6 months.
CT scans show solitary or multiple low-density
masses with lobular or irregular outlines, clear or ill-
defined margins, and curvilinear calcification. Post-
contrast CT scans display progressive and isodense
enhancement.

FIGURE 8-42
Angiosarcoma. Spindled cells with long tapering nuclei densely arranged
PATHOLOGIC FEATURES in a tissue fragment. Diagnosis usually hinges upon immunostaining
with vascular markers. Diff Quik stain, high power.

As in other organs, angiosarcoma is characterized by


formation of anastomosing vascular channels lined by
pleomorphic epithelioid or spindled endothelial cells. ANCILLARY STUDIES
‘Hobnail’ morphology is common. The tumor grows
along the sinusoids.
Cytopathologic characteristics include hypercellular Electron microscopy may be used to identify Weibel-
smears with variably sized fragments and numerous Palade bodies. Endothelial markers (CD34, CD31,
single cells with prominent pleomorphism (Fig. 8-42). Factor VIII, and Ulex europaeus) may be used to confirm
The malignant cells are predominantly spindly/fusiform the vascular origin of this neoplasm. Lack of expression
and less often epithelioid in shape. Necrosis is often of smooth muscle markers (actin, desmin, calponin)
observed. may be used to rule out a metastatic leiomyosarcoma.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

The differential diagnosis includes epithelioid heman-


ANGIOSARCOMA – PATHOLOGIC FEATURES gioendothelioma and metastatic sarcomas, particularly
leiomyosarcoma.
Cytopathologic Findings
៉ Hypercellular smears, variably sized fragments, numerous single
cells
៉ Pleomorphism, with predominantly spindly cells, less often
PRIMARY HEPATIC LYMPHOMA
epithelioid cells
៉ Necrosis

CLINICAL FEATURES
Histopathologic Findings
៉ Anastomosing vascular channels
៉ Channels lined by pleomorphic ‘hobnail’ epithelioid or spindled Primary hepatic lymphoma is defined as lymphoma
endothelial cells predominantly involving the liver in the absence of
៉ Growth along the sinusoids
nodal or disseminated disease. Primary hepatic lym-
phoma is exceedingly rare, representing less than
Ancillary Studies
0.5% of extranodal lymphomas. Only approximately
៉ Positive for endothelial markers (CD34, CD31, Factor VIII, and
100 cases have been reported in the literature.
Ulex europaeus)
More commonly, the liver is involved by lymphoma-
៉ Electron microscopy: Weibel-Palade bodies
tous infiltrate as part of disseminated lymphoma or
Differential Diagnosis and Pitfalls
hepatosplenic T-cell lymphoma. Liver involvement is
present at diagnosis in up to 10% of patients with
៉ Epithelioid hemangioendothelioma
៉ Metastatic sarcoma (particularly leiomyosarcoma)
Hodgkin lymphoma and up to 40% of patients with
non-Hodgkin lymphoma. There is a wide age spectrum,
ranging from 7 to 87 years, with a median age of 55

Ch008-F06731.indd 244 10/26/2006 10:30:20 AM


CHAPTER 8 Liver 245

lymphoma, mantle cell lymphoma, lymphoplasmacytic


PRIMARY HEPATIC LYMPHOMA – CLINICAL FEATURES lymphoma, MALT lymphoma, and angiocentric lym-
phoma. Histologic and cytologic features of primary
Incidence tumors correspond to the patterns characteristic of the
៉ Exceedingly rare nodal disease.
៉ Approximately 100 cases reported in the literature FNA shows hypercellular smears with a distinct
population of monotonous-appearing lymphocytes (Figs
Gender and Age Distribution 8-43–8-45). Mitoses and karyorrhexis are often observed,
៉ Male to female ratio is 3 : 1 reflecting the high-grade phenotype of the lymphoma
៉ Wide age spectrum (7 to 87 years; median age, 55 years) process. In cases of Hodgkin lymphoma, the smears are
often not as cellular, and depending on the subtype of
Treatment the disease, Reed-Sternberg cells are observed in varying
៉ Non-surgical numbers. The differential diagnosis involves small
round blue cell tumors (metastatic islet cell tumor in
particular) and, in rare instances, nodular form of extra-
medullary hematopoiesis (Figs 8-46 & 8-47).
years. The male to female ratio is 3 : 1. Therapy is non-
surgical. The chemotherapeutic regimen depends on
lymphoma type.

PATHOLOGIC FEATURES

Primary hepatic lymphoma may present as either a


solitary or multiple liver masses. All described cases
are of a non-Hodgkin lymphoma variety. The vast
majority of primary hepatic lymphomas are of B-cell
origin, with over 75% representing the diffuse large
B-cell lymphoma. Other patterns of primary B-cell lym-
phoma include follicular lymphoma, small lymphocytic

PRIMARY HEPATIC LYMPHOMA – PATHOLOGIC FEATURES FIGURE 8-43


Primary hepatic lymphoma. Monotonous population of small lympho-
Cytopathologic Findings cytes. Immunostaining and flow analysis was consistent with a MALT-
type lymphoma. Papanicolaou stain, high power.
៉ Depends on the type of lymphoma; similar to nodal disease

Histopathologic Findings
៉ Gross:
៉ Solitary or multiple masses
៉ Microscopic:
៉ All cases non-Hodgkin lymphoma
៉ Vast majority B-cell lymphoma
៉ 75% diffuse large B-cell lymphoma

Ancillary Studies
៉ Pan-hematopoietic marker, LCA (CD45), distinguishes lymphoma
from carcinoma, melanoma, or sarcoma
៉ Lymphomas further subtyped with CD3 (T-cell marker), CD20
(B-cell marker) and various other hematopoietic markers
៉ Fresh specimen should be submitted for flow cytometry

Differential Diagnosis and Pitfalls


៉ Poorly differentiated carcinoma, melanoma, and sarcoma
៉ Hepatic involvement by disseminated lymphoma: most common
type of hepatic lymphoma; liver involved in 40% of patients FIGURE 8-44
with non-Hodgkin lymphoma Primary hepatic lymphoma. High-grade tumor shows medium-sized lym-
៉ Hepatic involvement by hepatosplenic T-cell lymphoma phocytes with abundant mitoses and fine cytoplasmic vacuoles consis-
tent with an aggressive Burkitt-type lymphoma. Diff Quik stain, high
power.

Ch008-F06731.indd 245 10/26/2006 10:30:21 AM


246 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 8-45 FIGURE 8-47


Plasmacytoma. Malignant plasma cells with the characteristic speckled Extramedullary hematopoiesis. Megakaryocyte in a background of mono-
chromatin and intranuclear inclusions. Papanicolaou stain, high power. nuclear cells of different lineages. Papanicolaou stain, high power.

this differential diagnosis. Primary hepatic lymphoma


should also be differentiated from hepatic involvement
by disseminated lymphoma and hepatosplenic T-cell
lymphoma.

METASTATIC TUMORS TO THE LIVER

CLINICAL FEATURES (Table 8-15)

Liver, along with the lung, is the organ that is most


frequently involved by metastatic tumors. In the United
States, metastases represent 98% of all hepatic malig-
nancies. However, in patients with cirrhosis, primary
liver malignancies (mainly HCC) are three times more
FIGURE 8-46
common than metastases.
Extramedullary hematopoiesis. Polymorphous cells with lymphoid and
erythroid elements. Diff Quik stain, high power.
In adults, malignancies that most frequently metas-
tasize to the liver include those from the colorectum,
pancreas, breast, lung, stomach, kidney, and skin
ANCILLARY STUDIES

Leukocyte common antigen, LCA (CD45), is a pan- METASTATIC TUMORS – DISEASE FACT SHEET
hematopoietic marker used to distinguish lymphoma
from carcinoma, melanoma, or sarcoma. Lymphomas Incidence
can be further subtyped with CD3 (T-cell marker), ៉ Most common type of hepatic malignancy
៉ Metastases represent 98% of hepatic malignancies (in the United
CD20 (B-cell marker), and various other hematopoietic
States)
antigens. If lymphoma is suspected, a fresh specimen
៉ In cirrhotic livers, primary neoplasms are three times more
should be submitted for a flow cytometric analysis. common than metastases

Gender and Age Distribution


DIFFERENTIAL DIAGNOSIS AND PITFALLS ៉ Depends on tumor type

Prognosis and Treatment


Lymphoma should be differentiated from poorly ៉ Depends on tumor type
differentiated carcinoma, melanoma, and sarcoma.
Immunohistochemical stains and flow cytometry aid in

Ch008-F06731.indd 246 10/26/2006 10:30:21 AM


CHAPTER 8 Liver 247

TABLE 8-15
Metastatic Tumors in the Liver – Common Facts

• Most common cancer in the liver


• Most common indication for hepatic FNA
• Most common – colon, pancreas, breast, lung, kidney,
melanoma
• Source of the primary tumor could be occult
• Cytomorphology alone may not be able to diagnose an
unknown primary (immunoperoxidase stains could be
helpful)

(melanoma). Leiomyosarcoma is the most common soft


tissue tumor to metastasize to the liver. In contrast, it
is unusual for prostate tumors to metastasize to the
liver. In children, neoplasms that commonly metastasize FIGURE 8-48
to the liver include neuroblastoma, Wilms tumor, and Hepatectomy specimen shows a subcapsular nodule of metastatic colonic
adenocarcinoma. This presents the most common scenario for a liver FNA
rhabdomyosarcoma. procedure, which provides an accurate answer within a short time.

PATHOLOGIC FEATURES

Metastatic tumors frequently present as multiple


masses. Histopathology and cytopathology depend on
the tumor of origin (Figs 8-48–8-58). Metastatic
melanoma is a frequently observed secondary tumor in
the liver and may depict significant morphologic overlap
with poorly differentiated HCC. This necessitates a
careful evaluation of subtle morphologic differences
between the two tumor types (Table 8-16).

METASTATIC TUMORS – PATHOLOGIC FEATURES

Cytopathologic Findings FIGURE 8-49


៉ Depends on the type of primary tumor Hepatic metastasis: colonic adenocarcinoma. Columnar-shaped cells in
a background of ‘dirty necrosis’. Papanicolaou stain, high power.
Histopathologic Findings
៉ Gross: usually present as multiple nodules
៉ Microscopic: depends of the tumor of origin

Ancillary Studies
៉ HepPar-1 to distinguish from primary HCC or hepatoblastoma
៉ CK7 and CK20
៉ Organ- and tissue-specific markers: ER/PR (breast, ovary),
TTF-1 (lung, thyroid), PSA (prostate), S-100/Melan A/HMB-45
(melanoma), actin/desmin (leiomyosarcoma)

Differential Diagnosis and Pitfalls


៉ Common metastatic malignancies in adults: colorectum,
pancreas, breast, lung, stomach, kidney, melanoma,
leiomyosarcoma
៉ Common metastatic malignancies in children: neuroblastoma,
Wilms tumor, rhabdomyosarcoma
៉ Primary hepatic neoplasms: hepatocellular carcinoma,
cholangiocarcinoma, and mesenchymal neoplasms are in the FIGURE 8-50
differential diagnosis for metastatic malignancies Hepatic metastasis: adenosquamous carcinoma from a pancreatic
primary. Note admixture of adenocarcinoma with brightly pink keratin-
ized squamous cells. Papanicolaou stain, high power.

Ch008-F06731.indd 247 10/26/2006 10:30:23 AM


248 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 8-52
FIGURE 8-51
Hepatic metastasis: malignant melanoma from a cutaneous primary.
Hepatic metastasis: renal cell carcinoma. Clear cell tumor with large Discohesive, heavily pigmented malignant cells with an epithelioid
nuclei and macronucleoli. Renal cell carcinoma shares several morpho- appearance. Note prominent nucleoli and focal resemblance to hepato-
logic features with hepatocellular carcinoma and usually requires immu- cytes. Papanicolaou stain, high power.
nostaining for definitive diagnosis. Papanicolaou stain, high power.

FIGURE 8-53 FIGURE 8-54


Hepatic metastasis of malignant melanoma. Large pleomorphic and Hepatic metastasis: squamous cell carcinoma from an esophageal
naked nuclei resembling poorly differentiated hepatocellular carcinoma. primary. Extensive cystic necrosis (left side) with densely keratinized
Macronucleoli are visible. Note lack of pigmented cells. Diff Quik stain, malignant cells (right side). Papanicolaou stain, low to high power.
high power.

TABLE 8-16
Hepatocellular Carcinoma (HCC) versus Metastatic
Malignant Melanoma (MM)

• Closest mimic of HCC


• Liver is a very common site for visceral spread of MM
• Cytomorphologic features (MM over HCC):
• More often single cells or sheet-like arrangement
• Plasmacytoid cells (eccentric nuclei)
• More frequent binucleation
• Cytoplasmic tails/prolongations
• Melanin-like pigment
• Necrosis

FIGURE 8-55
Hepatic metastasis: small cell carcinoma from a lung primary. Small,
hyperchromatic nuclei with prominent molding, nuclear ‘streak’ artifact,
and lack of nucleoli. Diff Quik stain, high power.

Ch008-F06731.indd 248 10/26/2006 10:30:24 AM


CHAPTER 8 Liver 249

FIGURE 8-56
Hepatic metastasis: islet cell tumor (pancreatic endocrine neoplasm).
Monotonous population of neoplastic cells with eccentric nuclei (‘plas- FIGURE 8-58
macytoid’ appearance) and speckled chromatin. Uniformity of cells with
Hepatic metastasis: gastrointestinal stromal tumor from a gastric
a relatively smaller size, lack of prominent nucleoli, and glandular for-
primary. Loose fragment of plump fusiform nuclei arranged in a hap-
mations differentiate this from an adenocarcinoma. Papanicolaou stain,
hazard fashion. Papanicolaou stain, intermediate power.
intermediate power.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

Metastatic neoplasms must be differentiated from


primary HCC, cholangiocarcinoma, and mesenchymal
neoplasms.

SUGGESTED READINGS
Nodular Regenerative Hyperplasia
Cohen MB, Haber MM, Holly EA, Ahn DK, Bottles K, Stoloff AC. Cytologic
criteria to distinguish hepatocellular carcinoma from nonneoplastic
liver. Am J Clin Pathol 1991;95:125–130.
Yang GC, Yang GY, Tao LC. Distinguishing well-differentiated hepatocel-
lular carcinoma from benign liver by the physical features of fine-needle
aspirates. Mod Pathol 2004;17:798–802.
FIGURE 8-57
Hepatic metastasis: hemangiopericytoma from a brain primary. Monoto- Focal Nodular Hyperplasia
nous, round to oval cells with mostly naked nuclei and scant to no
cytoplasm. Diff Quik stain, intermediate power. Kong CS, Appenzeller M, Ferrell LD. Utility of CD34 reactivity in evaluating
focal nodular hepatocellular lesions sampled by fine needle aspiration
biopsy. Acta Cytol 2000;44:218–222.
Ruschenburg I, Droese M. Fine needle aspiration cytology of focal nodular
hyperplasia of the liver. Acta Cytol 1989;33:857–860.
ANCILLARY STUDIES
Hydatid Disease
Various markers may be applied to determine the site Agarwal PK, Husain N, Singh BN. Cytologic findings in aspirated hydatid
of origin of a metastatic carcinoma. For example: ER fluid. Acta Cytol 1989;33:652–654.
and PR support breast or gynecologic tract primary; Parwani AV, Burroughs FH, Ali SZ. Echinococcal cyst of the liver. Diagn
TTF-1, lung or thyroid primary; CDX-2, colorectal Cytopathol 2004;31:111–112.
primary; and PSA or PSAP, prostate primary. Meta-
static melanoma reacts with S-100, Melan A, and HMB- Extramedullary Hematopoiesis
45. Metastatic leiomyosarcoma expresses actin, desmin,
and calponin. In contrast, primary HCC may be distin- Lemos LB, Baliga M, Benghuzzi HA, Cason Z. Nodular hematopoiesis of
the liver diagnosed by fine-needle aspiration cytology. Diagn Cytopa-
guished from metastatic carcinomas based on expres- thol 1997;16:51–54.
sion of HepPar-1, AFP, and canalicular polyclonal CEA Parwani AV, Ali SZ. Pathologic quiz case: a 52-year-old woman with a liver
and CD10. mass. Arch Pathol Lab Med 2003;127:631–632.

Ch008-F06731.indd 249 10/26/2006 10:30:25 AM


250 FINE NEEDLE ASPIRATION CYTOLOGY

Hemangioma Fibrolamellar Hepatocellular Carcinoma


Guy CD, Yuan S, Ballo MS. Spindle-cell lesions of the liver: diagnosis by Davenport RD. Cytologic diagnosis of fibrolamellar carcinoma of the liver
fine-needle aspiration biopsy. Diagn Cytopathol 2001;25:94–100. by fine-needle aspiration. Diagn Cytopathol 1990;6:275–279.
Layfield LJ, Mooney EE, Dodd LG. Not by blood alone: diagnosis of hem- Kunz G Jr, Chung J, Ali SZ. Hepatocellular carcinoma-fibrolamellar variant:
angiomas by fine-needle aspiration. Diagn Cytopathol 1998;19: cytopathology of an unusual case. Diagn Cytopathol 2002;26:257–
250–254. 261.

Hepatocellular Adenoma Intrahepatic Cholangiocarcinoma

Pedio G, Landolt U, Zobeli L, Gut D. Fine needle aspiration of the liver. Dalton-Clarke HJ, Pearse E, Krause T, McPherson GA, Benjamin IS, Blumgart
Significance of hepatocytic naked nuclei in the diagnosis of hepatocel- LH. Fine needle aspiration cytology and exfoliative biliary cytology in
lular carcinoma. Acta Cytol 1988;32:437–442. the diagnosis of hilar cholangiocarcinoma. Eur J Surg Oncol 1986;12:
Suen KC. Diagnosis of primary hepatic neoplasms by fine-needle aspiration 143–145.
cytology. Diagn Cytopathol 1986;2:99–109. Pitman MB. Fine needle aspiration biopsy of the liver. Principal diagnostic
challenges. Clin Lab Med 1998;18:483–506, vi.

Ciliated Hepatic Foregut Cyst


Hepatoblastoma
Hornstein A, Batts KP, Linz LJ, Chang CD, Galvanek EG, Bardawil RG. Fine
needle aspiration diagnosis of ciliated hepatic foregut cysts: a report Wakely PE Jr, Silverman JF, Geisinger KR, Frable WJ. Fine needle aspiration
of three cases. Acta Cytol 1996;40:576–580. biopsy cytology of hepatoblastoma. Mod Pathol 1990;3:688–693.
Zaman SS, Langer JE, Gupta PK. Ciliated hepatic foregut cyst. Report of Weir EG, Ali SZ. Hepatoblastoma: cytomorphologic characteristics in
a case with findings on fine needle aspiration. Acta Cytol 1995;39: serious cavity fluids. Cancer 2002;96:267–274.
781–784.
Primary Malignant Mesenchymal Tumors
Bile Duct Adenoma Smith MB, Silverman JF, Raab SS, Towell BD, Geisinger KR. Fine-needle
aspiration cytology of hepatic leiomyosarcoma. Diagn Cytopathol 1994;
Bhathal PS, Hughes NR, Goodman ZD. The so-called bile duct adenoma is 11:321–327.
a peribiliary gland hamartoma. Am J Surg Pathol 1996;20:858–864. Wee A, Nilsson B. Fine needle aspiration biopsy of hepatic leiomyosar-
Shibahara H, Tamada S, Goto M, et al. Pathologic features of mucin- coma. An unusual epithelioid variant posing a potential diagnostic
producing bile duct tumors: two histopathologic categories as counter- pitfall in a hepatocellular carcinoma-prevalent population. Acta Cytol
parts of pancreatic intraductal papillary-mucinous neoplasms. Am J 1997;41:737–743.
Surg Pathol 2004;28:327–338.

Epithelioid Hemangioendothelioma
Hepatobiliary Cystadenoma
Pettinato G. Fine-needle aspiration cytology of epithelioid hemangio-
Devaney K, Goodman ZD, Ishak KG. Hepatobiliary cystadenoma and cyst- endothelioma. Diagn Cytopathol 1990;6:438–439.
adenocarcinoma. A light microscopic and immunohistochemical study Soslow RA, Yin P, Steinberg CR, Yang GC. Cytopathologic features of
of 70 patients. Am J Surg Pathol 1994;18:1078–1091. hepatic epithelioid hemangioendothelioma. Diagn Cytopathol 1997;17:
Logrono R, Rampy BA, Adegboyega PA. Fine needle aspiration cytology 50–53.
of hepatobiliary cystadenoma with mesenchymal stroma. Cancer 2002;
96:37–42
Angiosarcoma
Boucher LD, Swanson PE, Stanley MW, Silverman JF, Raab SS, Geisinger
Infl ammatory Pseudotumor
KR. Cytology of angiosarcoma. Findings in fourteen fine-needle aspira-
Hosler GA, Steinberg DM, Sheth S, Hamper UM, Erozan YS, Ali SZ. Inflam- tion biopsy specimens and one pleural fluid specimen. Am J Clin Pathol
matory pseudotumor: a diagnostic dilemma in cytopathology. Diagn 2000;114:210–219.
Cytopathol 2004;31:267–270. Nguyen GK, Husain M. Fine-needle aspiration biopsy cytology of angio-
Malhotra V, Gondal R, Tatke M, Sarin SK. Fine needle aspiration cytologic sarcoma. Diagn Cytopathol 2000;23:143–145.
appearance of inflammatory pseudotumor of the liver. A case report.
Acta Cytol 1997;41:1325–1328.
Primary Hepatic Lymphomas
Collins KA, Geisinger KR, Raab SS, Silverman JF. Fine needle aspiration
Hepatocellular Carcinoma biopsy of hepatic lymphomas: cytomorphology and ancillary studies.
Acta Cytol 1996;40:257–262.
Kulesza P, Torbenson M, Sheth S, Erozan YS, Ali SZ. Cytopathologic grading
Rappaport KM, DiGiuseppe JA, Busseniers AE. Primary hepatic lymphoma:
of hepatocellular carcinoma on fine-needle aspiration. Cancer 2004;102:
report of two cases diagnosed by fine-needle aspiration. Diagn Cytopa-
247–258.
thol 1995;13:142–145.
Soyuer I, Ekinci C, Kaya M, Genc Y, Bahar K. Diagnosis of hepatocellular
carcinoma by fine needle aspiration cytology. Cellular features. Acta
Cytol 2003;47:581–589. Metastatic Tumors
Takenaka A, Kaji I, Kasugai H, et al. Usefulness of diagnostic criteria for
aspiration cytology of hepatocellular carcinoma. Acta Cytol 1999;43: Bizjak-Schwarzbartl M. Fine-needle aspiration biopsy in the diagnosis of
610–616. metastases in the liver. Diagn Cytopathol 1987;3:278–283.
Wee A, Nilsson B, Tan LK, Yap I. Fine needle aspiration biopsy of hepa- Parwani AV, Chan TY, Mathew S, Ali SZ. Metastatic malignant melanoma
tocellular carcinoma. Diagnostic dilemma at the ends of the spectrum. in liver aspirate: cytomorphologic distinction from hepatocellular car-
Acta Cytol 1994;38:347–354. cinoma. Diagn Cytopathol 2004;30:247–250.

Ch008-F06731.indd 250 10/26/2006 10:30:27 AM


9 Pancreas
Martha Bishop Pitman

serous cyst, and solid neoplasms with cystic degenera-


INTRODUCTION tion. Cysts with thick septa and significant internal
debris are suspicious for malignancy, and the presence
of a solid mass in the wall of a cyst in suspicious for an
Cytologic specimens are obtained from the pancreas invasive cystic neoplasm.
most commonly by fine needle aspiration biopsy
(FNAB). The aspirated tissue from any lesion can be
prepared in a number of different ways: direct smears,
cytospins (Thermo Shandon Instruments), liquid-
based preparations (ThinPrep® [Cytyc Corporation, NORMAL PANCREAS
Marlborough, MA] or SurePathTM [TriPath, Inc, Burl-
ington, NC]) and cell blocks. Direct smears are the most
common, particularly for solid masses, but specimens A significant pitfall in the interpretation of pancreatic
aspirated as a fluid from a cyst are often processed as cytology is the over-interpretation of normal pancreatic
cytospins or liquid-based specimens. Liquid-based epithelium as neoplastic. The pancreas is composed of
preparations eliminate obscuring blood and inflam- exocrine (acinar cells and ducts) and endocrine (islets
mation and provide excellent cellular preservation; of Langerhans) cells. Eighty-five percent of the pan-
however, cellular and extracellular material such as creas is exocrine tissue, predominantly acinar epithe-
mucin is attenuated and not as apparent as on direct lium. In benign processes, except in late chronic
smears. Preserving some fluid in the liquid form from pancreatitis, acinar cells dominate aspiration smears.
cyst aspirates allows for additional slides dedicated to Acinar epithelium presents as cohesive, small, grape-
special stains for mucin, which may assist in detecting like clusters of cells and scattered polygonal single cells
subtle background or intracellular mucin. Aspirates with occasional stripped nuclei (Fig. 9-1). The round,
rinsed in saline can be used for flow cytometry analysis regular nuclei are central to eccentric, with uniform
in cases suspicious of lymphoma. Cell block prepara- chromatin and often quite prominent nucleoli. The
tions are advantageous given the readily available tissue typically abundant granular cytoplasm stains blue–
for immunocytochemistry. green with the Papanicolaou stain and purple with a
Percutaneous FNAB with computed tomography Romanowsky stain (Fig. 9-2). The Romanowsky stain
(CT) guidance is most commonly used; however, endo- highlights scattered small vacuoles in the cytoplasm.
scopic ultrasound (EUS)-guided FNAB is becoming The cytoplasmic granules may not be very visible. The
increasingly utilized. It is important to recognize that architectural arrangement of the cells is key to differ-
EUS-guided biopsies traverse the stomach or duodenal entiating benign acinar cells from a neoplastic acinar
wall, thereby introducing the potential for epithelial and cell proliferation, the latter generally forming large
mucin contamination. sheets and clusters as apposed to the small, uniform,
The interpretation of pancreatic cytology should not grape-like clusters of a benign process (see acinar cell
occur in a vacuum, and all clinical, radiologic, and ancil- carcinoma).
lary tests need to be incorporated in order to provide an Normal pancreatic ducts present as large, flat, cohe-
accurate diagnosis. The differential diagnosis of pancre- sive sheets of epithelium with round, uniform, evenly
atic masses is initially established from the radiographic spaced nuclei, yielding the classic glandular ‘honey-
appearance of the mass, and generally falls into two combed’ appearance (Fig. 9-3). The cytoplasm of normal
categories: solid versus cystic mass lesions. Solid masses ductal cells is not visibly mucinous and can best be seen
are much more common than cystic masses, and adeno- when the epithelium is present in strips yielding a
carcinoma is by far the most common neoplasm in the ‘picket fence’ arrangement, or in sheets with a luminal
pancreas. Solid mass lesions in the differential diagnosis edge (Fig. 9-4). Ductal cells have round to oval nuclei,
include chronic pancreatitis, pancreatic endocrine neo- even chromatin, and generally small, inconspicuous
plasm, acinar cell carcinoma, pancreatoblastoma, and nucleoli. Features consistent with a non-neoplastic
metastatic neoplasms. The most common cyst in the process on cytology include cohesion and nuclei with
pancreas is a pseudocyst. This non-neoplastic cyst needs uniform size, shape, chromatin distribution, and spacing.
to be distinguished from a neoplastic mucinous cyst, Distinguishing benign pancreatic ductal epithelium
251

Ch009-F06731.indd 251 10/26/2006 10:31:42 AM


252 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-1
Normal pancreatic acinar epithe-
lium. The smear pattern of normal
acinar epithelium is one in which
the majority of acinar epithelial
cells maintain a cohesive small
grape-like clustering. Occasional
stripped nuclei can be identified in
the background. Papanicolaou stain,
low power.

A B

FIGURE 9-2
Benign pancreatic acinar epithelium. Acinar cells are uniform with round, regular, central to slightly eccentric nuclei and often quite prominent
nucleoli. The cytoplasm is granular, staining blue–green with the Papanicolaou stain (A) and purple with the Romanowsky stain, which also high-
lights scattered small cytoplasmic vacuoles (B). A, Papanicolaou stain, high power. B, Romanowsky stain, high power.

Ch009-F06731.indd 252 10/26/2006 10:31:43 AM


CHAPTER 9 Pancreas 253

FIGURE 9-3
Benign pancreatic ductal epithe-
lium. Benign pancreatic ducts often
present as fl at cohesive sheets
with round, uniform, evenly spaced
nuclei, yielding the classic glandu-
lar honeycombed appearance. Papa-
nicolaou stain, high power.

Cytologically, individual islet cells appear uniform and


polygonal with round nuclei, coarse clumped chroma-
tin, small inconspicuous nucleoli, and pale amphophilic
cytoplasm.

NON-NEOPLASTIC MASS LESIONS

CHRONIC PANCREATITIS

Chronic pancreatitis is a progressive inflammatory


disease of the pancreas that destroys the exocrine com-
ponent – and, in some severe cases, the endocrine com-
ponent – of the gland, leading to irreversible morphologic
changes and function.
FIGURE 9-4
Benign pancreatic ductal epithelium. Occasionally, ductal epithelial
cells can be seen in strips on edge demonstrating basal nuclei and apical
columnar cytoplasm. Note that the cytoplasm is non-mucinous. Papa- CLINICAL FEATURES
nicolaou stain, high power.

In the United States, chronic alcohol abuse accounts for


from gastrointestinal epithelium, especially gastric roughly 70% of all cases of chronic pancreatitis. Other
epithelium, may be impossible. The misinterpretation causes of chronic pancreatitis include obstruction
of gastrointestinal epithelium as benign ductal epithe- (stones, abnormal anatomy, scarring, extrinsic com-
lium aspirated from a solid mass can lead to a false pression), autoimmune disease (lymphoplasmacytic
negative biopsy that, in reality, has not sampled the sclerosing pancreatitis), chronic malnutrition, and
mass at all. heredity. The condition is more common in men than
Benign islet cells are not often appreciated on in women, and occurs over a wide age range, generally
aspirate smears of a non-neuroendocrine neoplasm. 40–60 years of age.

Ch009-F06731.indd 253 10/26/2006 10:31:44 AM


254 FINE NEEDLE ASPIRATION CYTOLOGY

CHRONIC PANCREATITIS – DISEASE FACT SHEET CHRONIC PANCREATITIS – PATHOLOGIC FEATURES

Incidence Cytopathologic Findings


៉ 70% associated with long-term alcohol abuse ៉ Mixed ductal and acinar epithelium
៉ Infl ammation in the background and embedded in stromal
Gender and Age Distribution fragments
៉ M > F ៉ Cellular stromal fragments with infl ammatory cells >30/60× HPF
៉ 40–60 years is associated with autoimmune etiology
៉ Fat necrosis and calcific debris may be present
៉ Ductal epithelium contains relatively evenly spaced, uniform,
Radiologic Features
polarized nuclei with even to slightly coarse chromatin and
៉ Irregular ductal dilation associated with strictures and
smooth nuclear membranes; nucleoli may be prominent
calcifications ៉ Cytoplasm is usually non-mucinous (may see mucinous
៉ Focal areas of expansive fibrosis produce a mass-like lesion
metaplasia in pancreatic intraepithelial neoplasia)
simulating carcinoma
Differential Diagnosis and Pitfalls
Prognosis and Treatment
៉ Ductal adenocarinoma
៉ A benign disease, but the variable loss of exocrine and endocrine
៉ Gastrointestinal contamination (pitfall)
function is irreversible and the risk of developing carcinoma is
increased
៉ Therapy includes enzyme or hormone supplementation

necrosis supports chronic pancreatitis over adenocarci-


The morphologic changes of the gland, e.g. loss of noma (Fig. 9-7). Ductal cells are almost always present,
exocrine or endocrine tissue and fibrosis, are irrevers- although they may be quite scant, and can show some
ible. The risk of developing carcinoma is increased, nuclear crowding and coarse chromatin. The benign
particularly with hereditary chronic pancreatitis. The ‘honeycombed’ pattern of nuclear spacing is generally
therapy for exocrine or endocrine dysfunction is enzyme maintained and chromatin is evenly distributed in the
or hormone supplementation. nucleus, although it may be somewhat coarse and
display conspicuous nucleoli (Fig. 9-8).
Patients with classic clinical and radiologic evidence
of chronic pancreatitis, however, rarely undergo FNAB
R ADIOLOGIC FEATURES for confirmation of the diagnosis. It is the expansive
sclerosis in chronic pancreatitis, especially in auto-
immune pancreatitis, and focal gland involvement by
Most cases of chronic pancreatitis are associated with chronic pancreatitis that results in a mass lesion often
non-focal lesions, irregular ductal dilation that is associated with irregular pancreatic duct contours and
often associated with stricture formation, obstruction, bile duct stricture, radiologic features highly suspicious
and calcifications. Pseudocyst formation may also be for adenocarcinoma. The dominance of ductal cells
present. Focal involvement of the gland may occur, as without acinar cells or calcific debris is the cytologic
well as dense expanses of fibrosis in more diffuse finding that raises the differential diagnosis of
disease, both conditions creating mass-like lesions that adenocarcinoma.
mimic pancreatic carcinoma.

ANCILLARY STUDIES
CYTOPATHOLOGIC FEATURES
Currently there is no reliable ancillary study that dis-
The classic cytologic findings in patients with clinical tinguishes between benign and malignant pancreatic
and endoscopic evidence of the more common alcohol- glandular epithelium. In early studies, immunocyto-
related disease include the presence of fibrotic acinar chemical staining using B72.3 was shown not to stain
tissue, fragments of relatively acellular fibrosis, scant benign pancreatic ductal epithelium, while demonstrat-
reactive ductal epithelium, and, occasionally, saponi- ing fine perinuclear punctate staining in gastric and
fied fat necrosis and/or chalky to granular calcific duodenal epithelium, and strong cytoplasmic staining
debris (Fig. 9-5). Mixed inflammation and histiocytes in malignant pancreatic ductal epithelium; however,
may also be present in the background. Cellular stromal not all carcinomas label with B72.3, so a negative stain
fragments populated with inflammatory cells (>30 per does not exclude carcinoma. Evaluation of the patient
60×) (Fig. 9-6) is strongly suggestive of autoimmune or pancreatic tissue for mutation of the K-ras gene, a
pancreatitis – a condition important to recognize, as proto-oncogene on chromosome 12p12, remains contro-
patients can be treated with steroids rather than versial. Although most carcinomas tend to harbor a
surgery. Fat necrosis in contrast to coagulative cellular K-ras gene mutation, some cases of chronic pancreatitis

Ch009-F06731.indd 254 10/26/2006 10:31:45 AM


CHAPTER 9 Pancreas 255

FIGURE 9-5
Chronic pancreatitis. A mixture of
fibrotic acinar tissue, acellular frag-
ments of fibrous tissue, scant ductal
epithelium, reactive acinar epithe-
lium, infl ammation, saponified fat
necrosis, and chalky granular cal-
cific debris are characteristic
features of chronic pancreatitis.
Papanicolaou stain, low power.

FIGURE 9-6
Autoimmune pancreatitis (lympho-
plasmacytic sclerosing pancreati-
tis). The presence of cellular stromal
fragments populated with infl am-
matory cells of at least 30 per 60×
high-power field correlates with an
autoimmune etiology. Papanicolaou
stain, medium power.

Ch009-F06731.indd 255 10/26/2006 10:31:45 AM


256 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 9-7
Fat necrosis compared to coagulative cellular necrosis. Fat necrosis is identified by the presence of foamy histiocytes, infl ammatory cells, and
background cellular debris, and is characteristic of chronic pancreatitis (A). Cellular necrosis of carcinoma is distinguished by the presence of ghost
cells, infl ammation, and typically residual atypical epithelial cells in the background (B). A and B, Papanicolaou stain, low power.

FIGURE 9-8
Reactive ductal epithelium of
chronic pancreatitis. The honey-
combed pattern of nuclei with rela-
tively even spacing is maintained
in reactive ductal groups of chronic
pancreatitis. The nuclei may dem-
onstrate slight chromatin clumping
and nucleoli. Papanicolaou stain,
high power.

and pancreatic intraepithelial neoplasia do as well, and DIFFERENTIAL DIAGNOSIS AND PITFALLS
not all carcinomas will demonstrate the mutation, so
the diagnostic utility of the test is limited. Patients
suspected of having autoimmune pancreatitis should The primary differential diagnosis of chronic pan-
undergo serologic testing for IgG4. Although not ele- creatitis is ductal adenocarcinoma. The distinction
vated in all patients with the condition, an elevated between benign and reactive ductal epithelium in
level is strongly associated with autoimmune pancre- chronic pancreatitis and the malignant ductal epithe-
atitis and supports a cytologic diagnosis. lium of ductal adenocarcinoma can be challenging. The

Ch009-F06731.indd 256 10/26/2006 10:31:46 AM


CHAPTER 9 Pancreas 257

A B

FIGURE 9-9
Reactive ductal epithelium compared to adenocarcinoma. The reactive ductal epithelium of pancreatitis maintains relative even nuclear spacing,
uniform nuclear size and nuclear membranes, mild nuclear enlargement, and variation within a single group (A). In contrast and although sometimes
quite subtle, adenocarcinomas demonstrate more variability in nuclear size within a single group, uneven nuclear spacing, a loss of polarity, irregular
nuclear membranes with notches, divots, and grooves, as well as nuclear membrane thickening (B). A and B, Papanicolaou stain, high power.

overall cellularity of smears in chronic pancreatitis of residual acinar epithelium can present a diagnostic
is relatively low compared to adenocarcinoma, and is pitfall. Cellular smears of reactive and injured acinar
often composed of ductal epithelium with acinar epi- cells with a loss of the normal cohesive grape-like clus-
thelium and inflammation, in contrast to a pure ductal tering, interpreted in the setting of a solid mass lesion,
cell population in adenocarcinoma. If biopsies are being can be misinterpreted as a neoplasm such as acinar cell
performed by EUS, care must be taken not to include carcinoma, solid-pseudopapillary neoplasm, or pancre-
the contaminating epithelium from the gastrointestinal atic endocrine neoplasm (Fig. 9-10). Ductal cells with
tract, which may present in the assessment of overall mucinous metaplasia (PanIN), especially with dysplasia
cellularity, or to misinterpret the gastrointestinal epi- (PanIN-2 or -3), may be aspirated in chronic pancreati-
thelium as benign ductal epithelium of pancreatitis. tis and can cause a false positive interpretation.
The architecture of cell groups in pancreatitis is gen-
erally organized, with flat, monolayered sheets of ductal
cells that display relatively polarized nuclei with minimal
crowding and overlap compared to the subtle loss of PSEUDOCYST
nuclear polarity and nuclear crowding in adenocarci-
noma. Nuclear size is also more uniform and nuclear
membrane abnormalities minimal in chronic pancreati- A pancreatic pseudocyst is a localized collection of
tis, whereas nuclear enlargement and variation of pancreatic secretions, necrotic debris, and blood that,
nuclear size within a single group of cells, and nuclear by definition, has no epithelial lining.
membranes with notches, divots, grooves, and thicken-
ing favors adenocarcinoma (Fig. 9-9). The nuclear chro-
matin pattern of benign and reactive cells in chronic
pancreatitis demonstrates evenly distributed granular CLINICAL FEATURES
chromatin, whereas in adenocarcinoma there is hyper-
chromasia or, in well-differentiated adenocarcinoma,
parachromatin clearing. The cytoplasm of ductal cells in Pseudocysts occur as a consequence of damage to the
chronic pancreatitis does not usually contain mucin pancreatic parenchyma that results in hemorrhage,
visible on routine light microscopy, but may be present necrosis, and autodigestion of pancreatic tissue from
and even abundant in well-differentiated adenocarci- the release and activation of pancreatic enzymes. The
noma, leading to an exaggerated uneven arrangement of incidence of this pancreatic injury is highest in patients
nuclei, a pattern also referred to as ‘drunken honey- with a well-established history of acute pancreatitis,
comb’ (see adenocarcinoma section). the most common etiology of which is alcohol abuse.
Pitfalls in the cytologic diagnosis include misinter- Approximately 10% of patients with acute pancreatitis
preting gastrointestinal epithelial contamination from will develop a pseudocyst.
endoscopic biopsies as originating from the pancreas, The age and gender of patients with pseudocysts
leading to a false adequacy assessment. In addition, aspi- parallels that of pancreatitis. Alcohol-related pseudo-
rates of chronic pancreatitis with significant amounts cysts are more common in middle-aged men, while

Ch009-F06731.indd 257 10/26/2006 10:31:47 AM


258 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-10
Disrupted acinar epithelial cells in
autoimmune pancreatitis. A rela-
tively cellular smear composed of
disrupted acinar epithelial cells
with a loss of the grape-like orga-
nized clustering and reactive atypia
can mimic a neoplasm such as
pancreatic endocrine neoplasm,
solid-pseudopapillary neoplasm,
and acinar cell carcinoma. Papani-
colaou stain, low power.

R ADIOLOGIC FEATURES
PSEUDOCYST – DISEASE FACT SHEET

Incidence Pseudocysts are usually solitary, small to very large (up


៉ Occurs in ∼10% of patients with acute pancreatitis; also to 20 cm), well-demarcated, thin-walled, unilocular
associated with chronic pancreatitis and trauma peripancreatic cysts that can occur anywhere in the
pancreas, but are most common in the pancreatic tail.
Gender and Age Distribution
៉ Age and gender are dependent on etiology
៉ Alcohol-related cysts are more common in middle-aged men
៉ Traumatic, biliary, and hereditary causes of pancreatitis lead to CYTOPATHOLOGIC FEATURES
pseudocysts relatively equally in men and women

Radiologic Features The cyst fluid aspirated from an uncomplicated pseu-


៉ Typically, single, unilocular, thin-walled cysts without septations, docyst is generally thin, brown to green, and non-
occurring anywhere in the pancreas mucinous. A complicated pseudocyst, however, may
produce thick mucoid-appearing fluid due to the pres-
Prognosis and Treatment ence of inflammation. Cytologically, the characteristic
៉ Pseudocysts may resolve spontaneously, but can grow in size features include degenerative cyst debris with acute
៉ Undrained pseudocysts can rupture, erode into vessels, cause and chronic inflammatory cells, histiocytes, hemosid-
obstruction, and get infected erin, and often bile (Fig. 9-11). By definition, there are
៉ Treatment is usually drainage or resection
no cyst-lining epithelial cells.

ANCILLARY STUDIES
pseudocysts secondary to trauma, biliary disease, and
heredity pancreatitis are relatively equal in men and
women. To exclude a mucinous cyst, testing the fluid for thin
Pseudocysts can be complicated by rupture, hemor- mucin not appreciated on routine cytology can be done
rhage due to erosion into a vessel, obstruction of sur- on cytospin preparations. The typical special stains
rounding structures, and infection. Pseudocysts may be for mucin include mucicarmine and Alcian blue
medically managed, but most are drained or resected. pH 2.5. Chemical analysis of the fluid for amylase and

Ch009-F06731.indd 258 10/26/2006 10:31:48 AM


CHAPTER 9 Pancreas 259

FIGURE 9-11
Pseudocyst. The characteristic
contents of a pseudocyst include
infl ammatory debris, histiocytes,
and, occasionally, bile pigment.
Papanicolaou stain, high power.

DIFFERENTIAL DIAGNOSIS AND PITFALLS


PSEUDOCYST – PATHOLOGIC FEATURES

Cytopathologic Features The differential diagnosis of pseudocyst includes true


៉ Scant cellularity cysts of the pancreas and peri-pancreas. True cysts of
៉ Amorphous cyst debris the pancreas that can be confused with pseudocyst
៉ Infl ammation and bile variable include lymphoepithelial cyst, oligocystic and unilocu-
៉ Histiocytes
lar variants of serous cystadenoma, mucinous cystic
៉ No epithelial cells
neoplasm, and intraductal papillary mucinous neo-
Ancillary Studies
plasm. Peripancreatic cysts in the differential diagnosis
include duodenal duplication cysts, cystic gastro-
៉ Histochemical stains: mucicarmine and/or Alcian blue pH 2.5 are
negative
intestinal stromal tumors, and mesenteric cysts. Solid
៉ Cyst fluid analysis: amylase high; CEA low
neoplasms with cystic degeneration should also be
considered, such as solid-pseudopapillary neoplasm
Differential Diagnosis and Pitfalls and pancreatic endocrine neoplasms.
៉ Lymphoepithelial cyst Contamination of the specimen with epithelial or
៉ Unilocular or oligolocular variant of serous cystadenoma mesothelial cells from the surrounding pancreatic paren-
៉ Mucinous cystic neoplasm chyma, peritoneum, and especially the gastrointestinal
៉ Intraductal papillary mucinous neoplasm tract from EUS-guided biopsies may produce a potential
៉ Solid-pseudopapillary neoplasm diagnostic pitfall. Duodenal epithelium is relatively
៉ Cystic pancreatic endocrine neoplasm characteristic in appearance, presenting as large, folded
៉ Gastrointestinal contamination (pitfall) sheets of glandular epithelium with uniformly spaced,
round, regular nuclei and studded with goblet cells (see
Fig. 9-49). Gastric epithelium is more difficult to recog-
nize and distinguish from the cyst-lining epithelium of
carcinoembryonic antigen (CEA) is also very helpful. a mucin-producing cystic neoplasm, as it often presents
Amylase is consistently elevated (thousands of U/L) in in smaller sheets and clusters as well as single cells, and
pseudocysts, due to the connectivity of the cyst with can display cytoplasmic mucin (see Fig. 9-50). Histio-
the pancreatic ductal system. A cyst fluid with a very cytes can also be mistaken for mucinous epithelial cells,
low amylase level is highly unlikely to be a pseudocyst. leading to the incorrect diagnosis of mucin-producing
Pseudocysts typically have an undetectable or very low neoplastic cyst. On the other hand, the absence of cyst-
CEA level, whereas mucinous cysts generally have lining epithelium and detectable mucin in mucinous
levels of CEA >200 ng/mL. cystic neoplasms may lead to the incorrect diagnosis of

Ch009-F06731.indd 259 10/26/2006 10:31:49 AM


260 FINE NEEDLE ASPIRATION CYTOLOGY

pseudocyst. As such, correlation of the cytologic fea-


tures and ancillary studies with the clinical and radio- LYMPHOEPITHELIAL CYST – PATHOLOGIC FEATURES
logic findings is imperative.
Cytopathologic Findings
៉ Keratinous debris with anucleated and nucleated squamous cells
៉ Background lymphocytes, histiocytes, and cholesterol crystals
៉ May see intact cyst wall with squamous lining cells and
LYMPHOEPITHELIAL CYST subepithelial lymphoid tissue

Differential Diagnosis and Pitfalls


Lymphoepithelial cysts are benign cysts lined by squa- ៉ Pseudocyst
mous epithelium with subepithelial non-neoplastic ៉ Dermoid cyst
lymphoid tissue. ៉ Epidermoid cyst with accessory splenic tissue

CLINICAL FEATURES
CYTOPATHOLOGIC FEATURES
These cysts are rare, representing approximately 0.05%
of all pancreatic cysts. They are much more common The cytologic appearance is similar to that of an epi-
in men than in women, with a male to female ratio of dermal inclusion cyst, with nucleated and anucleated
4 : 1. It is a cyst of older adults, with a mean age of 56 squamous cells, keratinous and cholesterol debris. His-
years. tiocytes and lymphocytes may be noted in the back-
Lymphoepithelial cysts are benign, with no reported ground. Intact cyst wall with subepithelial lymphoid
cases of malignant transformation. Conservative resec- tissue may also be seen (Fig. 9-12).
tion is curative.

DIFFERENTIAL DIAGNOSIS AND PITFALLS


R ADIOLOGIC FEATURES
The differential diagnosis is with other squamous-lined
cysts such as a dermoid cyst of the pancreas and splenic
Lymphoepithelial cysts may be unilocular or multi- epidermoid cyst, both benign cysts more rare than lym-
locular and generally have thick-appearing walls with phoepithelial cyst. A pseudocyst is also in the differential
internal debris that corresponds to keratinous debris. diagnosis due to the necrotic appearance of the kerati-
The cysts average almost 5 cm in diameter, with a wide nous debris. The keratinous nature of the cyst contents
range of little over 1 cm to 17 cm. They can occur any- is usually apparent on close inspection, however.
where in the pancreas, and may even appear
extra-pancreatic.

SOLID NEOPLASMS

LYMPHOEPITHELIAL CYST – DISEASE FACT SHEET


DUCTAL ADENOCARCINOMA
Incidence AND ITS VARIANTS
៉ ∼0.05% of all pancreatic cysts

Gender and Age Distribution Ductal adenocarcinoma is an invasive malignant neo-


៉ Male to female ratio is 4 : 1 plasm arising from the glandular epithelium of the pan-
៉ Mean age 56 years creatic ducts which may produce mucin and which can
have variant morphology including signet-ring cells,
Radiologic Features squamous cells, pleomorphic giant cells, and osteoclast-
៉ Unilocular or multilocular cysts averaging ∼5 cm anywhere in the type giant cells.
pancreas or extra-pancreatic area, with thick-appearing walls and
internal debris

Prognosis and Treatment CLINICAL FEATURES


៉ The cysts are benign and prognosis is excellent
៉ Conservative resection is curative
Ductal adenocarcinoma is the fourth leading cause of
death from cancer in men, women, and overall in the

Ch009-F06731.indd 260 10/26/2006 10:31:50 AM


CHAPTER 9 Pancreas 261

FIGURE 9-12
Lymphoepithelial cyst. A back-
ground of anucleated squamous
cells and keratinous debris are
typical of the contents of a lympho-
epithelial cyst. The presence of an
intact cyst wall with squamous cell
lining and subepithelial lymphoid
tissue may occasionally be seen, as
shown here. Papanicolaou stain,
high power.

sis is directly related to resectability, surgical resection


DUCTAL ADENOCARCINOMA – DISEASE FACT SHEET being the treatment of choice. Patients with unresect-
able tumors typically survive less than 6 months.
Incidence
៉ 11 per 100,000
៉ 4th leading cause of cancer death in men and women
R ADIOLOGIC FEATURES
Gender and Age Distribution
៉ M > F by 30%
៉ Peak incidence, 7th to 8th decade Ductal carcinomas are most commonly staged by high-
resolution helical CT and/or EUS. On CT, tumors
Radiologic Features appear as poorly defined, hypodense masses with
៉ Hypodense mass with a poorly defined periphery and often a central attenuation that deform the normal lobulations
‘double duct’ sign from dilation of both the pancreatic and bile of the pancreas and frequently demonstrate an abrupt
ducts stricture in the main pancreatic duct. When tumors
occur in the pancreatic head, the characteristic ‘double
Prognosis and Treatment duct’ sign corresponding to the dually dilated main
៉ 5-year survival rate is 3–4% pancreatic and bile ducts may be present. On EUS, the
៉ Surgical resection is the treatment of choice tumors typically appear hypoechoic. A loss of a tissue
interface between the tumor and the large peripancre-
atic vessels (superior mesenteric or celiac axis) indi-
cates invasion and stages the tumor as unresectable.
United States. It is the eighth leading cause of cancer
death worldwide. This malignancy, including variants,
represents approximately 90% of all pancreatic malig-
nancies. Heredity accounts for approximately 10% of CYTOPATHOLOGIC FEATURES
cases.
Most cases of pancreatic cancer occur between 60 CONVENTIONAL TUBULAR-TYPE DUCTAL ADENOCARCINOMA
and 80 years of age, and it is more common in men than
in women by about 30%. The morphologic features of ductal adenocarcinoma
The prognosis of pancreatic cancer is universally vary by tumor grade. High-grade (moderately and poorly
poor, with a 5-year survival rate of less than 5%, a rate differentiated) tumors typically do not pose a diagnostic
that has not significantly improved in decades. Progno- challenge, given the overt malignant features of the

Ch009-F06731.indd 261 10/26/2006 10:31:50 AM


262 FINE NEEDLE ASPIRATION CYTOLOGY

Smears of carcinoma typically contain only ductal


DUCTAL ADENOCARCINOMA – PATHOLOGIC FEATURES groups. Smears composed of acinar and ductal groups
should increase the threshold for a malignant diagnosis.
Cytopathologic Findings The presence of granulation tissue and fibrous tissue
៉ Cellularity varies with degree of sclerosis fragments with inflammation are also features that
៉ Relatively pure population of ductal cells are more associated with pancreatitis than with
៉ Loss of honeycomb architecture with nuclear crowding, carcinoma.
overlapping, loss of polarity, and uneven spacing (‘drunken The architectural arrangement of epithelial cells in
honeycomb’)
carcinoma ranges from large crowded sheets to small
៉ Irregular nuclear membranes; may be subtle in well-
differentiated carcinoma
three-dimensional clusters and balls to single cells.
៉ Hyperchromasia generally, but often pale nuclei with Single intact cells are common in high-grade carcinoma
parachromatin clearing in well-differentiated carcinoma and may be scattered in the smear background or seen
៉ Cytoplasm may be scant to abundant and may show at the edge of more cohesive groups (Fig. 9-13). The
vacuolization presence of single atypical intact epithelial cells in well-
៉ Variant cytology: adenosquamous; colloid (mucinous non- differentiated carcinoma is significant and supports a
cystic); undifferentiated (anaplastic); undifferentiated with malignant interpretation.
osteoclast-type giant cells; signet-ring cell In high-grade carcinoma, the cellular features of
malignancy are overt. The cells have a high nuclear to
Ancillary Studies cytoplasmic (N/C) ratio and thus increased nuclear
៉ Immunocytochemical stains (+): cytokeratins AE1/AE3, CAM 5.2, density in cell clusters and sheets relative to benign
CK7, CK8, CK13, CK18, and CK19; CA 19-9, B72.3, CA 125, and ductal groups (Fig. 9-14). Dense nucleation results in
DUPAN-2; MUC1, MUC3, MUC4, and MUC5C
cellular crowding, nuclear overlapping, and loss of
nuclear polarity. The nuclei are enlarged, usually at
Differential Diagnosis and Pitfalls
least two to three times the size of a normal ductal
៉ Chronic pancreatitis, including autoimmune or
cell nucleus, with anisonucleosis of at least three
lymphoplasmacytic sclerosing pancreatitis
៉ Intraductal papillary mucinous neoplasm with invasive
times within a single sheet, hyperchromasia, irregular
carcinoma nuclear membranes, and, often, prominent nucleoli
៉ Mucinous cystic neoplasm with invasive carcinoma (Fig. 9-15).
៉ Metastatic adenocarcinoma In well-differentiated carcinomas, the nuclei are not
៉ Acinar cell carcinoma as enlarged as in high-grade carcinomas (2.5 times the
៉ Pancreatic endocrine neoplasm size of a red blood cell on air-dried smears) (Fig. 9-16),
and can be subtly crowded or show wide and uneven
spacing. When crowded, the nuclei have a tendency to
overlap at least focally and demonstrate a loss of polarity
and loss of smoothness of the nuclear membranes. With
neoplasm, whereas well-differentiated tumors present more abundant, often mucinous cytoplasm, the varia-
more of a challenge. tion in cell size and cytoplasmic volume leads to an
In general, aspiration biopsy of carcinoma produces irregular and uneven spacing of the nuclei in the sheet
cellular smears; however, cellularity can vary greatly or group, which has been termed ‘drunken honeycomb’,
and aspirates may be quite paucicellular in tumors with a feature that contrasts to the generally regular and even
dense sclerosis. Overall cellularity is also related to the nuclear spacing seen in benign ductal groups (Fig. 9-17).
experience of the interventional radiologist and the An extremely benign-appearing, well-differentiated
method of biopsy. The technical difficulty of EUS-guided variant of adenocarcinoma with voluminous mucinous
biopsy relative to percutaneous biopsy can result in a cytoplasm, termed ‘foamy gland adenocarcinoma’ (Fig.
lower cellular yield. Gastrointestinal contamination can 9-18), has been described. The nuclear chromatin in
contribute to what appears to be a highly cellular speci- well-differentiated carcinomas can be hyperchromatic,
men on EUS-guided biopsy and care must be taken to but commonly demonstrates more clearing than clump-
carefully distinguish epithelium from the stomach and ing (parachromatin clearing) (Fig. 9-19). These subtle
duodenum from pancreatic ductal epithelium. nuclear changes coupled with slight nuclear crowding
The smear background can contain helpful clues to and overlapping are the key to a diagnosis of well-
the diagnosis. Abundant background mucin and coagu- differentiated carcinoma.
lative necrosis are features suspicious for, but not inde-
pendently diagnostic of, carcinoma. Coagulative necrosis CYTOMORPHOLOGY OF ADENOCARCINOMA VARIANTS
is cellular necrosis with ghost tumor cells still visible,
which contrasts with fat necrosis, where saponification Adenosquamous carcinoma is an extremely aggressive
and liquefactive necrosis contain foamy histiocytes with variant of ductal adenocarcinoma in which dual lines
the inflammation (see Fig. 9-7). If the biopsy is by EUS, of glandular and squamous differentiation are present.
background mucin may be a contaminant, but contami- The cytologic diagnosis is dependent on recognizing
nating mucin is typically not abundant, as can be present these two malignant cellular components on aspirate
in mucin-producing invasive adenocarcinoma. smears. The diagnosis is straightforward when both the
The composition of the cellular elements on the slide glandular and squamous components are prominent
is important in assessing benign versus malignant. (Fig. 9-20); however, when either component is scant,

Ch009-F06731.indd 262 10/26/2006 10:31:51 AM


CHAPTER 9 Pancreas 263

FIGURE 9-13
Ductal adenocarcinoma. Hyperchro-
matic crowded groups and intact
single malignant cells are architec-
tural features typical of adeno-
carcinoma. Papanicolaou stain, high
power.

FIGURE 9-14
Ductal adenocarcinoma, high grade.
High-grade adenocarcinomas are
relatively easy to recognize due to
the overt features of malignancy,
including dense nucleation, high
N/C ratio, loss of nuclear polarity,
and irregular nuclear membranes.
Papanicolaou stain, high power.

Ch009-F06731.indd 263 10/26/2006 10:31:51 AM


264 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-15
Adenocarcinoma compared to
benign ductal epithelium. The group
of adenocarcinoma on the right
is distinguished from the benign
ductal group on the left by
the nuclear enlargement, uneven
nuclear spacing, and anisonucleosis
of at least three times within a
single sheet. Papanicolaou stain,
low power.

FIGURE 9-16
Adenocarcinoma, well differenti-
ated. The anisonucleosis in well-
differentiated adenocarcinoma is
not as dramatic as with high-grade
carcinomas, demonstrating mild
variability and nuclear enlargement
of approximately 2 1/2 times the size
of a red blood cell (which is noted
to the right of the cell cluster).
Hema III stain, high power.

Ch009-F06731.indd 264 10/26/2006 10:31:52 AM


CHAPTER 9 Pancreas 265

FIGURE 9-17
Adenocarcinoma, well differenti-
ated. The uneven nuclear spacing
combined with abundant volumi-
nous cytoplasm yields a disarray
often referred to as ‘drunken honey-
comb’. Papanicolaou stain, high
power.

FIGURE 9-18
Foamy gland adenocarcinoma. This
variant of well-differentiated ade-
nocarcinoma is characterized by
voluminous mucinous cytoplasm
and a very bland, almost benign
cytologic appearance. Papanicolaou
stain, high power.

Ch009-F06731.indd 265 10/26/2006 10:31:53 AM


266 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-19
Adenocarcinoma, well differenti-
ated. In contrast to hyperchromasia
typical of most adenocarcinomas
of the pancreas, the presence of
nuclear clearing with peripheral
clumping (also known as para-
chromatin clearing) is a feature
characteristic of well-differentiated
neoplasms. Papanicolaou stain,
medium power.

FIGURE 9-20
Adenosquamous carcinoma. This
variant of ductal adenocarcinoma is
recognized by the presence of both
malignant glandular and squamous
components. Papanicolaou stain,
medium power.

Ch009-F06731.indd 266 10/26/2006 10:31:54 AM


CHAPTER 9 Pancreas 267

FIGURE 9-21
Signet-ring cell adenocarcinoma.
This variant of ductal adenocarci-
noma is recognized by the promi-
nence of the signet-ring cell, a
small malignant cell with a cyto-
plasmic vacuole that indents the
nucleus. Papanicolaou stain, high
power.

the diagnosis can be challenging. A careful search for Undifferentiated (anaplastic) carcinoma is a variant
glandular differentiation is warranted in squamous- of ductal adenocarcinoma in which there is a loss of
predominant cases, in particular, given the virtual non- glandular differentiation and tumor cells display large,
existence of primary squamous cell carcinoma of the polygonal to spindled, often bizarre (anaplastic) and
pancreas and thus the potential for a misdiagnosis of a multinucleated features. The aspirate smears may not
metastasis. be hypercellular, but the malignant cells present are
Colloid (mucinous non-cystic) carcinoma on histology unquestionably malignant, with wildly pleomorphic
is similar to colloid carcinomas of the colon and breast, mononuclear and multinuclear tumor giant cells (Fig.
where the vast majority of the infiltrating tumor cells 9-22). These large tumor cells are present in small clus-
(>80% in the pancreas) are present floating in pools of ters, and singly may appear spindled and sarcomatoid.
mucin. These neoplasms are rare and most are found Frequent mitotic activity, cellular cannibalism, and
in association with an intraductal papillary mucinous background necrosis are commonly noted.
neoplasm. Aspiration produces thick viscous mucin that Undifferentiated carcinoma with osteoclast-type giant
may or may not contain an identifiable epithelial com- cells is a rare variant of ductal adenocarcinoma in which
ponent. Recognition of malignant cells in the thick the malignant, usually mononuclear, epithelial cells are
mucin, however, is necessary for the diagnosis of a admixed with benign osteoclast-type giant cells. Approx-
malignant mucin-producing neoplasm. imately 40% of these tumors arise in association with
The distinction from an in-situ neoplasm that may a glandular neoplasm such as conventional adenocarci-
also produce only thick viscous mucin is by the fact that noma and mucinous cystic neoplasms. Aspirate smears
the aspiration is of a solid mass that may or may not be are typically hypercellular with two cell populations, an
arising in the wall of a cyst or dilated duct. The issue of obviously malignant mononuclear epithelial prolifera-
gastrointestinal contamination should not be of concern, tion and benign-appearing osteoclast-like giant cells.
given the abundance of thick mucin. The giant cells vary in number and often contain 10 or
Signet-ring cell carcinoma is a variant of ductal adeno- more bland-appearing, centrally clustered and slightly
carcinoma in which the predominant infiltrating tumor overlapping nuclei with even chromatin and occasion-
cell is a signet-ring cell. Aspirate smears are character- ally prominent nucleoli (Fig. 9-23). The mononuclear
ized by small collections of cells or individual cells with cells appear singly or in small clusters and can range
a vacuole of intracellular mucin that typically indents from medium-sized polygonal epithelioid cells with
the nucleus, a feature that is helpful in distinguishing clear cytoplasm to large bizarre sarcomatoid cells with
the tumor cells from histiocytes (Fig. 9-21). dense and/or spindled cytoplasm.

Ch009-F06731.indd 267 10/26/2006 10:31:55 AM


268 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-22
Undifferentiated carcinoma. This
variant of ductal adenocarcinoma is
characterized by large, bizarre, ana-
plastic or pleomorphic, mononuclear
to multinucleated giant tumor cells.
Papanicolaou stain, high power.

FIGURE 9-23
Undifferentiated carcinoma with
osteoclast-type giant cells. This
rare variant of ductal adenocarci-
noma is composed of a malignant
mononuclear epithelial cell popu-
lation admixed with benign
osteoclast-type giant cells. Papani-
colaou stain, low power.

Ch009-F06731.indd 268 10/26/2006 10:31:56 AM


CHAPTER 9 Pancreas 269

ANCILLARY STUDIES clustered with scattered single cells, whereas acinar


cell carcinoma and pancreatic endocrine neoplasms
produce a more single cell smear pattern with occa-
Special stains for mucin (mucicarmine, Alcian blue sional cell clusters. The cells of high-grade carcinoma
pH 2.5) may demonstrate intracytoplasmic mucin that demonstrate much more nuclear atypia than the cells
helps to classify the neoplasm as an adenocarcinoma. of the typical acinar cell carcinoma and neuroendo-
Immunocytochemical stains can help specify the ade- crine neoplasm, showing significant nuclear membrane
nocarcinoma as pancreatic in origin. Most pancreatic abnormalities and marked hyperchromasia. This is in
adenocarcinomas are positive for cytokeratins AE1/ contrast to the rounder, more central and less convo-
AE3, CAM 5.2, CK7, CK8, CK18, and CK19, with about luted nuclei of both acinar cell carcinoma and pancre-
half expressing CK17 and less than 20% expressing atic endocrine neoplasms. Nucleoli may be prominent
CK20. CEA, B72.3, CA 125, and DUPAN-2 also stain in all three of these neoplasms, but are usually more
tumor cells of pancreatic adenocarcinoma. Mucin prominent and irregular in adenocarcinoma. The chro-
glycoproteins (MUC) are variably expressed, with matin is also more coarse and clumped (‘salt and
most tumors expressing MUC1, MUC3, MUC4, and pepper’) in endocrine neoplasms than in adenocarci-
MUC5AC, and a minority expressing MUC6 (∼25%) noma. The cytoplasmic differences are also important,
and MUC2 (<10%). Antibodies against the protein as high-grade adenocarcinomas display either scant,
product dpc4 correlates with the DPC4/MADH4 gene wispy cytoplasm or abundant, often mucinous cyto-
status, so the loss of dpc4, noted in a little more than plasm, whereas acinar cells tend to have granular cyto-
50% of pancreatic adenocarcinomas, helps to distin- plasm and endocrine neoplasms display eccentric,
guish malignant from benign, since all benign glandu- dense to finely granular, non-mucinous cytoplasm.
lar epithelium of the pancreas demonstrate dpc4 Although extremely rare compared to primary ade-
positivity. nocarcinoma, metastatic malignancies should always be
considered in the differential diagnosis of a high-grade
adenocarcinoma. A common carcinoma metastatic to
DIFFERENTIAL DIAGNOSIS AND PITFALLS the pancreas is renal cell carcinoma. Renal cell carci-
noma is recognized by the large polygonal cells which
may be single, in small clusters, or loosely attached to
The differential diagnosis of pancreatic carcinoma is traversing vessels. The nuclei are usually round and
dependent on grade. High-grade adenocarcinoma can central with large macronucleoli frequently surrounded
be confused with acinar cell carcinoma, pancreatic by a clear halo (‘owl’s eye appearance’) (Fig. 9-24). The
endocrine neoplasms, and metastatic malignancies. cytoplasm is distinguished from that of ductal adenocar-
The smear pattern of adenocarcinoma tends to be more cinoma by the presence of multiple fine vacuoles (best

FIGURE 9-24
Metastatic renal cell carcinoma.
Metastases should always be con-
sidered in the evaluation of carci-
nomas in the pancreas. One of the
more common carcinomas meta-
static to the pancreas, renal cell
carcinoma is characterized by a
polygonal cell shape, clear to dense
cytoplasm, and a prominent central
nucleolus that may be surrounded
by a clear halo, yielding an ‘owl’s
eye’ appearance. Papanicolaou
stain, high power.

Ch009-F06731.indd 269 10/26/2006 10:31:57 AM


270 FINE NEEDLE ASPIRATION CYTOLOGY

appreciated on air-dried smears), in contrast to the often such as the absence of vascular or perineural invasion,
large single vacuoles of adenocarcinoma. and less than two mitoses per high-powered field. As
Well-differentiated adenocarcinoma can be confused such, it is prudent to confine the cytologic diagnosis to
with benign ductal epithelium in chronic pancreatitis ‘pancreatic endocrine neoplasm’ in most cases.
(discussed more fully in the section on chronic pancre-
atitis), high-grade dysplastic ductal epithelium (pancre-
atic intraepithelial neoplasia, PanIN-2 or -3), and with
reactive glandular epithelium from the stomach or R ADIOLOGIC FEATURES
duodenum contaminating the specimen in EUS-guided
biopsies.
By CT scan, most PENs appear as enhancing, solid,
well-circumscribed masses. Cystic degeneration can
occur and calcification is occasionally seen.
PANCREATIC ENDOCRINE NEOPLASM

Pancreatic endocrine neoplasms (PENs) are mostly CYTOPATHOLOGIC FEATURES


low-grade well-differentiated malignant neoplasms of
the endocrine cells of the pancreas, which are classified
Aspirate smears are generally hypercellular, often with
by grade, size, and functional status.
a bloody background, and are composed of a monoto-
nous population of small to medium-sized polygonal
cells. The tumor cells are mostly single (Fig. 9-25), but
CLINICAL FEATURES small to medium-sized groups can also be seen. The
nuclei are round, usually bland and uniform, with
coarse, stippled ‘salt and pepper’ chromatin (Fig. 9-26).
The vast majority of PENs are well-differentiated Nucleoli may be inconspicuous, but can be quite promi-
tumors, representing about 5% of all pancreatic tumors. nent (Fig. 9-26). Nuclear atypia is variable. Significant
The peak age of incidence is 50–60 years of age, but pleomorphism may be present (Fig. 9-27), but this
PENs can occur in all age groups. Men and women are feature should not lead to a malignant diagnosis. The
equally affected. relatively scant cytoplasm is usually dense and eccen-
The biologic behavior of PENs is difficult, if not tric, yielding a plasmacytoid appearance. Rare variants
impossible, to predict from cytologic evaluation. The of PEN, such as oncocytic (Fig. 9-28) and clear cell
architectural pattern and cellular pleomorphism, fea-
tures of the neoplasms that can be evaluated on cytol-
ogy, do not correlate with prognosis. Prognosis is highly
dependent on tumor size at detection, a feature that is PANCREATIC ENDOCRINE NEOPLASM – PATHOLOGIC FEATURES
often related to the association of the neoplasm with a
syndrome, in addition to ‘favorable’ histologic features Cytopathologic Findings
៉ Dyshesive, single cell smear pattern; few small clusters
៉ Uniform, monotonous population of cells with plasmacytoid
features
៉ Coarse, speckled, ‘salt and pepper’ chromatin pattern
PANCREATIC ENDOCRINE NEOPLASM – DISEASE FACT SHEET
៉ Nucleoli may be prominent
៉ Dense, finely granular cytoplasm; rarely clear or oncocytic
Incidence
cytoplasm
៉ ∼2–5% of pancreatic neoplasms
៉ ∼50% are functional and 50% non-functional
Ancillary Studies
៉ Histochemical stains: positive for Grimelius stain and PAS
Gender and Age Distribution
៉ Immunocytochemical stains: cytokeratins CAM 5.2 and AE1/AE3
៉ M = F are usually positive; chromogranin, synaptophysin, Leu-7, and
៉ Any age, but most between 40 and 60 years NSE are typically positive; insulin, glucagon, somatostatin, and
pancreatic polypeptide are variably positive; gastrin, vasoactive
Radiologic Features intestinal polypeptide, cholecystokinin, and adrenocorticotropic
៉ Solid, well-circumscribed masses, usually small (<2 cm), but may hormone may occasionally be positive
be large (>6 cm); can be cystic ៉ Electron microscopy: neurosecretory granules

Prognosis and Treatment Differential Diagnosis and Pitfalls


៉ Prognosis is related to tumor size, mitotic rate, necrosis, extra- ៉ Acinar cell carcinoma
pancreatic invasion, vascular invasion, and nodal or distant ៉ Solid-pseudopapillary neoplasm
metastases ៉ Pancreatoblastoma
៉ Small neoplasms without adverse prognostic features are curable ៉ Plasmacytoma
by surgical resection ៉ Normal acinar cell epithelium (pitfall)

Ch009-F06731.indd 270 10/26/2006 10:31:58 AM


CHAPTER 9 Pancreas 271

FIGURE 9-25
Pancreatic endocrine neoplasm. The
typical aspirate smears are hyper-
cellular, composed of a predomi-
nantly single cell population of
small to medium-sized, monotonous
polygonal cells that may be associ-
ated with a prominent vascular
component. Papanicolaou stain,
medium power.

FIGURE 9-26
Pancreatic endocrine neoplasm
(PEN) with cystic degeneration.
Cystic PENs are cytologically similar
to solid PENs. The typical coarse
stippled ‘salt and pepper’ chromatin
pattern is similar in both neo-
plasms. Although nucleoli are not
very prominent in most PENs, they
can be quite prominent, as illus-
trated here. Papanicolaou stain,
high power.

Ch009-F06731.indd 271 10/26/2006 10:31:58 AM


272 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-27
Pancreatic endocrine neoplasm. The
presence of nuclear pleomorphism
and mitotic activity cannot be used
as criteria for the diagnosis of
malignancy. Papanicolaou stain,
high power.

FIGURE 9-28
Pancreatic endocrine neoplasm,
oncocytic variant. The presence of
abundant dense, oncocytic cyto-
plasm characterizes this variant.
Papanicolaou stain, medium power.

Ch009-F06731.indd 272 10/26/2006 10:31:59 AM


CHAPTER 9 Pancreas 273

FIGURE 9-29
Pancreatic endocrine neoplasm
(PEN), clear cell variant. In contrast
to the typical dense granular cyto-
plasm of most PENs, the cytoplasm
of the clear cell variant demon-
strates cytoplasmic vacuolization
ranging from large vacuoles to small
multiple vacuoles. These neoplasms
can identically mimic renal cell car-
cinomas. Hema III stain, high
power.

(Fig. 9-29), exist. Mitotic figures and necrosis are DIFFERENTIAL DIAGNOSIS AND PITFALLS
uncommon features but may be seen on cytology and
are features that should be noted in the cytology report.
The cytologic features of a cystic PEN are similar to The differential diagnosis is dependent on the degree
those of solid neoplasms. High-grade large cell neuro- of cellular atypia. The differential diagnosis of
endocrine carcinoma and small cell neuroendocrine well-differentiated PEN most commonly includes solid-
carcinoma are neoplasms that appear cytologically pseudopapillary tumor, acinar cell carcinoma, non-
malignant, resembling similar neoplasms in the lung. Hodgkin lymphoma, and plasmacytoma. Clear cell and
oncocytic PEN variants can be confused with primary
and metastatic neoplasms with similar cytoplasmic fea-
tures. High-grade PEN demonstrating significant cel-
ANCILLARY STUDIES lular atypia and pleomorphism can be misdiagnosed as
high-grade pancreatic adenocarcinoma and other high-
grade malignancies. Normal acinar cells when aspi-
Documenting endocrine differentiation is most com- rated in abundance and with a disrupted architecture
monly performed with immunocytochemical staining are a pitfall for a false positive diagnosis of PEN (see
for chromogranin and/or synaptophysin, markers posi- Fig. 9-10).
tive in over 95% of tumors. Synaptophysin produces One of the more common neoplasms to be confused
a consistently strong and diffuse staining pattern, with PEN in women especially is the solid-pseudopapil-
whereas chromogranin staining can be quite focal. lary tumor. This tumor is distinguished by papillary
CD57 (Leu-7) and CD56 (neural cell adhesion mole- groups with vascular cores and myxoid stroma, but this
cule) also stain most tumors. Labeling the tumors for smear pattern may not be well demonstrated in all cases.
specific peptides demonstrates a wide range of positiv- When absent, the bland, oval more than round nuclei,
ity, the nature of which does not necessarily correlate nuclear grooves, small perinuclear vacuoles, and scant
with a syndrome. Most PENs also stain with cytokera- non-plasmacytoid cytoplasm help to diagnose a solid-
tins CAM 5.2 and AE1/AE3. pseudopapillary tumor. Recognizing the stippled chro-
Flow cytometry of aspirated tissue may be useful in matin of PEN is also helpful.
distinguishing PEN from non-Hodgkin lymphoma and Acinar cell carcinoma can also closely resemble PEN
plasmacytoma. on smears. Aspirates of PEN produce mostly single cells
Electron microscopy will identify numerous and and some cell clusters, in contrast to acinar cell carci-
randomly distributed neurosecretory granules in PEN. noma, where cell clusters predominate. Although nucle-

Ch009-F06731.indd 273 10/26/2006 10:32:00 AM


274 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-30
Plasmacytoma. Although rare in the
pancreas, plasmacytoma with the
typical single cell smear pattern,
eccentric nucleus, and coarse stip-
pled chromatin mimics a pancreatic
endocrine neoplasm (PEN). Notice,
however, the chromatin clumps in a
clock-face distribution, in contrast
to the even distribution throughout
the nucleus in PEN. Also notice the
pale perinuclear huff (Golgi zone)
in the cell at 11 o’clock. Papanico-
laou stain, high power.

oli are typically more prominent in acinar cell carcinoma,


PEN can demonstrate very prominent nucleoli, so this ACINAR CELL CARCINOMA – DISEASE FACT SHEET
feature is not discriminatory, but the characteristic
stippled ‘salt and pepper’ chromatin pattern of PEN, a Incidence
pattern absent in acinar cell carcinoma, is useful. ៉ ∼1–2% of adult pancreatic neoplasms
Granular cytoplasm, when present and visible on smears,
helps to define an acinar cell carcinoma, in contrast Gender and Age Distribution
to the dense to finely granular eccentric cytoplasm of ៉ Male to female ratio is 4 : 1
PEN. ៉ Children << adults; peak incidence, 7th decade
Non-Hodgkin lymphoma enters the differential diag-
nosis owing to the single cell smear pattern common Radiologic Features
to both neoplasms and the tendency of the delicate ៉ Solid masses with well-demarcated borders; rarely cystic
cytoplasm of some PENs to get stripped from the cells,
yielding small round cells mimicking lymphoma. The Prognosis and Treatment
differences in the chromatin pattern, cytoplasm of intact ៉ Prognosis is directly related to tumor stage and is better than for
PEN cells, and the absence of lymphoglandular bodies, ductal adenocarcinoma
៉ Resection is the treatment of choice
stripped globules of cytoplasm from the delicate lym-
phoma cells, help to differentiate PEN from lymphoma.
Extracellular plasmacytoma shares features of eccentric
cytoplasm and stippled chromatin, but the distribution
of the chromatin is more ‘clock-face’ in plasmacytoma CLINICAL FEATURES
and the eccentric cytoplasm of PEN does not contain a
perinuclear huff (Golgi zone) as may be seen in plasma-
Acinar cell carcinoma is a malignant neoplasm of adults
cytoma (Fig. 9-30).
and children, comprising 1–2% of pancreatic neoplasms
in adults and 15% of pancreatic neoplasms in children.
The tumors are most common in adults. Most tumors
ACINAR CELL CARCINOMA occur in the seventh decade and the male to female
ratio is almost 4 : 1. In the pediatric group, the tumors
generally occur between 10 and 20 years of age.
Acinar cell carcinoma is a malignant proliferation of Acinar cell carcinomas generally have a poor progno-
the enzyme-producing exocrine (acinar) cells of the sis, with a median survival of about 11/2 years. The
pancreas. reported 5-year survival is between 6% and 25%. Chil-

Ch009-F06731.indd 274 10/26/2006 10:32:01 AM


CHAPTER 9 Pancreas 275

FIGURE 9-31
Acinar cell carcinoma. The presence
of stripped naked nuclei with a
granular background from the dis-
persed cytoplasm is a rather char-
acteristic feature of this neoplasm.
Hema III stain, high power.

dren are believed to have a slightly better prognosis,


with reports of long-term survival and cure in this ACINAR CELL CARCINOMA – PATHOLOGIC FEATURES
population.
Cytopathologic Findings
៉ Small glandular clusters, single cells
៉ Stripped naked nuclei; +/− loose cytoplasmic granules
R ADIOLOGIC FEATURES ៉ May be disarmingly bland, with a polygonal cell shape and low
N/C ratio
៉ Coarse chromatin usually with prominent nucleoli, but not
Acinar cell carcinomas are typically solid, occasionally always
cystic, well-circumscribed, usually large masses ៉ Granular cytoplasm, variably prominent
(average size, 10 cm) that may arise in any portion of
the pancreas. Ancillary Studies
៉ Histochemical stains: positive for PAS/dPAS
៉ Immunocytochemical stains: trypsin, chymotrypsin, lipase, and
elastase usually positive; amylase is usually negative; positive
CYTOPATHOLOGIC FEATURES for CAM 5.2 and AE1/AE3, but negative for cytokeratins 7, 19,
and 20; EMA is positive in about 50%
Aspirate smears of acinar cell carcinoma are usually
Differential Diagnosis and Pitfalls
moderately cellular and often display a relatively clean
៉ Pancreatic endocrine neoplasms
background devoid of necrosis and cyst debris, but the
៉ Solid-pseudopapillary tumor
background may contain cytoplasmic granules from the
៉ Pancreatoblastoma
stripped cytoplasm of tumor cells. Stripped cells leave ៉ Normal acinar epithelium (pitfall)
naked tumor nuclei, a rather characteristic feature of
acinar cell carcinoma (Fig. 9-31). These nuclei can
resemble lymphocytes, especially when nucleoli are not
prominent, but when compared to the intact tumor
cells, the similarity is apparent. The clusters of tumor able degree the normal acinar cell. The cells can be
cells form irregular-shaped groups, sheets, and small pleomorphic, and may occasionally appear plasmacy-
glandular clusters (Fig. 9-32), a pattern that contrasts toid; however, the marked and rather diffuse plasma-
to the organoid, grape-like clustering typical of benign cytoid appearance common in endocrine neoplasms is
acinar epithelium. Individual tumor cells are frequently not present in acinar cell carcinoma.
very bland with a polygonal cell shape, low N/C ratio, The nuclear chromatin in the tumor cells is generally
and uniform nuclei (Fig. 9-33), resembling to a remark- coarsely clumped, and one, sometimes two, prominent

Ch009-F06731.indd 275 10/26/2006 10:32:01 AM


276 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-32
Acinar cell carcinoma. In contrast
to the organoid grape-like cluster-
ing of benign acinar epithelial cells,
the cell clusters of acinar cell car-
cinoma are of varying sizes and are
irregular in shape. Papanicolaou
stain, low power.

FIGURE 9-33
Acinar cell carcinoma. The individ-
ual tumor cells are frequently quite
bland with a polygonal cell shape,
low N/C ratio, and uniform nucleus.
Prominent nucleoli may not be
present, and the cells can resemble
normal acinar epithelial cells. Papa-
nicolaou stain, high power.

Ch009-F06731.indd 276 10/26/2006 10:32:02 AM


CHAPTER 9 Pancreas 277

FIGURE 9-34
Acinar cell carcinoma. The dense
granularity of the cytoplasm is best
appreciated on H&E-stained smears.
H&E stain, high power.

nucleoli may be seen. This feature and the coarsely endocrine markers chromogranin and synaptophysin
granular cytoplasm are helpful diagnostic findings when should not be misinterpreted.
present. The cytoplasmic granularity is not always
readily apparent and is more easily identified on
hematoxylin and eosin (H&E)-stained smears than on DIFFERENTIAL DIAGNOSIS AND PITFALLS
Papanicolaou- or Romanowsky-stained smears (Fig.
9-34). The absence of striking cytoplasmic granularity
and large, prominent nucleoli are not uncommon in The cytologic differential diagnosis includes PEN,
acinar cell carcinoma, adding to the diagnostic diffi- pancreatoblastoma, solid-pseudopapillary tumor, and
culty. Identification of mitotic activity supports a neo- non-neoplastic acinar parenchyma. The most common
plastic process. neoplasm mistaken cytologically for acinar cell carci-
noma is the PEN. These neoplasms have in common
a uniformity of nuclei, the presence of numerous
ANCILLARY STUDIES individual cells, and a moderate amount of dense non-
vacuolated cytoplasm. Endocrine neoplasms tend to
have a more uniform, dyshesive, single cell smear pattern
Documenting the presence of zymogen granules or exo- with plasmacytoid cells, and less well-developed micro-
crine enzyme production by the tumor cells establishes glandular (acinar) groups than has acinar cell carci-
the diagnosis of acinar cell carcinoma. When sufficient noma. Acinar cell carcinoma also lacks the typical
in quantity, a periodic acid–Schiff (PAS) stain with endocrine ‘salt and pepper’ chromatin pattern. However,
and without diastase digestion will highlight the red- given that endocrine tumors can demonstrate prominent
staining intracytoplasmic granules. Electron micros- nucleoli and acinar cell carcinomas may not demonstrate
copy can also be used to detect the granules. Although obviously granular cytoplasm, many cases require
not widely used, the butyrate esterase stain detects immunohistochemical staining for accurate diagnosis.
lipase activity in about 75% of cases, and 75–95% of Features helpful in distinguishing solid-pseudo-
cases will label with antibodies to exocrine enzymes papillary tumor from acinar cell carcinoma include the
including trypsin, chymotrypsin, lipase, and elastase. formation of pseudopapillary structures with the char-
Low and high molecular weight cytokeratins (CAM 5.2 acteristic myxoid stroma, the fragility of the cytoplasm
and AE1, respectively) as well as cytokeratins 8 and 18 (with numerous stripped nuclei), and the presence of
stain the carcinoma cells, but cytokeratins 7, 19, and longitudinal nuclear grooves and small perinuclear
20 are typically negative. Because acinar cell carcino- vacuoles in solid-pseudopapillary tumors.
mas can contain a small number of endocrine cells (1– Pancreatoblastoma is nearly impossible to distinguish
2%), positive immunocytochemical staining with the from acinar cell carcinoma on the basis of cytology,

Ch009-F06731.indd 277 10/26/2006 10:32:03 AM


278 FINE NEEDLE ASPIRATION CYTOLOGY

since the characteristic squamoid corpuscles of pancre- presence of metastases at the time of diagnosis. Patients
atoblastoma are rarely detectable in cytologic smears. typically present with unresectable tumors and over a
Since both of these neoplasms exhibit acinar differentia- third have metastases upon diagnosis.
tion by immunohistochemistry, it may be impossible to
exclude a pancreatoblastoma in a neoplasm otherwise
having cytologic features of acinar cell carcinoma. R ADIOLOGIC FEATURES
The misinterpretation of benign acinar cells as neo-
plastic (acinar cell carcinoma), and vice versa, are diag-
nostic pitfalls. Rarely, aspiration of normal pancreatic CT scans reveal a well-defined, often large (up to 20 cm),
parenchyma may produce a smear with many acini heterogenous and mostly enhancing pancreatic mass,
without the typical organoid grape-like clustering and the minority of which will demonstrate scattered
uniform acinar formation, raising the possibility of calcifications.
acinar cell carcinoma. Similarly, a scantily cellular aspi-
rate of a neoplasm may be mistaken for normal acini.
CYTOPATHOLOGIC FEATURES
PANCREATOBLASTOMA
Aspirate smears of pancreatoblastoma are cellular,
composed of cell clusters and single cells. The appear-
Pancreatoblastoma is a malignant neoplasm with mul- ance of the epithelial cells will vary depending on the
tiple lines of epithelial differentiation, most commonly direction of differentiation. Cells with acinar differen-
acinar differentiation, which also contains squamous tiation have an oval to cuboidal cell shape, round,
nests and usually a mesenchymal component. central to eccentric nuclei, one or more small nucleoli,
and a moderate amount of granular, amphophilic to
eosinophilic cytoplasm that is impossible to distinguish
CLINICAL FEATURES from acinar cell carcinoma (Fig. 9-35). Cells with endo-
crine differentiation can resemble acinar cells but
demonstrate a more cuboidal shape, a higher N/C ratio,
Pancreatoblastoma is an extremely rare neoplasm of denser, less granular cytoplasm, and less conspicuous
adults and children, with approximately two-thirds nucleoli. Squamoid cells, singly or in clusters, are rec-
occurring in children and one-third occurring in adults.
It is the most common malignant pancreatic tumor in
children, representing approximately 25% of all pan- PANCREATOBLASTOMA – PATHOLOGIC FEATURES
creatic neoplasms in the pediatric population.
There is a bimodal age distribution when pediatric Cytopathologic Findings
and adult populations are evaluated separately. In chil- ៉ Cellular smears with varying sized cell clusters and single cells
dren, the peak age is almost 21/2 years, whereas in the ៉ Epithelial cells will vary in appearance depending on their
adult, the peak age is 40 years. As a whole, the average direction of differentiation: cells with acinar differentiation
age is about 10 years. Across all ages, the male to female dominate and have a polygonal shape, round, central to
ratio is equal. eccentric nucleus, one or more small nucleoli, and a moderate
Prognosis appears to be worse in adults than in chil- amount of granular cytoplasm
៉ Squamoid nests are recognized in cell block preparations
dren, and is dependent on tumor resectability and the
៉ Stromal fragments with traversing capillaries may be present

Ancillary Studies
PANCREATOBLASTOMA – DISEASE FACT SHEET ៉ Histochemical stains:
៉ Acinar cells: positive for PAS/dPAS
Incidence ៉ Immunocytochemical stains:
៉ Acinar cells: positive for pancytokeratin, trypsin,
៉ Rare in adults; 25% of pancreatic tumors in children
chymotrypsin, and lipase
៉ Endocrine cells: positive for chromogranin, synaptophysin,
Gender and Age Distribution
and NSE, but usually negative for insulin, glucagon, or
៉ M = F somatostatin
៉ Mean age in children is 21/2 years, and in adults, 40 years ៉ Ductal cells: positive for cytokeratin, CEA, B72.3, and
DUPAN-2
Radiologic Features ៉ Stroma: positive for vimentin
៉ A well-defined heterogeneous pancreatic or peripancreatic mass ៉ Squamoid corpuscles are generally not immunoreactive
that may contain calcifications
Differential Diagnosis and Pitfalls
Prognosis and Treatment ៉ Acinar cell carcinoma
៉ Prognosis is generally poor and is worse in adults than in ៉ Pancreatic endocrine neoplasm
children ៉ Solid-pseudopapillary neoplasm
៉ Surgical resection is the treatment of choice ៉ Normal acinar epithelium (pitfall)

Ch009-F06731.indd 278 10/26/2006 10:32:04 AM


CHAPTER 9 Pancreas 279

FIGURE 9-35
Pancreatoblastoma. Acinar differ-
entiation is the most common cell
line of differentiation in this neo-
plasm, which can undergo multiple
lines of epithelial differentiation.
As such, pancreatoblastoma can
identically mimic an acinar cell
carcinoma. Hema III stain, high
power.

ognized in cell block preparations rather than on direct


smears (Fig. 9-36). Stromal fragments may be scant or SEROUS CYSTADENOMA – DISEASE FACT SHEET
prominent, and traversing capillaries may be seen.
Incidence
៉ 2% of all pancreatic neoplasms but ∼10% of resected cysts
ANCILLARY STUDIES
Gender and Age Distribution
៉ F >> M
The neoplastic cells will be highlighted by histochemi- ៉ Mean age 65 years
cal and immunohistochemical markers depending on
the line of differentiation of the cells. Most neoplasms Radiologic Features
develop an acinar cell phenotype and therefore the ៉ Large, mostly microcystic, masses with characteristic ‘soap bubble’
tumor cells will demonstrate zymogen granules that pattern on ultrasound
stain with PAS/dPAS and exocrine antibodies such as ៉ Central stellate scar frequently with ‘starburst’ pattern of
trypsin, chymotrypsin and lipase. microcalcifications noted on CT
៉ Oligocystic, macrocystic, and unilocular variants can occur

DIFFERENTIAL DIAGNOSIS AND PITFALLS Prognosis and Treatment


៉ The cysts are benign and prognosis is excellent
៉ Therapy includes resection, or observation for small (<4 cm) cysts
The primary neoplasm in the differential diagnosis is in poor surgical candidates
acinar cell carcinoma, which is discussed in detail on
page 277–278.

CLINICAL FEATURES
CYSTIC NEOPLASMS
The incidence of these often asymptomatic neoplasms
is increasing with the common use of CT scanning for
SEROUS CYSTADENOMA the work-up of patients for other conditions. Of all the
neoplasms arising from the exocrine pancreas, serous
cystadenomas account for roughly 2%, but of resected
Serous cystadenoma is a benign, typically microcystic, cysts of the pancreas, the percentage increases to 10%.
proliferation of small cuboidal epithelial cells rich in There is an association with von Hippel–Lindau
cytoplasmic glycogen which produces serous fluid. syndrome.

Ch009-F06731.indd 279 10/26/2006 10:32:04 AM


280 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-36
Pancreatoblastoma. This cell block
preparation demonstrates a cluster
of squamoid cells surrounded by
neoplasm with acinar cell differen-
tiation. The cell block is the best
preparation to identify squamoid
cells. This finding helps distinguish
pancreatoblastoma from acinar cell
carcinoma. H&E stain, high power.

Serous cystadenomas are much more common in


women than in men and have greater than a 70-year age SEROUS CYSTADENOMA – PATHOLOGIC FEATURES
range (18–91 years) with a mean of approximately 65
years. Cytopathologic Findings
These neoplasms are benign, with very rare reports ៉ Scantily cellular smears with clean or bloody background
of malignant transformation. Complications of large neo- ៉ Uniform cuboidal cells with round nuclei, smooth nuclear
plasms such as rupture or erosion into a large vessel can membranes, and inconspicuous nucleoli
៉ Scant, pale to finely vacuolated but non-mucinous cytoplasm
cause death. Complete surgical resection is curative.
Ancillary Studies
R ADIOLOGIC FEATURES ៉ Histochemical stains: PAS-positive, dPAS-negative; confirms
cytoplasmic glycogen
៉ Immunocytochemical stains (+): cytokeratins AE1/AE3, CAM 5.2,
Microcystic serous cystadenoma often demonstrates CK7, CK8, CK18, and CK19; CEA, MUC2, and MUC5
very characteristic radiographic findings precluding the ៉ Cyst fluid analysis: low amylase and CEA
need for tissue confirmation. Ultrasound demonstrates
a ‘soap bubble’ pattern owing to the numerous micro- Differential Diagnosis and Pitfalls
cysts that average less than 2 cm each. CT scan shows ៉ Pseudocyst
៉ Mucinous cysts: mucinous cystic neoplasm and side-branch
a well-defined mass with multiple, sometimes innumer-
intraductal papillary mucinous neoplasm
able, small cysts separated by delicate septa. Many
៉ Gastrointestinal contamination (pitfall)
tumors will contain a central stellate scar, and about ៉ Histiocytes (pitfall)
30% of these will demonstrate a ‘starburst’ pattern of
microcalcifications within the scar. Oligocystic and
unilocular variants of serous cystadenomas do exist
and produce radiologic features that usually cannot be
most mucinous cysts. Smears are frequently very
distinguished from other cysts in the pancreas. A solid
paucicellular and it is not uncommon for smears to be
variant has also been reported.
acellular. Many cases are considered inadequate for
interpretation due to insufficient cellularity, but iden-
CYTOPATHOLOGIC FEATURES tification of even a couple of small clusters of cells with
bland cuboidal morphology can be very helpful in the
appropriate clinical setting, even if not technically con-
The fluid aspirated from serous cystadenomas is clear sidered ‘diagnostic’. Tumor cells are uniform cuboidal
and thin and may be bloody, but is not mucoid as in cells in small clusters and flat sheets with round, central

Ch009-F06731.indd 280 10/26/2006 5:58:33 PM


CHAPTER 9 Pancreas 281

FIGURE 9-37
Serous cystadenoma. The tumor
cells are uniform cuboidal cells with
bland central nuclei, smooth nuclear
membranes, and even chromatin.
The cytoplasm is finely vacuolated
but non-mucinous. The cells may
resemble histiocytes. Papanicolaou
stain, high power.

to slightly eccentric nuclei and scant but visible cyto-


plasm that is homogeneous to finely vacuolated (Fig.
9-37). The nuclei have smooth nuclear membranes,
an even chromatin pattern, and inconspicuous to no
nucleoli, and the cytoplasm is scant and finely vacuo-
lated or dense, but not mucinous (Fig. 9-38).

ANCILLARY STUDIES

PAS stain with and without diastase will confirm the


presence of cytoplasmic glycogen and exclude the pres-
ence of mucin. Tumor cells immunolabel with cyto-
keratins CAM 5.2, AE1/AE3, CK7, CK8, CK18, and
CK19. MUC6 and α-inhibin are positive in most cases,
and MUC1 is positive in about one-third. CEA, MUC2
and MUC5, and all endocrine markers are negative.
Cyst fluid analysis typically shows low levels of amylase
and CEA.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

Bloody specimens that may contain inflammation and


debris and lack epithelial cells can be misinterpreted as
pseudocysts. Cyst fluid analysis helps by showing an FIGURE 9-38
elevated amylase level in pseudocysts. Smears with epi- Serous cystadenoma. This air-dried preparation highlights the non-
thelial cells demonstrating vacuolated cytoplasm may mucinous finely vacuolated cytoplasm. Hema III stain, high power.
indicate the presence of a neoplastic mucinous cyst (see
below).

Ch009-F06731.indd 281 10/26/2006 10:32:05 AM


282 FINE NEEDLE ASPIRATION CYTOLOGY

Gastrointestinal epithelium and mucin contaminat- demonstrate a typically large, well-demarcated, solid
ing the smears from EUS-guided biopsies adds to this and cystic heterogeneous neoplasm commonly located
diagnostic pitfall. Cyst fluid analysis is helpful if CEA is in the tail of the pancreas of a young woman. Calcifica-
elevated above 200 ng/mL. tions may also be noted.

SOLID-PSEUDOPAPILLARY NEOPLASM CYTOPATHOLOGIC FEATURES

Solid-pseudopapillary neoplasm is a low-grade malig- Aspirate smears are hypercellular with a population of
nant solid neoplasm with secondary cystic degenera- small, uniform cells in cohesive, often branching and
tion that produces a proliferation of small bland cells papillary cell clusters, interspersed with many single
of uncertain lineage in solid nests and around delicate cells (Fig. 9-39). The background may be clean or filled
blood vessels, creating a pseudopapillary appearance on with hemorrhagic cyst debris laden with foamy histio-
histology. cytes and multinucleated giant cells. Delicate fibrovas-
cular cores with myxoid stroma and a zone of cytoplasm
sometimes separating the nuclei of the cells from the
vessel is a diagnostic finding (Fig. 9-40). Individual
CLINICAL FEATURES tumor cells are uniform with round to oval nuclei,
smooth to slightly indented or grooved nuclear mem-
branes, even and finely granular chromatin, and incon-
This neoplasm is relatively rare, accounting for less spicuous nucleoli (Fig. 9-41). The cytoplasm is scant to
than 3% of all pancreatic malignancies. It occurs almost moderate, non-granular to finely granular, and may be
exclusively in women, with an approximate female to stripped from the nucleus. When intact, a small peri-
male ratio of 9 : 1. Most women are in their third decade, nuclear vacuole and, occasionally, intracytoplasmic
but the reported ages range from 7 to 79 years. hyaline globule may be noted (Fig. 9-42).
Surgical resection is the treatment of choice. The
overall prognosis is excellent, with only about 15% of
patients developing recurrence or metastatic disease,
and this is typically the result of incomplete resection. SOLID-PSEUDOPAPILLARY NEOPLASM – PATHOLOGIC FEATURES
Complete resection of node-negative patients is con-
sidered curative. Even with recurrence or metastatic Cytopathologic Findings
disease, long-term survival is common. ៉ Highly cellular smears with branching and papillary cell clusters
interspersed with single cells
៉ Smear background may be clean or filled with hemorrhagic cyst
debris, foamy histiocytes, and multinucleated giant cells
R ADIOLOGIC FEATURES ៉ Fibrovascular cores with myxoid stroma is characteristic
៉ Individual tumor cells are homogeneous with little
anisonucleosis and no mitotic activity
Like for serous cystadenomas, solid-pseudopapillary ៉ Nuclei are round to oval with smooth nuclear membranes except
neoplasms are often diagnosed from the characteristic for frequent nuclear grooves or focal indentations, finely
clinical and radiologic features. CT and ultrasound granular chromatin, and inconspicuous nucleoli
៉ Cytoplasm is scant to moderate and may contain a small
perinuclear vacuole or intracytoplasmic hyaline globule

SOLID-PSEUDOPAPILLARY NEOPLASM – DISEASE FACT SHEET Ancillary Studies


៉ Histochemical stains: myxoid stroma is positive for PAS/dPAS
Incidence ៉ Immunocytochemical stains: positive for vimentin, α-1-
antitrypsin, CD10, NSE, CD56, and progesterone receptor;
៉ ∼1–3% of all pancreatic malignancies, 6% of exocrine tumors,
synaptophysin and cytokeratin (AE1/AE3, CAM 5.2) are variably
and ∼24% of all surgically resected cystic lesions in the pancreas
positive; CK7 and CK19 are usually negative; β-catenin will
demonstrate abnormal cytoplasmic and nuclear staining in
Gender and Age Distribution
>90% of tumors; intracytoplasmic hyaline globules stain with
៉ ∼90% of patients are female antibodies to α-1-antitrypsin
៉ Most are in their 20s; mean age is 28 years ៉ Molecular analysis: almost all harbor somatic point mutations in
exon 3 of the β-catenin gene
Radiologic Features
៉ Well-circumscribed mass with solid and cystic components, Differential Diagnosis and Pitfalls
commonly in the pancreatic tail ៉ Pancreatic endocrine neoplasm
៉ Pseudocysts
Prognosis ៉ Acinar cell carcinoma
៉ Excellent – only 15% develop recurrence or metastases ៉ Normal acinar epithelium (pitfall)

Ch009-F06731.indd 282 10/26/2006 10:32:06 AM


CHAPTER 9 Pancreas 283

FIGURE 9-39
Solid-pseudopapillary neoplasm.
The typical aspirate smears are
hypercellular, composed of a popu-
lation of small uniform cells in
cohesive, often branching and pap-
illary cell clusters interspersed with
many single cells. Papanicolaou
stain, high power.

FIGURE 9-40
Solid-pseudopapillary neoplasm.
The myxoid stroma that separates
the tumor cells from the vessels can
be seen as globules within acinar
structures, as demonstrated here.
Papanicolaou stain, medium power.

Ch009-F06731.indd 283 10/26/2006 10:32:06 AM


284 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-41
Solid-pseudopapillary neoplasm.
The individual tumor cells are bland
with oval to indented nuclei, even
chromatin, nuclear grooves, and no
nucleoli. Note the characteristic
small perinuclear vacuole seen in
some of the cells. Papanicolaou
stain, high power.

FIGURE 9-42
Solid-pseudopapillary neoplasm.
Intracytoplasmic hyaline globules
may also be seen in some tumor
cells, as demonstrated in this neo-
plasm, most prominent in the cell
at 2 o’clock. Note also the occa-
sional perinuclear vacuole. Hema
III stain, high power.

Ch009-F06731.indd 284 10/26/2006 10:32:08 AM


CHAPTER 9 Pancreas 285

ANCILLARY STUDIES
MUCINOUS CYSTIC NEOPLASM – DISEASE FACT SHEET

Solid-pseudopapillary neoplasms often do not express Incidence


cytokeratin (CAM 5.2 or AE1/AE3) and usually do not ៉ ∼6% of all primary pancreatic neoplasms and pancreatic cysts
express cytokeratins 7 and 19. Most tumors stain with
antibodies to vimentin, CD10 (neprilysin), neuron- Gender and Age Distribution
specific enolase (NSE), CD56, β-catenin, cyclin D1, ៉ Almost all female
progesterone receptors, and α-1-antitrypsin, a marker ៉ 40–50 years
that also stains the intracytoplasmic hyaline globules.
Cyst fluid analysis demonstrates low amylase and CEA Radiologic Features
levels. ៉ Typically, solitary, multiloculated, well-circumscribed masses,
often with peripheral calcifications and a thick capsule; 90% in
the body or tail
៉ Communication with the large pancreatic ducts is absent in
almost all cases
DIFFERENTIAL DIAGNOSIS AND PITFALLS
៉ A cyst wall mass suggests an invasive component

The primary solid neoplasm in the differential diagno- Prognosis and Treatment
sis is PEN. The uniform, bland-appearing, often dis- ៉ Prognosis is directly related to the presence or absence of
persed epithelial cells of solid-pseudopapillary neoplasm invasive carcinoma
៉ Surgical resection is the treatment of choice and is curative for
can greatly resemble those of PEN. Careful attention to
non-invasive neoplasms
the nuclear details, including the presence of nuclear
grooves, indentations, and the occasional perinuclear
vacuole or intracytoplasmic hyaline droplet seen in
many solid-pseudopapillary neoplasms, will help dis-
tinguish the two when papillary architecture is
absent. component, given that these neoplasms tend to be very
Pseudocyst enters the differential diagnosis when heterogeneous in their lining and that an invasive com-
only the hemorrhagic and necrotic cyst debris is aspi- ponent may not be apparent from gross inspection of
rated. Clinical correlation and cyst fluid analysis helps the resected cyst. Complete resection of thoroughly
distinguish these entities. Pseudocysts are more common evaluated, non-invasive neoplasms is considered
in men with a history of pancreatitis. Unlike pseudo- curative.
cysts, solid-pseudopapillary neoplasms have a low
amylase level on cyst fluid analysis.
CYTOPATHOLOGIC FEATURES

MUCINOUS CYSTIC NEOPLASM Aspiration of MCNs produces highly variable amounts


of extracellular mucin and cyst-lining epithelium. In
Mucinous cystic neoplasm (MCN) is a neoplastic mucin- addition, the degree of epithelial atypia may be variable
producing cyst that, in almost all cases, does not com- and not representative of the highest degree of atypia
municate with the pancreatic ductal system, is lined by of the cyst, owing to the heterogeneity of the cyst lining.
mucinous epithelial cells with varying degrees of atypia, As such, the cytologic diagnosis often underestimates
and contains subepithelial ovarian-type stroma. the ultimate histologic grade of the tumor. In addition,
a specific diagnosis of MCN is less common than a more
general diagnosis of neoplastic mucinous cyst, a generic
term that encompasses both intraductal papillary muci-
CLINICAL FEATURES nous neoplasm (IPMN) and MCN. This is primarily
due to a lack of architectural specificity of the glandular
epithelium.
Mucinous cystic neoplasms comprise approximately The presence of a neoplastic mucinous cyst is often
6% of all primary pancreatic tumors. With rare excep- first suspected at the time of aspiration when thick,
tion, patients are female, and the average age is between viscous mucus is grossly appreciated by the aspirator.
40 and 50 years. Mucinous cystic neoplasms with inva- This mucus is difficult to draw into and express from
sive carcinoma often occur in patients in their 60s. the needle. Such thick and viscous cyst fluid is reflected
Complete surgical resection is the treatment of choice. on the slide as a thick sheet of ‘colloid-like’ mucin that
Regardless of the atypia of the lining epithelium, the frequently covers most of the slide (Fig. 9-43). Mucin
prognosis is directly related to the presence or absence contamination from the gastrointestinal tract will not
of an invasive carcinoma. As such, cytologic evaluation be ‘colloid-like’. Degenerated inflammatory cells and
cannot predict outcome. Complete and thorough his- histiocytes within the mucin is a feature that also helps
tologic sampling is essential to rule out an invasive to distinguish contaminating mucin from neoplastic

Ch009-F06731.indd 285 10/26/2006 10:32:09 AM


286 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-43
Mucinous cystic neoplasm (MCN).
Colloid-like mucin is typical of a
neoplastic mucinous cyst, including
MCN and intraductal papillary muci-
nous neoplasm. This quality of
mucin is not seen from gastrointes-
tinal contamination. Papanicolaou
stain, medium power.

mucin. However, not all MCNs will have such mucoid


MUCINOUS CYSTIC NEOPLASM – PATHOLOGIC FEATURES contents. Extracellular mucin can appear as focally
thick clumps, thin wisps, and as focal or diffuse thin
Cytopathologic Findings background mucin, not easily visualized on routine
៉ Mucin quality and quantity can vary: thick, ‘colloid-like’ mucin preparations, especially liquid-based preparations where
is consistent with origin from the cyst (rather than the mucin can be quite attenuated during processing
gastrointestinal contamination from EUS biopsy) (Fig. 9-44). An aliquot of the cyst fluid, if sufficient in
៉ Epithelial cells vary in quantity and may be absent in high- quantity, can be used to prepare cytospin slides for
grade neoplasms with only mucin on smears
mucicarmine and/or Alcian blue pH 2.5 to assess for
៉ Epithelial cells range from fl at sheets and single bland columnar
mucinous cells (adenoma) to high-grade dysplastic cells with a
mucin. Negative special stains, however, do not exclude
high N/C ratio and irregular nuclei with or without cytoplasmic the presence of a MCN.
mucin Mucinous cystadenomas are typically scantily cellular,
aspirates producing single cells, small clusters, and flat
Ancillary Studies sheets of bland glandular epithelial cells that typically
៉ Histochemical stains: mucicarmine and Alcian blue pH 2.5 +/− demonstrate cytoplasmic mucin visible on routine light
៉ Immunocytochemistry (+): cytokeratins AE1/AE3, CAM 5.2, CK7, microscopy (Fig. 9-45). The nuclei are basally located,
CK8, CK18, CK19; CEA, DUPAN-2, CA 19-9, and MUC5AC round and regular, with even chromatin and inconspicu-
៉ Cyst fluid analysis: low amylase and variable CEA (usually ous to occasionally prominent nucleoli. Occasionally,
>200 ng/mL) muciphages (foamy histiocytes) may be the only cells in
the cyst contents. The epithelial lining of MCNs with
Differential Diagnosis and Pitfalls moderate dysplasia and higher-grade neoplasms demon-
៉ Pseudocyst strates epithelial cells with nuclear crowding, loss of
៉ Side-branch IPMN polarity, nuclear elongation or rounding, hyperchroma-
៉ Serous cystadenoma sia, and increased N/C ratio (Fig. 9-46). Cells in small
៉ Gastrointestinal contamination (pitfall)
tight bud-like clusters or singly with these same nuclear
features, and cytoplasm with or without mucin is also
consistent with the presence of a MCN with moderate
dysplasia or higher. Crowded groups of cells with open
chromatin, irregular nuclear membranes and nucleoli,
significant background inflammation, and necrosis
support the interpretation of malignancy (Fig. 9-47).

Ch009-F06731.indd 286 10/26/2006 10:32:09 AM


CHAPTER 9 Pancreas 287

FIGURE 9-44
Mucinous cystic neoplasm. Mucin
can appear fragmented and quite
attenuated in liquid-based prepara-
tions, making recognition difficult.
ThinPrep®, Papanicolaou stain,
medium power.

FIGURE 9-45
Mucinous cystic neoplasm-adenoma.
Bland adenomatous epithelium
maintains a uniform honeycomb
arrangement and single columnar
cells demonstrate diffuse cytoplas-
mic mucin. ThinPrep®, Papanicolaou
stain, high power.

Ch009-F06731.indd 287 10/26/2006 10:32:09 AM


288 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-46
Mucinous cystic neoplasm with
moderate dysplasia. Increased
nuclear density, nuclear crowding,
loss of polarity, and increased N/C
ratio that may be associated with
a loss of cytoplasmic mucin and
cellular degeneration are features
that correlate with a neoplasm of at
least moderate dysplasia. Papanico-
laou stain, high power.

FIGURE 9-47
Mucinous cystadenocarcinoma.
Crowded three-dimensional groups
containing cells with either hyper-
chromasia or open chromatin, irreg-
ular nuclear membranes, and
nucleoli in a background with sig-
nificant infl ammation and necrosis
support the interpretation of malig-
nancy. Invasion, however, can only
be determined from aspiration of an
associated mural nodule. Papanico-
laou stain, low power.

Ch009-F06731.indd 288 10/26/2006 10:32:10 AM


CHAPTER 9 Pancreas 289

FIGURE 9-48
Mucinous cystic neoplasm. Thin
background mucin may not be
appreciated on routine stains.
Special stains for mucin, including
this Alcian blue at pH 2.5, can high-
light extracellular and intracellular
mucin. Alcian blue pH 2.5, medium
power.

Invasion can be determined only by the aspiration of a by the presence of large, uniform, flat sheets of
mural nodule, as the cytologic features distinguishing glandular epithelium with a brush-bordered luminal
in-situ from invasive carcinoma have not been definitely edge studded with goblet cells containing clear
established. cytoplasm (Fig. 9-49), gastric epithelium more often
presents as small groups and occasionally as single
columnar glandular epithelial cells that can be con-
ANCILLARY STUDIES fused with the bland epithelium of an adenoma (Fig.
9-50). A cup-like apical pocket of mucinous cytoplasm
seen in the foveolar cells of the gastric epithelium is a
Aspirates producing thin white fluid, not grossly feature that can help differentiate it from the cyst lining
mucoid, may benefit from special stains for mucin on of a MCN adenoma that demonstrates a full columnar
cystospin preparations, as mentioned above (Fig. 9-48). column of mucinous epithelium (see Fig. 9-45).
The epithelial cells of MCN stain with antibodies to The differential diagnosis of MCN also includes
cytokeratins (pancytokeratin, CAM 5.2, CK7, CK8, IPMN, pseudocyst, oligocystic and unilocular variants
CK18, and CK19), DUPAN-2, CA 19-9, and MUC5AC. of serous cystadenoma, and cystic PEN. Mucinous cystic
Almost all MCNs are negative for CK20, MUC1, and neoplasms devoid of identifiable mucin or epithelial
MUC6. MUC2 will stain goblet cells. cells can be mistaken for a pseudocyst. Oligocystic, mac-
Chemical analysis of the cyst fluid can aid in the diag- rocystic, and unilocular variants of serous cystadenoma
nosis and classification of pancreatic cysts. CEA levels radiologically mimic MCN. The presence of thick,
above 200 ng/mL have been found to aid in the distinc- viscous mucin excludes the diagnosis of a serous cyst
tion of non-mucinous from mucinous cysts, with very and pseudocyst. Serous cysts produce thin, clear, non-
high levels of CEA correlating with malignancy. Amylase viscous fluid and scant epithelium. The epithelial cells
levels are not high in MCN as in IPMN, due to the are bland cuboidal cells with non-mucinous cytoplasm.
absence of connectivity with the pancreatic duct. In the absence of background mucin, the small, often
individual cells of a MCN with moderate dysplasia or
carcinoma are similar to the cells of cystic PEN. The
DIFFERENTIAL DIAGNOSIS AND PITFALLS most helpful distinguishing features are those of the
nucleus. In cystic PEN, the nuclei are round with rela-
tively smooth nuclear membranes and have chromatin
Distinction of mucinous cystadenomas from con- that is coarse and stippled in the typical ‘salt and pepper’
taminating gastrointestinal epithelium is particularly neuroendocrine pattern. The presence of even focal
challenging, especially gastric epithelium. In con- intracytoplasmic vacuoles also supports a diagnosis of
trast to duodenal epithelium, which is recognized MCN over cystic PEN.

Ch009-F06731.indd 289 10/26/2006 10:32:11 AM


290 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-49
Duodenal epithelium. During EUS-
guided biopsies of lesions in the
pancreatic head, duodenal epithe-
lium may contaminate the speci-
men. The epithelium is often
recognized by the presence of large
monolayered sheets with a uniform
luminal edge of columnar cells with
a brush border. Goblet cells fre-
quently stud the sheets of epithe-
lium but may not be overtly apparent
in all groups, especially small
groups. Papanicolaou stain, medium
power.

FIGURE 9-50
Gastric epithelium. EUS-guided
biopsies of neoplasms or lesions in
the pancreatic body and tail tra-
verse the gastric wall. Gastric epi-
thelium may demonstrate mucinous
cytoplasm which can be confused
with mucin-producing neoplastic
cysts. The mucinous cytoplasm is
often present in an apical cup-like
compartment. Papanicolaou stain,
high power.

Ch009-F06731.indd 290 10/26/2006 10:32:12 AM


CHAPTER 9 Pancreas 291

INTRADUCTAL PAPILLARY MUCINOUS varies by study and institution, but most studies show
NEOPLASM a slight male predominance.
Complete surgical resection is currently the treat-
ment of choice. Prognosis is directly related to the pres-
Intraductal papillary mucinous neoplasm (IPMN) is a ence or absence of an invasive carcinoma. Complete and
mucin-producing neoplastic cyst that arises from and thorough histologic sampling is essential to rule out an
is directly connected with the pancreatic ductal system, invasive component, given that these neoplasms tend to
either the main duct and/or side-branch duct, and is be very heterogenous in their lining and that an invasive
lined by typically papillary and variably atypical muci- component may not be apparent from gross inspection
nous epithelium. These neoplasms are classified by the of the resected cyst. Non-invasive IPMNs have a greater
WHO as IPMN-adenoma, IPMN with moderate dyspla- than 90% 5-year survival rate. This rate drops to 40%
sia (also know as borderline tumor), IPMN with carci- in IPMN with invasive carcinoma, but this rate is still
noma in situ, and IPMN with invasive carcinoma significantly better than for conventional ductal adeno-
(either tubular type or colloid type). carcinoma. The prognosis also depends on the type of
IPMN and the type and size of the invasive component.
Side-branch IPMN is reported to behave better than
main duct and combined-type IPMN, and the invasive
CLINICAL FEATURES IPMN with colloid carcinoma is reported to have a
better prognosis than one with invasive conventional
It is difficult to establish an accurate incidence of IPMN tubular type carcinoma.
owing to its relatively recent distinction as a specific
entity separate from mucinous cystic neoplasms in the
mid 1980s and from serous cysts in the 1970s. In addi- CYTOPATHOLOGIC FEATURES
tion, many small asymptomatic neoplasms are being
incidentally recognized because of the increasing use
of CT scans for other conditions. An estimated inci- The gross features of the cyst fluid of IPMNs are identi-
dence from recent reports places IPMN at approxi- cal to those of MCNs and the same discorrelation is
mately 20% of all neoplastic pancreatic cysts and 5% present between the quality and quantity of mucin and
of all pancreatic neoplasms. cells on FNAB with the final grade of the tumor on
Most IPMNs occur in the elderly population, with a histology.
peak age of close to 65 years. The male to female ratio

INTRADUCTAL PAPILLARY MUCINOUS NEOPLASM –


PATHOLOGIC FEATURES
INTRADUCTAL PAPILLARY MUCINOUS NEOPLASM –
DISEASE FACT SHEET Cytopathologic Findings
៉ Mucin quality and quantity can vary: thick, ‘colloid-like’ mucin
Incidence
is consistent with origin from the cyst (rather than
៉ ∼5% of all pancreatic neoplasms and ∼20% of all neoplastic gastrointestinal contamination from EUS biopsy)
pancreatic cysts ៉ Epithelial cells vary in quantity and may be absent even in
high-grade neoplasms
Gender and Age Distribution ៉ Epithelial cells range from fl at sheets to papillary clusters and
៉ M > F single cells with bland columnar mucinous cells (adenoma) to
៉ Mean age ∼65 years high-grade dysplastic cells singly or in tight epithelial clusters
with a high N/C ratio and irregular nuclei with or without
Radiologic Features cytoplasmic mucin; abundant background infl ammation suggests
៉ Main duct, side-branch, and combined types exist at least moderate dysplasia, and necrosis at least carcinoma in
៉ Copious ductal mucin produces filling defects situ
៉ Cysts are single or multiple and connect to the large pancreatic
ducts Ancillary Studies
៉ 70% occur in the pancreatic head ៉ Histochemical stains: mucicarmine and Alcian blue pH 2.5 +/−
៉ Cyst wall nodules and markedly dilated main pancreatic duct ៉ Immunocytochemical stains (+): cytokeratins AE1/AE3, CAM 5.2,
suggest malignancy CK7, CK8, CK18, CK19; CEA, CA 19-9, and MUC5AC
៉ Cyst fluid analysis: high amylase and variable CEA (usually
Prognosis and Treatment >200 ng/mL)
៉ Prognosis is directly related to the presence or absence of
invasive carcinoma Differential Diagnosis and Pitfalls
៉ Surgical resection is the treatment of choice and is curative for ៉ Pseudocyst
non-invasive neoplasms ៉ Mucinous cystic neoplasm
៉ Invasive side-branch IPMN has a better prognosis than main duct ៉ Serous cystadenoma
IPMN ៉ Gastrointestinal contamination (pitfall)

Ch009-F06731.indd 291 10/26/2006 10:32:13 AM


292 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-51
Intraductal papillary mucinous neo-
plasm-adenoma. The bland muci-
nous epithelium is appreciated by
the low N/C ratio, round, regular
nuclei, and abundant columnar
cytoplasm that may or may not
appear visibly mucinous. Papanico-
laou stain, high power.

IPMN-adenoma is lined by bland columnar mucinous ANCILLARY STUDIES


glandular cells arranged in small clusters and flat to
folded sheets with a honeycombed pattern. Single cells
may also be seen. Mucinous papillary epithelial frag- Aspirates producing thin white fluid, not grossly
ments are not often appreciated in adenomas. Columnar mucoid, may benefit from special stains for mucin on
cells with round basal nuclei and abundant columnar cystospin preparations, as mentioned above. Negative
cytoplasm that may or may not appear visibly mucinous mucin stains, however, do not exclude the diagnosis of
(Fig. 9-51). Dense, oncocytic cytoplasm is consistent IPMN. The epithelial cells of IPMN label with anti-
with intraductal oncocytic papillary neoplasm (Fig. 9- bodies to cytokeratins (AE1/AE3, CAM 5.2, CK7, CK8,
52). Other epithelial types (intestinal, biliary, and gastric CK18, and CK19; occasionally with CK20), CEA, and
foveolar) are not readily distinguished on cytology. CA 19-9. Very few IPMNs stain for MUC1 and almost
IPMN-moderate dysplasia and IPMN-carcinoma in situ none stain for MUC7, but most stain for MUC2, MUC3,
are lined by atypical to malignant-appearing glandular MUC4, and MUC5AC. Staining patterns for the MUC
epithelium. The cells display nuclear crowding, loss of proteins have been correlated with the type of lining
polarity, nuclear elongation, and hyperchromasia. epithelium of the villi and the presence or absence of
Hyperchromatic cells with irregular nuclear membranes an invasive component on histology.
and a high N/C ratio may be arranged in papillary clus- Like in MCN, CEA levels above 200 ng/mL support
ters, where the length is usually twice the width of a mucinous cyst, with very high levels of CEA correlat-
the group (Fig. 9-53), small tight epithelial cells clusters ing with malignancy. Amylase levels are high in IPMN
(Fig. 9-54), or singly (Fig. 9-55). Even if very scant in but not MCN, due to the connectivity of IPMN with the
amount, the presence of these groups is significant. Open pancreatic duct.
chromatin, irregular nuclear membranes and nucleoli,
significant background inflammation, and necrosis
support the interpretation of an in-situ or invasive car-
cinoma (Figs 9-56 & 9-57). Only necrosis appears to DIFFERENTIAL DIAGNOSIS AND PITFALLS
correlate with the presence of invasion, but this distinc-
tion cannot be made on aspirates of cyst contents alone.
A mural nodule is indicative of an invasive carcinoma, The differential diagnosis of IPMN is essentially the
and currently, aspiration of this nodule is necessary to same as for MCN and is discussed in the above
cytologically document an invasive carcinoma. section.

Ch009-F06731.indd 292 10/26/2006 10:32:13 AM


CHAPTER 9 Pancreas 293

FIGURE 9-52
Intraductal papillary mucinous
neoplasm, oncocytic variant. The
presence of an intraductal lesion
with abundant dense, granular,
oncocytic cytoplasm characterizes
this variant. Hema III stain, high
power.

FIGURE 9-53
Intraductal papillary mucinous
neoplasm (IPMN) with moderate
dysplasia. Papillary clusters of epi-
thelial cells with nuclear crowding,
atypia, and overlap characterizes an
IPMN with at least moderate dyspla-
sia. Papanicolaou stain, medium
power.

Ch009-F06731.indd 293 10/26/2006 10:32:14 AM


294 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 9-54
Intraductal papillary mucinous
neoplasm with moderate dysplasia.
Small clusters of high N/C ratio
cells with hyperchromatic nuclei
and with or without mucinous cyto-
plasm suggests the presence of a
neoplastic mucinous cyst with at
least moderate dysplasia. This
finding does not separate moderate
dysplasia from carcinoma in situ or
invasive carcinoma. Papanicolaou
stain, high power.

FIGURE 9-55
Intraductal papillary mucinous
neoplasm with carcinoma in situ.
Some neoplasms may demonstrate
only single dysplastic cells that
resemble severe dysplasia of the
cervix. Note the necrosis and
infl ammatory debris surrounding
this dysplastic cell. Papanicolaou
stain, high power.

Ch009-F06731.indd 294 10/26/2006 10:32:15 AM


CHAPTER 9 Pancreas 295

FIGURE 9-56
Intraductal papillary mucinous neo-
plasm with carcinoma in situ. The
hyperchromatic crowded group con-
tains cells with nuclear overlapping
and distinct parachromatin clear-
ing, features that are consistent
with carcinoma; however, they do
not distinguish in-situ from inva-
sive carcinoma. Papanicolaou stain,
high power.

FIGURE 9-57
Intraductal papillary mucinous
neoplasm with invasive carcinoma.
The presence of coagulative cellular
necrosis is the one feature that
correlates with invasive carcinoma.
Note the thick mucin in the right
of the image. Papanicolaou stain,
medium power.

Ch009-F06731.indd 295 10/26/2006 10:32:16 AM


296 FINE NEEDLE ASPIRATION CYTOLOGY

a 3-year experience and review of the literature. Cancer 2002;96:


NON-EPITHELIAL AND METASTATIC 362–369.
TUMORS
Pseudocyst

Primary non-epithelial neoplasms account for approxi- Brugge WR, Lewandrowski K, Lee-Lewandrowski E, et al. Diagnosis of
pancreatic cystic neoplasms: a report of the cooperative pancreatic cyst
mately 1–2% of all pancreatic neoplasms, and metasta- study. Gastroenterology 2004;126:1330–1336.
ses represent up to 4% of pancreatic tumors. Frossard JL, Amouyal P, Amouyal G, et al. Performance of endosonography-
The clinical behavior and cytologic features of neo- guided fine needle aspiration and biopsy in the diagnosis of pancreatic
plasms such as lymphangioma, schwannoma, lipoma, cystic lesions. Am J Gastroenterol 2003;98:1516–1524.
Kloppel G. Pseudocysts and other non-neoplastic cysts of the pancreas.
and (extra-) gastrointestinal stromal tumor are similar Semin Diagn Pathol 2000;17:7–15.
to those outside of the pancreas. Non-Hodgkin lym- Kloppel G, Maillet B. Pseudocysts in chronic pancreatitis: a morphological
phoma may be primary or secondary. Carcinomas, par- analysis of 57 resection specimens and 9 autopsy pancreata. Pancreas
ticularly from the lung, are the most common malignant 1991;6:266–274.
neoplasm metastatic to the pancreas. Other common Kosmahl M, Pauser U, Peters K, et al. Cystic neoplasms of the pancreas
and tumor-like lesions with cystic features: a review of 418 cases and
metastatic carcinomas include those originating in the a classification proposal. Virchows Arch 2004;445:168–178.
breast, kidney, stomach, and skin (melanoma). Lewandrowski K, Lee J, Southern J, Centeno B, Warshaw A. Cyst fluid
analysis in the differential diagnosis of pancreatic cysts: a new approach
to the preoperative assessment of pancreatic cystic lesions. AJR Am J
Roentgenol 1995;164:815–819.
Pinto M, Meriano F. Diagnosis of cystic pancreatic lesions by cytologic
Acknowledgement examination and carcinoembryonic antigen and amylase levels of cyst
contents. Acta Cytol 1991;35:456–463.

I would like to thank my co-authors on the 4th series


AFIP fascicle on Tumors of the Pancreas, Drs Ralph Lymphoepithelial Cyst
Hruban and David Klimstra, for providing a rich refer- Adsay NV, Hasteh F, Cheng JD, Klimstra DS. Squamous-lined cysts of the
ence on which I relied heavily to produce this chapter. pancreas: lymphoepithelial cysts, dermoid cysts (teratomas), and
accessory-splenic epidermoid cysts. Semin Diagn Pathol 2000;17:56–65.
Adsay NV, Hasteh F, Cheng JD, et al. Lymphoepithelial cysts of the pan-
creas: a report of 12 cases and a review of the literature. Mod Pathol
SUGGESTED READINGS 2002;15:492–501.
General Bolis GB, Farabi R, Liberati F, Maccio T. Lymphoepithelial cyst of the
pancreas. Report of a case diagnosed by fine needle aspiration biopsy.
Centeno BA, Pitman MB. Fine Needle Aspiration Biopsy of the Pancreas. Acta Cytol 1998;42:384–386.
Boston: Butterworth-Heinemann, 1999. Cappellari JO. Fine-needle aspiration cytology of a pancreatic lymphoepi-
DeMay R. Pancreas. In: The Art and Science of Cytopathology. Chicago: thelial cyst. Diagn Cytopathol 1993;9:77–81.
ASCP Press, 1996:1053–1082. Centeno BA, Stockwell JW, Lewandrowski KB. Cyst fluid cytology and
Geisinger KR, Stanley MW, Raab SS, Silverman JF, Abati A. Modern Cyto- chemical features in a case of lymphoepithelial cyst of the pancreas: a
pathology. Philadelphia: Churchill Livingstone, 2004. rare and difficult preoperative diagnosis. Diagn Cytopathol 1999;21:
Hruban RH, Pitman MB, Klimstra DS. Atlas of Tumor Pathology: Tumors of 328–330.
the Pancreas, 4th ed. Silver Spring, MD: ARP Press, In press. Liu J, Shin HJ, Rubenchik I, Lang E, Lahoti S, Staerkel GA. Cytologic
Klimstra DS. Pancreas. In: Sternberg SS, ed. Histology for Pathologists. features of lymphoepithelial cyst of the pancreas: two preoperatively
Philadelphia: Lippincott-Raven, 1997:613–647. diagnosed cases based on fine-needle aspiration. Diagn Cytopathol
Lack EE. Pathology of the Pancreas, Gallbladder, Extrahepatic Biliary Tract, 1999;21:346–350.
and Ampullary Region. New York: Oxford University Press, 2003. Mitchell ML. Fine needle aspiration biopsy of peripancreatic lymphoepi-
thelial cysts. Acta Cytol 1990;34:462–463.

Chronic Pancreatitis
Ductal Adenocarcinoma
Bentz JS, Kochman ML, Faigel DO, Ginsberg GG, Smith DB, Gupta PK.
Endoscopic ultrasound-guided real-time fine-needle aspiration: clinico- Al-Kaisi N, Siegler EE. Fine needle aspiration cytology of the pancreas.
pathologic features of 60 patients. Diagn Cytopathol 1998;18:98– Acta Cytol 1989;33:145–152.
109. Cohen MB, Egerter DP, Holly EA, Ahn DK, Miller TR. Pancreatic adenocar-
Deshpande V, Mino-Kenudson M, Pitman MB, Lauwers GY. Endoscopic cinoma: regression analysis to identify improved cytologic criteria.
ultrasound guided fine needle aspiration biopsy of autoimmune pan- Diagn Cytopathol 1991;7:341–345.
creatitis: diagnostic criteria and pitfalls. Am J Surg Pathol 2005;29: Fekete PS, Nunez C, Pitlik DA. Fine-needle aspiration biopsy of the pan-
1464–1471. creas: a study of 61 cases. Diagn Cytopathol 1986;2:301–306.
Fritscher-Ravens A, Brand L, Knofel WT, et al. Comparison of endoscopic Henke A, Jensen C, Cohen M. Cytologic diagnosis of adenocarcinoma in
ultrasound-guided fine needle aspiration for focal pancreatic lesions biliary and pancreatic duct brushings. Adv Anat Pathol 2002;9:
in patients with normal parenchyma and chronic pancreatitis [see 301–308.
comment]. Am J Gastroenterol 2002;97:2768–2775. Lin F, Staerkel GA. Cytologic criteria for well differentiated adenocarci-
Hollerbach S, Klamann A, Topalidis T, Schmiegel WH. Endoscopic ultraso- noma of the pancreas in fine-needle aspiration biopsy specimens.
nography (EUS) and fine-needle aspiration (FNA) cytology for diagnosis Cancer 2003;99:44–50.
of chronic pancreatitis. Endoscopy 2001;33:824–831. Mitchell ML, Carney CN. Cytologic criteria for the diagnosis of pancreatic
Stelow EB, Bardales RH, Lai R, et al. The cytological spectrum of chronic carcinoma. Am J Clin Pathol 1985;83:171–176.
pancreatitis. Diagn Cytopathol 2005;32:65–69. Nagle J, Wilbur DC, Pitman MD. Cytomorphology of gastric and duodenal
Voss M, Hammel P, Molas G, et al. Value of endoscopic ultrasound guided epithelium and reactivity to B72.3: a baseline for comparison to pan-
fine needle aspiration biopsy in the diagnosis of solid pancreatic creatic neoplasms aspirated by EUS-FNAB. Diagn Cytopathol 2005;33:
masses. Gut 2000;46:244–249. 381–386.
Ylagan LR, Edmundowicz S, Kasal K, Walsh D, Lu DW. Endoscopic ultra- Robins DB, Katz RL, Evans DB, Atkinson EN, Green L. Fine needle aspira-
sound guided fine-needle aspiration cytology of pancreatic carcinoma: tion of the pancreas. In quest of accuracy. Acta Cytol 1995;39:1–10.

Ch009-F06731.indd 296 10/26/2006 10:32:17 AM


CHAPTER 9 Pancreas 297

Van Heek T, Rader A, Offerhaus G. K-ras, p53, and DPC4 (MAD4) alterations Klimstra DS. Pancreas. In: Sternberg SS, ed. Histology for Pathologists.
in fine-needle aspirates of the pancreas: a molecular panel correlates Philadelphia: Lippincott-Raven, 1997:613–647.
with and supplements cytologic diagnosis. Am J Surg Oncol 2002;117: Klimstra DS, Heffess CS, Oertel JE, Rosai J. Acinar cell carcinoma of the
755–765. pancreas. A clinicopathologic study of 28 cases. Am J Surg Pathol
1992;16:815–837.
Labate AM, Klimstra DL, Zakowski MF. Comparative cytologic features of
Ductal Adenocarcinoma Variants pancreatic acinar cell carcinoma and islet cell tumor. Diagn Cytopathol
Adsay NV, Pierson C, Sarkar F, et al. Colloid (mucinous noncystic) carci- 1997;16:112–116.
noma of the pancreas. Am J Surg Pathol 2001;25:26–42. Samuel LH, Frierson HJ. Fine needle aspiration cytology of acinar cell
Chow LT, Chow WH. Signet-ring mucinous adenocarcinoma of the pan- carcinoma of the pancreas: a report of two cases. Acta Cytol 1996;
creas. Chin Med Sci J 1994;9:176–178. 40:585–591.
Gupta RK, Wakefield SJ. Needle aspiration cytology, immunocytochemis- Villanueva RR, Nguyen-Ho P, Nguyen GK. Needle aspiration cytology of
try, and electron microscopic study of unusual pancreatic carcinoma acinar-cell carcinoma of the pancreas: report of a case with diagnostic
with pleomorphic giant cells. Diagn Cytopathol 1992;8:522–527. pitfalls and unusual ultrastructural findings. Diagn Cytopathol 1994;10:
Manci EA, Gardner LL, Pollock WJ, Dowling EA. Osteoclastic giant cell 362–364.
tumor of the pancreas. Aspiration cytology, light microscopy, and
ultrastructure with review of the literature. Diagn Cytopathol 1985;1: Pancreatoblastoma
105–110.
Mullick SS, Mody DR. ‘Osteoclastic’ giant cell carcinoma of the pancreas. Dhebri A, Connor S, Campbell F, Ghaneh P, Sutton R, Neoptolemos J.
Report of a case with aspiration cytology. Acta Cytol 1996;40:975– Diagnosis, treatment and outcome of pancreatoblastoma. Pancreatology
979. 2004;4:441–453.
Paal E, Thompson LD, Frommelt RA, Przygodzki RM, Heffess CS. A clinico- Henke AC, Kelley CM, Jensen CS, Timmerman TG. Fine-needle aspira-
pathologic and immunohistochemical study of 35 anaplastic carcinomas tion cytology of pancreatoblastoma. Diagn Cytopathol 2001;25:118–
of the pancreas with a review of the literature. Ann Diagn Pathol 121.
2001;5:129–140. Klimstra D, Longnecker D. Pancreatoblastoma. In: Hamilton S, Aaltonen
Rahemtullah A, Misdraji J, Pitman MB. Adenosquamous carcinoma of the L, eds. World Health Organization Classification of Tumours. Pathology
pancreas: cytologic features in 14 cases. Cancer 2003;99:372–378. and Genetics of Tumours of the Digestive System. Lyon, France: IARC
Silverman JF, Dabbs DJ, Finley JL, Geisinger KR. Fine-needle aspiration Press, 2000:244–245.
biopsy of pleomorphic (giant cell) carcinoma of the pancreas. Cytologic, Klimstra DS, Wenig BM, Adair CF, Heffess CS. Pancreatoblastoma. A clini-
immunocytochemical, and ultrastructural findings. Am J Clin Pathol copathologic study and review of the literature. Am J Surg Pathol
1988;89:714–720. 1995;19:371–389.
Silverman JF, Finley JL, Berns L, Unverferth M. Significance of giant cells Pitman MB, Faquin WC. The fine-needle aspiration biopsy cytology of
in fine-needle aspiration biopsies of benign and malignant lesions of pancreatoblastoma. Diagn Cytopathol 2004;31:402–406.
the pancreas. Diagn Cytopathol 1989;5:388–391. Silverman JF, Holbrook CT, Pories WJ, Kodroff MB, Joshi VV. Fine needle
Tracey KJ, O’Brien MJ, Williams LF, et al. Signet ring carcinoma of the aspiration cytology of pancreatoblastoma with immunocytochemical
pancreas, a rare variant with very high CEA values. Immunohistologic and ultrastructural studies. Acta Cytol 1990;34:632–640.
comparison with adenocarcinoma. Dig Dis Sci 1984;29:573–576.
Walts AE. Osteoclast-type giant-cell tumor of the pancreas. Acta Cytol
1983;27:500–504. Serous Cystadenoma
Hittmair A, Pernthaler H, Totsch M, Schmid KW. Preoperative fine needle
Pancreatic Endocrine Neoplasm aspiration cytology of a microcystic adenoma of the pancreas. Acta
Cytol 1991;35:546–548.
Al-Kaisi N, Weaver MG, Abdul KFW, Siegler E. Fine needle aspiration cytol- Lal A, Bourtsos EP, DeFrias DV, Nemcek AA, Nayar R. Microcystic adenoma
ogy of neuroendocrine tumors of the pancreas. A cytologic immuno- of the pancreas: clinical, radiologic, and cytologic features. Cancer
cytochemical and electron microscopic study. Acta Cytol 1992;36: 2004;102:288–294.
655–660. Lewandrowski K, Lee J, Southern J, Centeno B, Warshaw A. Cyst fluid
Bell DA. Cytologic features of islet-cell tumors. Acta Cytol 1987;31: analysis in the differential diagnosis of pancreatic cysts: a new approach
485–492. to the preoperative assessment of pancreatic cystic lesions. AJR Am J
Collins BT, Cramer HM. Fine-needle aspiration cytology of islet cell tumors. Roentgenol 1995;164:815–819.
Diagn Cytopathol 1996;15:37–45. Logrono R, Vyas SH, Molina CP, Waxman I. Microcytic adenoma of the pan-
Dodd LG, Evans DB, Symmans F, Katz RL. Fine-needle aspiration of pan- creas: cytologic appearance on percutaneous and endoscopic ultrasound-
creatic extramedullary plasmacytoma: possible confusion with islet cell guided fine-needle aspiration. Report of a case. Diagn Cytopathol
tumor. Diagn Cytopathol 1994;10:371–375. 1999;20:298–301.
Labate AM, Klimstra DL, Zakowski MF. Comparative cytologic features of Nguyen GK, Vogelsang PJ. Microcystic adenoma of the pancreas. A report
pancreatic acinar cell carcinoma and islet cell tumor. Diagn Cytopathol of two cases with fine needle aspiration cytology and differential
1997;16:112–116. diagnosis. Acta Cytol 1993;37:908–912.
Ligneau B, Lombard-Bohas C, Partensky C. Cystic endocrine tumors of the
pancreas: clinical, radiologic and histopathologic features in 13 cases.
Am J Surg Pathol 2001;25:752–760. Solid-Pseudopapillary Neoplasm
Shaw JA, Vance RP, Geisinger KR, Marshall RB. Islet cell neoplasms. A Abraham SC, Klimstra DS, Wilentz RE, et al. Solid-pseudopapillary tumors
fine-needle aspiration cytology study with immunocytochemical corre- of the pancreas are genetically distinct from pancreatic ductal adeno-
lations. Am J Clin Pathol 1990;94:142–149. carcinomas and almost always harbor beta-catenin mutations. Am J
Sneige N, Ordonez NG, Veanattukalathil S, Samaan NA. Fine-needle aspira- Pathol 2002;160:1361–1369.
tion cytology in pancreatic endocrine tumors. Diagn Cytopathol Brugge WR, Lauwers GY, Sahani D, Fernandez-del Castillo C, Warshaw AL.
1987;3:35–40. Erratum in: Diagn Cytopathol 1987;3:176. Cystic neoplasms of the pancreas. N Engl J Med 2004;351:1218–
Sun W, Sneige N, Staerkel G. Comparison of endoscopic ultrasound-guided 1226.
and computed tomography-guided fine needle aspiration biopsy in the Greenberg ML, Rennie Y, Grierson JM, Quin JW, Boadle RA. Solid and
diagnosis of islet cell tumor of pancreas. Mod Pathol 2004;17(suppl. papillary epithelial tumour of the pancreas: cytological case study with
1):82A. ultrastructural and flow cytometric evaluation. Diagn Cytopathol
1993;9:541–546.
Acinar Cell Carcinoma Pelosi G, Iannucci A, Zamboni G, Bresaola E, Iacono C, Serio G. Solid and
cystic papillary neoplasm of the pancreas: a clinico-cytopathologic and
Caruso RA, Inferrera A, Tuccari G, Barresi G. Acinar cell carcinoma of the immunocytochemical study of five new cases diagnosed by fine-needle
pancreas. A histologic, immunocytochemical and ultrastructural study. aspiration cytology and a review of the literature. Diagn Cytopathol
Histol Histopathol 1994;9:53–58. 1995;13:233–246.

Ch009-F06731.indd 297 10/26/2006 10:32:17 AM


298 FINE NEEDLE ASPIRATION CYTOLOGY

Pettinato G, Di Vizio D, Manivel JC, Pambuccian SE, Somma P, Insabato Hruban RH, Takaori K, Klimstra DS, et al. An illustrated consensus on the
L. Solid-pseudopapillary tumor of the pancreas: a neoplasm with dis- classification of pancreatic intraepithelial neoplasia and intraductal
tinct and highly characteristic cytological features. Diagn Cytopathol papillary mucinous neoplasms. Am J Surg Pathol 2004;28:977–987.
2002;27:325–334. Jhala NC, Jhala D, Eltoum I, et al. Endoscopic ultrasound-guided fine
Pettinato G, Manivel JC, Ravetto C, et al. Papillary cystic tumor of needle aspiration biopsy: a powerful tool to obtain samples from small
the pancreas. A clinicopathologic study of 20 cases with cytologic, lesions. Cancer (Cancer Cytopathol) 2004;102:239–246.
immunohistochemical, ultrastructural, and flow cytometric observa- Klapman JB, Logrono R, Dye CE, Waxman I. Clinical impact of on-site
tions, and a review of the literature. Am J Clin Pathol 1992;98:478– cytopathology interpretation on endoscopic ultrasound-guided fine
488. needle aspiration. Am J Gastroenterol 2003;98:1289–1294.
Layfield LJ, Cramer H. Fine-needle aspiration cytology of intraductal
papillary-mucinous tumors: a retrospective analysis. Diagn Cytopathol
2005;32:16–20.
Mucinous Cystic Neoplasms Maire F, Couvelard A, Hammel P, et al. Intraductal papillary mucinous
Brugge WR. Role of endoscopic ultrasound in the diagnosis of cystic tumors of the pancreas: the preoperative value of cytologic and histo-
lesions of the pancreas. Pancreatology 2001;1:637–640. pathologic diagnosis. Gastrointest Endosc 2003;58:701–706.
Jhala NC, Jhala D, Eltoum I, et al. Endoscopic ultrasound-guided fine Michaels PJ, Brachtel EF, Bounds BC, Brugge WR, Pitman MB. Intraductal
needle aspiration biopsy: a powerful tool to obtain samples from small papillary mucinous neoplasm of the pancreas: cytologic features predict
lesions. Cancer (Cancer Cytopathol) 2004;102:239–246. histologic grade. Cancer (Cancer Cytopathology) 2006;108:163–173.
Klapman JB, Logrono R, Dye CE, Waxman I. Clinical impact of on-site Recine M, Kaw M, Evans DB, Krishnamurthy S. Fine-needle aspiration
cytopathology interpretation on endoscopic ultrasound-guided fine cytology of mucinous tumors of the pancreas. Cancer 2004;102:
needle aspiration. Am J Gastroenterol 2003;98:1289–1294. 92–99.
Lau SK, Lewandrowski KB, Brugge WR, Warshaw AL, Centeno BA. Diagnos- Shin HJ, Lahoti S, Sneige N. Endoscopic ultrasound-guided fine-needle
tic significance of mucin in fine needle aspiration samples of pancreatic aspiration in 179 cases: the M. D. Anderson Cancer Center experience.
cysts. Mod Pathol 2000;13:48A. Cancer 2002;96:174–180.
Lewandrowski KB, Southern JF, Pins MR, Compton CC, Warshaw AL. Cyst Stelow EB, Stanley MW, Bardales RH, et al. Intraductal papillary-mucinous
fluid analysis in the differential diagnosis of pancreatic cysts: a neoplasm of the pancreas. The findings and limitations of cytologic
comparison of pseudocysts, serous cystadenomas, mucinous cystic samples obtained by endoscopic ultrasound-guided fine needle aspira-
neoplasms, and mucinous cystadenocarcinoma. Ann Surg 1993;217:41– tion. Am J Clin Pathol 2003;120:398–404.
47.
Mallery JS, Centeno BA, Hahn PF, Chang Y, Warshaw AL, Brugge WR.
Pancreatic tissue sampling guided by EUS, CT/US, and surgery: a com- Non-Epithelial and Secondary Neoplasms
parison of sensitivity and specificity. Gastrointest Endosc 2002;56: Adsay NV, Andea A, Basturk O, Kilinc N, Nassar H, Cheng JD. Secondary
218–224. tumors of the pancreas: an analysis of a surgical and autopsy database
Nagle J, Wilbur DC, Pitman MD. The cytomorphology of gastric and duo- and review of the literature. Virchows Arch 2004;444:527–535.
denal epithelium and reactivity to B72.3: a baseline for comparison to Caffert L, Katz R, Ordonez N, Carrasco C, Cabanillas F. Fine needle aspira-
pancreatic neoplasms aspirated by EUS-FNAB. Diagn Cytopathol 2005;33: tion diagnosis of intra-abdominal and retroperitoneal lymphomas by a
381–386. morphologic and immunocytochemical approach. Cancer 1990;65:72–
Pinto M, Meriano F. Diagnosis of cystic pancreatic lesions by cytologic 77.
examination and carcinoembryonic antigen and amylase levels of cyst Dong HY, Harris NL, Preffer FI, Pitman MB. Fine-needle aspiration biopsy
contents. Acta Cytol 1991;35:456–463. in the diagnosis and classification of primary and recurrent lymphoma:
Recine M, Kaw M, Evans DB, Krishnamurthy S. Fine-needle aspiration a retrospective analysis of the utility of cytomorphology and flow
cytology of mucinous tumors of the pancreas. Cancer 2004;102: cytometry. Mod Pathol 2001;14:472–481.
92–99. Ferrozzi F, Zuccoli G, Bova D, Calculli L. Mesenchymal tumors of the pan-
Shin HJ, Lahoti S, Sneige N. Endoscopic ultrasound-guided fine-needle creas: CT findings. J Comput Assist Tomogr 2000;24:622–627.
aspiration in 179 cases: the M. D. Anderson Cancer Center experience. Paal E, Thompson LD, Heffess CS. A clinicopathologic and immunohisto-
Cancer 2002;96:174–180. chemical study of ten pancreatic lymphangiomas and a review of the
literature. Cancer 1998;82:2150–2158.
Wieczorek TJ, Faquin WC, Rubin BP, Cibas ES. Cytologic diagnosis of gastro-
Intraductal Papillary Mucinous Neoplasm intestinal stromal tumor with emphasis on the differential diagnosis
with leiomyosarcoma. Cancer (Cancer Cytopathol) 2001;93:276–287.
Faigel DO, Ginsberg GG, Bentz JS, Gupta PK, Smith DB, Kochman ML. Young NA, Al-Saleem TI, Ehya H, Smith MR. Utilization of fine-needle
Endoscopic ultrasound-guided real-time fine-needle aspiration biopsy aspiration cytology and flow cytometry in the diagnosis and subclassi-
of the pancreas in cancer patients with pancreatic lesions. J Clin Oncol fication of primary and recurrent lymphoma. Cancer 1998;84:252–
1997;15:1439–1443. 261.

Ch009-F06731.indd 298 10/26/2006 10:32:18 AM


10 Kidney and Adrenal Gland
Yener S Erozan

KIDNEY RENAL CELL CARCINOMA (RCC) – DISEASE FACT SHEET

Incidence
៉ About 3% of adult malignancies
Fine needle aspiration (FNA), performed under ultra-
៉ In USA, higher in black population
sound (US) or computed tomography (CT) guidance, is
a safe and accurate technique for the diagnosis of mass Gender and Age Distribution
lesions of the kidney (Table 10-1). It has been used in
៉ M : F = 1.5 : 1
the diagnosis of single and multiple masses, as well as ៉ Median age, 55 years
selected cystic lesions of kidney. The majority of single
masses in the kidney are primary renal neoplasms, Clinical Features
with renal cell carcinomas being the most common. ៉ Most confined to kidney for a long time and stay asymptomatic
Other single masses include urothelial carcinoma ៉ Many detected incidentally (on CT or US)
arising from kidney pelvis, benign neoplasms such as ៉ Hematuria, flank pain, or flank mass occurs in advanced stages
angiomyolipoma and oncocytoma, and tumor-like
lesions such as xanthogranulomatous pyelonephritis Genetic Abnormalities
and abscess. ៉ Multiple chromosomal abnormalities (specifically chromosome #3)
Childhood tumors of kidney are rarely sampled by ៉ Up to 55% of patients with von Hippel–Lindau syndrome develop
FNA. Exceptions are nephroblastoma (Wilms tumor) RCC
and rhabdoid tumor, which can be diagnosed by FNA ៉ Increased risk in patients with tuberculous sclerosis and acquired
and will be presented in this chapter. cystic kidney disease
Cystic lesions in kidney are common, and most are
benign, simple cysts (Table 10-2). Only those for which Prognosis
imaging studies suggest a neoplasm, such as complex ៉ Determined by stage, nuclear grading, and tumor type
multilocular cysts, are aspirated.
As in other FNAs, on-site evaluation of the speci-
mens for adequacy improves the diagnosis. Based on the
results, core biopsies can be performed or additional renal neoplasms, as well as for obtaining FNA samples
samples can be obtained for ancillary studies. and core biopsies from the lesions.
Most RCCs have multiple chromosomal abnormalities,
specifically involving chromosome 3. Up to 55% of
RENAL CELL CARCINOMA (RCC) patients with von Hippel–Lindau (VHL) syndrome
develop RCC. There is an increased risk of RCC in
patients with tuberous sclerosis and acquired cystic
CLINICAL FEATURES kidney disease, and an association between genetic alter-
ations and specific types of RCC has been documented.
Prognosis for RCC is determined by stage, nuclear
Renal cell carcinomas comprise about 3% of adult
grading, and type of the tumor.
malignancies and occur in older age groups (median
age, 55 years). They are more common in men than in
women (approximately 1.5 : 1), and in the United States
the incidence is higher in blacks than in whites. Heavy CLASSIFICATION
smoking and obesity increase the risk for RCC.
Most RCCs are confined to the kidney for a long
period of time, stay asymptomatic, and are detected inci- The current classification of RCC is shown in Table
dentally during imaging studies. When they become 10-3. The most common type of RCC is clear cell (about
symptomatic (i.e. hematuria, flank pain, or flank mass), 70%), followed by papillary (10–15%) and chromo-
they are usually in an advanced stage. Radiologic tech- phobe cell (approximately 5%). Collecting duct and
niques (CT and US) are very effective for detecting medullary types of RCCs are very rare.
299

Ch010-F06731.indd 299 10/26/2006 10:33:34 AM


300 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-1
Low-grade renal cell carcinoma
(grade 1). Uniform, small, round
nuclei with dense chromatin. No
visible nucleoli. Papanicolaou stain,
low power.

TABLE 10-1 TABLE 10-3


Mass Lesions of Kidney Classification of Renal Cell Carcinoma

• Neoplasms:
• Renal cell carcinoma
• Oncocytoma
• Nephroblastoma (Wilms tumor)
• Rhabdoid tumor
• Urothelial carcinoma
• Angiomyolipoma
• Lymphoma
• Metastatic neoplasms
• Non-neoplastic masses:
• Xanthogranulomatous pyelonephritis From Murphy WM, Grignon DJ, Perlman EJ. Tumors of the
• Abscess Kidney, Bladder, and Related Urinary Structures. Atlas of
Tumor Pathology, 4th Series, Fascicle 1. Washington, D.C:
Armed Forces Institute of Pathology, 2004.

TABLE 10-2
Cystic Lesions of Kidney cytologic specimens. Grade 1 RCCs have small, round
nuclei with dense chromatin and inconspicuous nucle-
• Benign cysts oli (Fig. 10-1). Grade 2 RCCs have somewhat larger,
• Malignant cystic lesions: round nuclei with finely granular chromatin and small
• Multilocular cystic clear cell carcinoma nucleoli which are not visible under low power (10×)
• Other renal cell carcinomas with cystic degeneration
(Fig. 10-2). Grade 3 RCCs have larger, round or oval
nuclei with a coarse granular chromatin pattern and
prominent nucleoli (Fig. 10-3). Grade 4 RCCs have
large pleomorphic nuclei with irregular nuclear borders
NUCLEAR GRADING and prominent nucleoli (Fig. 10-4). Correlation between
cytopathologic and histopathologic grading is moderate.
Accuracy increases when a two-grade system, ‘low
Nuclear grading (Fuhrman’s nuclear grading system) of grade’ (combined grades 1 and 2) and ‘high grade’ (com-
RCC has been shown to be a significant prognostic tool, bined grades 3 and 4), is used. When adequate cellular
and similar criteria are applicable to both histologic and samples are obtained, discrepancies between cytopa-

Ch010-F06731.indd 300 10/26/2006 10:33:35 AM


CHAPTER 10 Kidney and Adrenal Gland 301

FIGURE 10-2
Low-grade renal cell carcinoma
(grade 2). Uniform, somewhat larger
nuclei. No visible nucleoli. Diff-Quik
stain, low power.

FIGURE 10-3
High-grade renal cell carcinoma
(grade 3). Large, round to oval
nuclei, some with irregular borders.
Prominent nucleoli. Papanicolaou
stain, medium power.

thologic (FNA specimens) and histopathologic (resected CYTOPATHOLOGIC FEATURES


tumor) specimens are usually caused by sampling error
due to tumor heterogeneity. In our experience, inter-
observer concordance in FNA samples is higher The cytopathologic features of RCC presented in most
than that of cytopathologic and histopathologic correla- textbooks are based on those of clear cell carcinoma,
tion, supporting sampling error as the major reason for which is the most common type. There is also adequate
disconcordance. information about papillary RCC in the literature;

Ch010-F06731.indd 301 10/26/2006 10:33:35 AM


302 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-4
High-grade renal cell carcinoma
(grade 4). Large pleomorphic nuclei
with macronucleoli. Papanicolaou
stain, medium power.

specimens for cell blocks and/or needle core biopsies,


RENAL CELL CARCINOMA (RCC) – PATHOLOGIC FEATURES when needed, increases the specificity of the diagnosis.

Classification
៉ Histologic subtypes of RCC:
៉ Clear cell (conventional) (~70%) CLEAR CELL (CONVENTIONAL-TYPE)
៉ Papillary (10–15%) RENAL CELL CARCINOMA
៉ Chromophobe cell (~5%)
៉ Collecting duct (very rare)
៉ Medullary (very rare) CLINICAL FEATURES
Nuclear Grading (Fuhrman’s Grading System)
៉ Significant prognostic tool Clear cell is the most common subtype, comprising
៉ Similar criteria applicable to histologic and cytologic specimens about 70% of RCCs. Granular cell RCCs, which were
៉ Moderate correlation between histopathologic and considered a separate category previously, are now
cytopathologic grading included in this group. Both sporadic and familial forms
៉ Accuracy increases when a two-grade system is used: (VHL) have genetic abnormalities involving chromo-
៉ ‘Low grade’ (grades 1 and 2)
some 3. The VHL gene is located in 3p25, which is
៉ ‘High grade’ (grades 3 and 4)
mutated or lost in clear cell papillary RCCs. Additional
Cytopathologic Findings
suppressor genes are found in chromosome 3, most
commonly in the region of 3p14. Continuous deletion
៉ Vary according to type and nuclear grading (discussed in detail
under specific subtype)

Differential Diagnosis and Pitfalls


៉ Well-differentiated RCC vs other clear cell neoplasms CLEAR CELL (CONVENTIONAL-TYPE) RENAL CELL CARCINOMA –
៉ Poorly differentiated RCC vs metastatic neoplasm or urothelial DISEASE FACT SHEET
carcinoma
Incidence
៉ Most common type of RCC (70%)

Genetic Abnormalities
however, FNA findings of chromophobe, collecting duct,
៉ Both sporadic and familial forms (genetic abnormalities involving
and medullary RCCs are limited. Subtypes of RCC, as
chromosome #3)
well as Fuhrman nuclear grading, can be determined ៉ Continuous deletion from 3p24.2 to 3p25 in 96% of clear cell
fairly accurately in FNA specimens. Errors usually RCCs
occur due to lack of adequate sampling or to the poor
differentiation of the tumor. Obtaining adequate

Ch010-F06731.indd 302 10/26/2006 10:33:36 AM


CHAPTER 10 Kidney and Adrenal Gland 303

FIGURE 10-5
Renal cell carcinoma, clear cell (con-
ventional) type. Many large tissue
fragments of tumor with scattered
single cells or small groups. Diff-
Quik stain, low power.

FIGURE 10-6
Renal cell carcinoma, clear cell
(conventional) type. A large frag-
ment of tumor. Note the thin-walled
blood vessels traversing the tumor.
Diff-Quik stain, medium power.

from 3p24.2 to 3p25 is found in up to 96% of clear cell Some, however, are predominantly blood and require
carcinomas. multiple attempts to obtain adequate cellular material.
Thin-walled blood vessels crossing the fragments is a
characteristic feature (Fig. 10-6). Branching blood
CYTOPATHOLOGIC FEATURES vessels surrounded by several layers of tumor cells
form a papillary-like architecture (Fig. 10-7). Tumor
cells have clear or finely vacuolated cytoplasm (Fig.
Most FNAs yield abundant material comprised pre- 10-8) which contains lipids, cholesterol, and glycogen.
dominantly of tissue fragments of tumor (Fig. 10-5). Other cells may have acidophilic granular cytoplasm

Ch010-F06731.indd 303 10/26/2006 10:33:37 AM


304 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-7
Renal cell carcinoma, clear cell
(conventional) type. A large frag-
ment of tumor with papillary-like
formations. Diff-Quik stain, low
power.

FIGURE 10-8
Renal cell carcinoma, clear cell
(conventional) type. Tumor cells
with intracytoplasmic, small,
uniform vacuoles which are best
seen in air-dried Romanowsky-
stained specimens. These vacuoles
contain lipids and glycogen, which
could be shown by staining with
Oil-Red O and PAS stains. Diff-Quik
stain, high power.

(Fig. 10-9). Intracytoplasmic hyaline globules may be DIFFERENTIAL DIAGNOSIS AND PITFALLS
present; the nuclei of the tumor cells are usually round
with finely granular chromatin. Nucleoli may be absent,
small, or prominent, depending on the grade of the Clear cell RCCs have distinct cytologic features, and the
neoplasm. diagnosis is not difficult when the specimen is obtained
by FNA of a mass in the kidney. The only challenging
differential diagnosis might be for oncocytoma when
the tumor contains a large number of tumor cells with
ANCILLARY STUDIES granular eosinophilic cytoplasm rather than clear cells.
Oncocytomas lack large nucleoli and mitoses. Unlike
clear cell RCC, oncocytoma does not react to antibodies
Clear cell RCCs react to RCC and CD10 antibodies.
for vimentin and RCC.
They also react to antibodies to low molecular weight
cytokeratins, epithelial membrane antigen (EMA), and
vimentin.

Ch010-F06731.indd 304 10/26/2006 10:33:38 AM


CHAPTER 10 Kidney and Adrenal Gland 305

FIGURE 10-9
Renal cell carcinoma, clear cell
(conventional) type. A fragment of
high-grade RCC. Moderately pleo-
morphic nuclei with prominent
nucleoli. Most of the cells have
dense cytoplasm; a few have clear
cell features. Papanicolaou stain,
medium power.

CLEAR CELL (CONVENTIONAL-TYPE) RENAL CELL CARCINOMA – PAPILLARY RENAL CELL CARCINOMA – DISEASE FACT SHEET
PATHOLOGIC FEATURES
Incidence
Cytopathologic Findings ៉ 10–15% of RCCs
៉ Abundant material, predominantly tissue fragments in FNAs ៉ Multiple tumors more common than in clear cell RCC
៉ Thin-walled blood vessels traversing tissue fragments
៉ Branching blood vessels surrounded by tumor cells form
papillary-like architecture
៉ Finely vacuolated cytoplasm (lipids and glycogen)
៉ Other cells may have acidophilic granular cytoplasm
៉ Intracytoplasmic hyaline globules may be present
CYTOPATHOLOGIC FEATURES
៉ Nuclei usually round with finely granular chromatin
៉ Nucleoli absent, small or prominent, depending on grade
In type 1 papillary RCC, which comprises more than
two-thirds of papillary RCCs, fine needle aspirates
Ancillary Studies
usually show abundant papillary structures with vas-
៉ Reacts antibodies to RCC, CD10, low molecular weight
cular cores (Fig. 10-10). Single or lobulated globular
cytokeratins, EMA, and vimentin
forms with sharp outlines may also be present (Fig.
Differential Diagnosis and Pitfalls
10-11). Psammoma bodies (Fig. 10-12), lipid-laden
histiocytes, and intracytoplasmic hemosiderin pigment
៉ Could be confused with oncocytoma
៉ Oncocytomas lack large nucleoli and mitoses and do not react
in both histiocytes and tumor cells are characteristic
to antibodies for vimentin and RCC features of this tumor, although they are not always
present (Figs 10-13 & 10-14). Evidence of necrosis and
hemorrhage, which commonly occur in the tumor, may
also be seen in FNA specimens. In type 2 papillary
RCC, the cells have large acidophilic cytoplasm and a
higher nuclear grade (Grade 3) with prominent
PAPILLARY RENAL CELL CARCINOMA nuclei.

CLINICAL FEATURES
ANCILLARY STUDIES
Approximately 10–15% of RCCs are papillary type, and
multiple tumors occur more commonly than in clear Most papillary RCCs react to RCC markers, pancyto-
cell renal carcinoma. keratins, low molecular weight cytokeratins, and CD9

Ch010-F06731.indd 305 10/26/2006 10:33:40 AM


306 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-10
Renal cell carcinoma, papillary type.
Papillary structure with vascular
core. Diff-Quik stain, low power.

FIGURE 10-11
Renal cell carcinoma, papillary type.
Lobulated, globular formations in
a background of loosely attached
tumor cells with ill-defined cyto-
plasm. One rosette-like structure
with an early forming psammoma
body in the center. Papanicolaou
stain, medium power.

Ch010-F06731.indd 306 10/26/2006 10:33:41 AM


CHAPTER 10 Kidney and Adrenal Gland 307

FIGURE 10-12
Renal cell carcinoma, papillary type.
Small tissue fragments and loose
aggregates of tumor cells mixed
with macrophages. Several psam-
moma bodies surrounded with tumor
cells. Papanicolaou stain, medium
power.

FIGURE 10-13
Renal cell carcinoma, papillary type.
A tissue fragment of tumor. Tumor
appears to be high grade. Hemo-
siderin-laden macrophages and
several tumor cells next to a larger
tissue fragment. Papanicolaou stain,
medium power.

Ch010-F06731.indd 307 10/26/2006 10:33:42 AM


308 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-14
Renal cell carcinoma, papillary type.
Slightly enlarged, round or oval
nuclei consistent with a low-grade
(grade 2) tumor. Note the intra-
cytoplasmic hemosiderin pigment
in the tumor cells.

and tumor cells, and psammoma bodies differentiates


PAPILLARY RENAL CELL CARCINOMA – PATHOLOGIC FEATURES these tumors from other types of RCC.

Cytopathologic Findings
៉ Tumor tends to have hemorrhage, necrosis, and cystic
formations CHROMOPHOBE RENAL CELL CARCINOMA
៉ Type 1 papillary RCC: abundant papillary structures with vascular
cores; single or lobulated globular forms; psammoma bodies,
lipid-laden histiocytes, intracytoplasmic hemosiderin in CLINICAL FEATURES
histiocytes and tumor cells
៉ Type 2 papillary RCC: large acidophilic cytoplasm; higher nuclear
grade, prominent nucleoli The chromophobe cell type comprises about 5% of
RCCs. It occurs predominantly in middle-aged patients
Ancillary Studies and in both sexes equally.
៉ React to RCC markers, pancytokeratins, low molecular weight
cytokeratins, CD9 and CD10

Differential Diagnosis and Pitfalls CYTOPATHOLOGIC FEATURES


៉ Differentiating characteristics: papillary structures, lipid-laden
histiocytes, intracytoplasmic hemosiderin in histiocytes and
tumor cells, and psammoma bodies In FNA specimens, chromophobe cell RCCs occur as
single cells and in tight groups (Fig. 10-16). Tumor cells

and CD10 (Fig. 10-15). Reaction to high molecular


weight cytokeratins is rare and focal. Reactivity to CHROMOPHOBE RENAL CELL CARCINOMA – DISEASE FACT SHEET
vimentin is inconsistent.
Incidence
៉ About 5% of RCCs

DIFFERENTIAL DIAGNOSIS AND PITFALLS Gender and Age Distribution


៉ No gender predilection
៉ Predominantly middle-aged patients
The presence of papillary structures, lipid-laden histio-
cytes, intracytoplasmic hemosiderin in both histiocytes

Ch010-F06731.indd 308 10/26/2006 10:33:43 AM


CHAPTER 10 Kidney and Adrenal Gland 309

A B

FIGURE 10-15
Renal cell carcinoma, papillary type. A, Strong positive reaction to RCC
antibody. Cell block immunostain for RCC, medium power. B, Papillary RCC
with psammoma bodies. Strong reactivity for AE1/AE3. Cell block, medium
power. C, Membranous staining with CD10. Cell block, medium power.

FIGURE 10-16
Renal cell carcinoma, chromophobe
cell type. Single cells and tight
groups of tumor cells. Nuclei vary in
size and are eccentrically located.
Diff-Quik stain, low power.

Ch010-F06731.indd 309 10/26/2006 10:33:44 AM


310 FINE NEEDLE ASPIRATION CYTOLOGY

not always clearly defined. Nuclei are hyperchromatic


CHROMOPHOBE RENAL CELL CARCINOMA – with mildly irregular borders and vary in size. Binucle-
PATHOLOGIC FEATURES ation is common (Fig. 10-17). Occasional intranuclear
pseudoinclusions may be seen (Fig. 10-18). Small nucle-
Cytopathologic Findings oli may be present.
៉ Single cells and tight groups
៉ Tumor cells vary in size with usually abundant cytoplasm
៉ Two types of cells:
៉ Classic: pale cytoplasm with distinct borders ANCILLARY STUDIES
៉ Eosinophilic: dense, granular cytoplasm
៉ Perinuclear halos rarely seen
៉ Hyperchromatic nuclei with mildly irregular borders Chromophobe cell RCCs react with antibodies to cyto-
៉ Binucleation is common keratins (i.e. 7, 14, and pancytokeratin) and EMA.
៉ Small nucleoli may be present
Approximately half of the chromophobe cell RCCs are
positive for the RCC marker. They do not react for
Ancillary Studies
vimentin. In cell blocks, chromophobe RCCs show
៉ Reacts to antibodies to cytokeratins and EMA
diffuse cytoplasmic staining with Hale’s colloidal iron
៉ About half are positive for RCC marker
៉ Do not react for vimentin
stain.
៉ Diffuse cytoplasmic staining with Hale’s colloidal iron stain

Differential Diagnosis and Pitfalls


DIFFERENTIAL DIAGNOSIS AND PITFALLS
៉ Most likely to be confused with oncocytoma
៉ Nuclear border irregularities and perinuclear halos are not
present in oncocytoma, binucleation is rare
The main differential diagnosis of chromophobe cell
៉ Hale’s colloidal iron stain is negative
RCC is from oncocytoma. The former shows frequent
binucleation, pleomorphism (mostly variation in the
size of nuclei), nuclear border irregularities, and peri-
nuclear halos, in contrast to the round to oval uniform
vary in size with usually abundant cytoplasm, which nuclei of oncocytomas. Binucleation is rare, and nuclear
could be pale with distinct borders or dense granular, border irregularities and perinuclear halos are not
corresponding to the ‘classic’ and ‘eosinophilic’ type present in oncocytomas. Hale’s colloidal iron stain,
cells in histologic preparations. Others have vacuolated which is positive in chromophobe cell RCC and nega-
or flocculent cytoplasm. Perinuclear halos may be seen; tive in oncocytomas, is also helpful in the differential
but, unlike in histologic preparations, they are rare and diagnosis.

FIGURE 10-17
Renal cell carcinoma, chromophobe
cell type. Tumor cells have abun-
dant cytoplasm with well-defined
borders. Note the cells with clear,
pale and dense cytoplasm. Thin
perinuclear clearing is noticeable
in some cells. Binucleation is
frequent. There is moderate aniso-
cytosis and some nuclear border
irregularity. Diff-Quik stain, medium
power.

Ch010-F06731.indd 310 10/26/2006 10:33:45 AM


CHAPTER 10 Kidney and Adrenal Gland 311

FIGURE 10-18
Renal cell carcinoma, chromophobe
cell type. Intranuclear pseudoinclu-
sions. Occasional large nuclei with
irregular borders. Tumor cells with
pale and dense cytoplasm. Diff-Quik
stain, high power.

COLLECTING DUCT RENAL CELL papillary RCCs. Multicystic formations, due to dilated
ducts, are frequent.
CARCINOMA

CLINICAL FEATURES CYTOPATHOLOGIC FEATURES

This type accounts for less than 1% of RCCs and occurs Fine needle aspirates reveal small tissue fragments and
in a younger age group (34–43 years) with a male to single cells with apparent malignant features (Figs
female ratio of 2 : 1. It is an aggressive tumor with a 10-19 & 10-20). The former could be monolayer sheets
poor prognosis and is located in the medulla. Cytoge-
netic abnormalities found in this tumor (loss of arms
6p, 8p, 13q, and 21q, and monosomies 1, 6, 14, 15, and COLLECTING DUCT RENAL CELL CARCINOMA –
22) are different from those in conventional and PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Small tissue fragments and single cells with apparent malignant
features
COLLECTING DUCT RENAL CELL CARCINOMA – ៉ Sheets and tubular, rarely papillary, forms
DISEASE FACT SHEET ៉ Eccentrically located, enlarged pleomorphic nuclei, irregular
nuclear borders, vesicular or coarse chromatin
Incidence ៉ Single or multiple, prominent nucleoli
៉ Less than 1% of RCCs ៉ Small to moderate amounts of cytoplasm
៉ Fine or larger vacuoles containing mucin may be present
Gender and Age Distribution
៉ Male to female ratio is 2 : 1 Ancillary Studies
៉ Occurs in younger age group (34–43 years of age) ៉ Reacts to pankeratin, low and high molecular weight
cytokeratins, EMA, and peanut agglutinin
Clinical Features ៉ RCC marker is negative
៉ Aggressive tumor with poor prognosis
៉ Located in the medulla Differential Diagnosis and Pitfalls
៉ Cytogenetic abnormalities are different from those in ៉ Differs from chromophobe RCC, which has low-grade nuclei
conventional and papillary RCCs ៉ Positive staining with high molecular weight cytokeratins and
៉ Multicystic formations are frequent intracytoplasmic mucin differentiate it from conventional RCC

Ch010-F06731.indd 311 10/26/2006 10:33:46 AM


312 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-19
Renal cell carcinoma, collecting
duct cell type. A tissue fragment of
tumor. Many malignant features are
present, including marked nuclear
pleomorphism and marked variation
in N/C ratio. Diff-Quik stain, medium
power.

FIGURE 10-20
Renal cell carcinoma, collecting
duct cell type. Markedly atypical
single tumor cells. Diff-Quik stain,
high power.

Ch010-F06731.indd 312 10/26/2006 10:33:47 AM


CHAPTER 10 Kidney and Adrenal Gland 313

FIGURE 10-21
Renal cell carcinoma, collecting
duct cell type. Tumor cells with
large nuclei, macronucleoli, and
intracytoplasmic vacuoles. Papani-
colaou stain, high power.

and tubular or, rarely, papillary forms. Desmoplastic Differential diagnosis from medullary type RCC and
stromal tissue fragments may be present. The tumor metastatic neoplasms is difficult on cytomorphologic
cells have eccentrically located, enlarged, pleomorphic bases. Correlation with the clinical findings is neces-
nuclei with irregular borders, vesicular or coarse chro- sary in most cases.
matin, single or multiple prominent nucleoli, and small
or moderate amounts of cytoplasm, which may contain
fine or larger vacuoles (Fig. 10-21). Mucin may be
present. MEDULLARY-TYPE RENAL CELL
CARCINOMA

ANCILLARY STUDIES CLINICAL FEATURES

The tumor cells react to pankeratin (AE1/AE3), low This is a very aggressive, rare type of RCC which occurs
and high molecular weight cytokeratins, EMA, cyto- in young African-American men with sickle cell disease
keratins 7, 8, and 18, and peanut agglutinin. RCC or sickle cell trait. It is usually diagnosed in the advanced
marker is reported to be negative. stage.

DIFFERENTIAL DIAGNOSIS AND PITFALLS

The collecting duct RCC has a distinctly different MEDULLARY-TYPE RENAL CELL CARCINOMA –
cytomorphology from those of conventional-type and DISEASE FACT SHEET
chromophobe RCCs. The latter usually have low-grade
Clinical Features
nuclei, in contrast to the high-grade nuclei of collecting
៉ Very aggressive, rare type of RCC
duct RCC. Positive staining with antibodies to high
៉ Occurs in young African-American men with sickle cell disease or
molecular weight cytokeratins, and the presence of
sickle cell trait
intracytoplasmic mucin (mucicarmine and periodic ៉ Usually diagnosed in advanced stage
acid–Shiff [PAS] with diastase digest), help to differen-
tiate collecting duct RCC from conventional type.

Ch010-F06731.indd 313 10/26/2006 10:33:48 AM


314 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-22
Medullary-type renal cell carcinoma.
A tissue fragment. Poorly differen-
tiated carcinoma with large nuclei,
prominent nucleoli, and occasional
intracytoplasmic vacuoles. H&E
stain, low power. Courtesy of Srini-
vas R Mandavilli, MD.

CYTOPATHOLOGIC FEATURES ANCILLARY STUDIES

There are limited data on the cytopathologic features The tumor cells react to cytokeratins. Reaction to EMA
of this tumor. Cytomorphology is similar to that of and carcinoembryonic antigen (CEA) is variable. Mucin
other high-grade carcinomas. Tumor cells appear pre- stains are positive in the majority of the tumors.
dominantly in loosely cohesive groups. The nuclei are
large and pleomorphic with prominent nucleoli. Cyto-
plasm may be dense or vacuolated (Fig. 10-22).
DIFFERENTIAL DIAGNOSIS AND PITFALLS

Cytopathologic features of this tumor overlap with


those of collecting duct RCC and other poorly differen-
tiated primary or metastatic carcinomas. Clinical cor-
MEDULLARY-TYPE RENAL CELL CARCINOMA –
PATHOLOGIC FEATURES
relation is usually necessary to establish a definitive
diagnosis, since it occurs exclusively in patients with
Cytopathologic Findings sickle cell disease or sickle cell trait.
៉ Cytomorphology similar to other high-grade carcinomas
៉ Tumor cells appear predominantly in loosely cohesive groups
៉ Large pleomorphic nuclei with prominent nucleoli
៉ Cytoplasm may be dense or vacuolated SARCOMATOID RENAL CELL CARCINOMA

Ancillary Studies
៉ Reacts to antibodies to cytokeratins
CLINICAL FEATURES
៉ Variable reaction for EMA and CEA
៉ Mucin is positive in the majority of the tumors
Sarcomatoid changes are present in 1% to 6.5% of
RCCs, and many occur in large portions of the tumor.
Differential Diagnosis and Pitfalls
They may be found in all types of RCC and are not
៉ Difficult to differentiate from collecting duct RCC, and other
considered a separate type. Sarcomatoid changes indi-
poorly differentiated primary or metastatic carcinomas; clinical
correlation is necessary
cate a poor prognosis. In spite of the sarcomatous
morphology, tumor cells have the ultrastructural and
histochemical features of epithelial differentiation.

Ch010-F06731.indd 314 10/26/2006 10:33:49 AM


CHAPTER 10 Kidney and Adrenal Gland 315

SARCOMATOID RENAL CELL CARCINOMA – DISEASE FACT SHEET SARCOMATOID RENAL CELL CARCINOMA – PATHOLOGIC FEATURES

Incidence Cytopathologic Findings


៉ Present in 1% to 6.5% of RCCs ៉ Spindle cells with varying degrees of atypia, multinucleated
៉ May occur in all types of RCC (not considered a separate type) giant cells, and significant pleomorphism may be present
៉ Associated with other types of RCC
Clinical Features
៉ Indicates a poor prognosis Ancillary Studies
៉ Reaction to antibodies to cytokeratins and EMA varies
៉ RCC markers usually negative in sarcomatous cells

Differential Diagnosis and Pitfalls


៉ Rare metastatic sarcomas can be ruled out by reaction for
cytokeratins and the presence of other types of RCC
CYTOPATHOLOGIC FEATURES

In fine needle aspirates, in addition to the cellular com-


ponent presenting features of one of the specific types
of RCC, there are spindle cells with varying degrees
of atypia (Fig. 10-23). Multinucleated giant cells and reaction for cytokeratins. RCC marker is usually nega-
significant nuclear pleomorphism may be present. tive in sarcomatous cells of the tumor.

ANCILLARY STUDIES DIFFERENTIAL DIAGNOSIS AND PITFALLS

Immunohistochemical reaction with antibodies to With adequate sampling, identification of other types
cytokeratins and EMA varies. Both sarcomatous and of RCC and positive reaction to antibodies to cytokera-
epithelial components in some tumors have a positive tins helps to differentiate it from metastatic sarcomas.

FIGURE 10-23
Sarcomatoid renal cell carcinoma.
Poorly differentiated atypical cells
with sarcomatoid features. Papani-
colaou stain, high power.

Ch010-F06731.indd 315 10/26/2006 10:33:49 AM


316 FINE NEEDLE ASPIRATION CYTOLOGY

ONCOCYTOMA
ONCOCYTOMA – PATHOLOGIC FEATURES

CLINICAL FEATURES Cytopathologic Findings


៉ Combination of single, loose aggregates and tissue fragments of
oncocytic cells
Oncocytomas are benign neoplasms comprising 3.5% ៉ Uniform, round nuclei with small, bright red nucleoli
of renal tumors in adults. The majority of these tumors ៉ Finely granular, acidophilic cytoplasm with well-defined borders
occur in persons over the age of 50 years and are more
common in men than in women (2.3 : 1). About two- Ancillary Studies
thirds of oncocytomas are incidentally detected by ៉ Reacts to antibodies to EMA, low molecular weight cytokeratins,
radiologic examinations performed for other reasons. and most pancytokeratin cocktails
The most common cytogenetic abnormalities are ៉ CK7 strongly positive in scattered cells
losses of chromosomes 1 and 7, deletion of chromosome ៉ Reaction to CK20 varies
14, and rearrangements involving chromosome 11q13. ៉ Most oncocytomas are non-reactive to antibodies to vimentin
៉ RCC marker is negative

Differential Diagnosis and Pitfalls


CYTOPATHOLOGIC FEATURES ៉ Differential diagnoses include RCCs with oncocytic component
៉ With adequate sampling, other cell types can be ruled out
៉ Necrosis, mitoses, and macronucleoli rule out oncocytoma
FNA usually provides a moderately cellular specimen ៉ Eosinophilic-type chromophobe RCC vs oncocytoma can be
with a combination of single, loose aggregates, and difficult
៉ Chromophobe RCCs’ irregular nuclear borders with perinuclear
tissue fragments of oncocytic cells (Figs 10-24 & 10-
halos contrast with the uniform nuclear borders of oncocytic
25). The cells have uniform, round nuclei with small, cells
៉ Hale’s colloidal iron stain is negative in oncocytoma

ONCOCYTOMA – DISEASE FACT SHEET

Incidence, and Age and Gender Distribution bright red nucleoli and abundant, finely granular,
៉ Benign neoplasms comprising 3.5% of RCCs in adults over the age acidophilic cytoplasm with well-defined borders.
of 50 Mitoses and background necrosis are absent.
៉ Male to female ratio is 2.3 : 1
៉ About two-thirds are incidentally detected by imaging studies

Genetic Abnormalities ANCILLARY STUDIES


៉ Cytogenetic abnormalities include loss of chromosomes 1 and 7,
deletion of 14, and rearrangement of 11q13
Oncocytomas react to EMA, low molecular weight
cytokeratins, and most pancytokeratin cocktails. CK7

FIGURE 10-24
Oncocytoma. Round, uniform nuclei
with prominent nucleoli and abun-
dant cytoplasm. Diff-Quik stain,
high power.

Ch010-F06731.indd 316 10/26/2006 10:33:50 AM


CHAPTER 10 Kidney and Adrenal Gland 317

FIGURE 10-25
Oncocytoma. These tumor cells
show characteristic features of
oncocytoma: single cells with
large, centrally located nuclei and
prominent nucleoli. Cytoplasm is
granular. Papanicolaou stain, high
power.

is strongly positive in scattered cells, and reaction to


CK20 varies. Most oncocytomas are non-reactive to NEPHROBLASTOMA (WILMS TUMOR) – DISEASE FACT SHEET
vimentin. The RCC marker is negative.
Incidence
៉ Most common genitourinary neoplasm in children
៉ Comprises ~90% of pediatric renal malignancies
DIFFERENTIAL DIAGNOSIS AND PITFALLS
Gender and Age Distribution
៉ 98% of nephroblastomas occur in children under 10 years of age
Many RCCs have oncocytic cells; however, entirely ៉ Mean age is 36 months for males and 42 months for females
oncocytic cell populations occur only in oncocytomas.
Extensive samplings of the tumor, along with clinical Clinical Features
and radiologic correlations, are necessary for a defini- ៉ Association with genetic syndromes involving WT1 and WT2 genes
tive diagnosis. The presence of necrosis, mitoses, and has been shown
macronucleoli rules out a diagnosis of oncocytoma. The ៉ The majority of tumors are single, but multiple synchronous
differential diagnosis between oncocytoma and the unilateral or bilateral tumors may occur
eosinophilic type of chromophobe RCC can be difficult.
Chromophobe RCCs have irregular nuclear borders
(‘raisinoid’ nuclei) with perinuclear halos, in contrast
to the uniform, smooth nuclear borders of oncocytic 10 years of age, the mean age being 36 months for males
cells of oncocytomas. Eosinophilic-type chromophobe and 42 months for females. Association with certain
RCC shows diffuse cytoplasmic staining with Hale’s genetic syndromes involving WT1 and WT2 genes
colloidal iron, which is negative in oncocytoma. Lack among others has been shown. The majority of nephro-
of intracytoplasmic glycogen and mucin, and non- blastomas are single tumors; however, multiple syn-
reactivity to RCC marker, can be helpful in distinguish- chronous unilateral or bilateral tumors may occur.
ing oncocytomas from RCCs.

CYTOPATHOLOGIC FEATURES
NEPHROBLASTOMA (WILMS TUMOR)
Cytopathologic diagnosis of nephroblastoma requires
CLINICAL FEATURES adequate sampling of the tumor. Blastemal cells occur
in sheets, groups, or single forms. Some molding is
present. The cells are small with very scant, fragile
Nephroblastoma is a malignant embryonal neoplasm of cytoplasm (Fig. 10-26). Nuclei show fine, evenly dis-
childhood. It is the most common genitourinary malig- tributed chromatin and inconspicuous nucleoli. Epithe-
nant neoplasm in children, and comprises approxi- lial cells form glandular structures of primitive cells.
mately 90% of pediatric renal malignancies. Ninety-eight Stromal cells are seen in cellular tissue fragments com-
percent of the nephroblastomas occur in children under posed of tightly arranged spindle cells and capillaries.

Ch010-F06731.indd 317 10/26/2006 10:33:51 AM


318 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-26
Wilms tumor. A fragment of undif-
ferentiated carcinoma. Tumor cells
have somewhat pleomorphic nuclei
and high N/C ratio. On morphology
alone, this tumor cannot be differ-
entiated from other small cell
tumors of childhood. Papanicolaou
stain, high power.

vimentin and may express neuron-specific enolase


NEPHROBLASTOMA (WILMS TUMOR) – PATHOLOGIC FEATURES (NSE). Nuclear staining for WT1 is positive in blaste-
mal cells and in epithelial cells in early differential
Cytopathologic Findings stages. Blastemal cells are also positive for desmin,
៉ Blastemal cells occur in sheets, groups, or singly but they are usually negative for primitive muscle
៉ Small cells with scant, very fragile cytoplasm markers.
៉ Fine, evenly dispersed chromatin, inconspicuous nucleoli
៉ Glandular structures of primitive epithelial cells
៉ Cellular tissue fragments of spindle cells and capillaries
៉ Admixture of blastemal epithelial and stromal cells DIFFERENTIAL DIAGNOSIS AND PITFALLS
៉ Anaplasia characterized by hyperchromatic nuclei and multipolar
mitotic figures
The differential diagnosis of nephroblastoma includes
Ancillary Studies other primary pediatric tumors of kidney, such as rhab-
៉ Blastemal cells positive for vimentin, may be positive with NSE, doid tumor, clear cell carcinoma, congenital mesoblas-
and are positive with WT1 in early differential stages tic nephroma, and so-called small blue cell tumors. Blue
៉ Also positive with desmin, but negative for primitive muscle
cell tumors in childhood include neuroblastoma, primi-
markers
tive neuroectodermal tumor (PNET), rhabdomyo-
Differential Diagnosis and Pitfalls
sarcoma, and lymphoma. These tumors are rare and
together comprise about 15% of pediatric renal neo-
៉ Include other primary pediatric tumors of kidney
៉ These tumors are rare and, with a few exceptions, specific
plasms. With a few exceptions, a specific diagnosis of
diagnosis can only be made on tissue samples representative of these tumors can be made only by examining tissue
the tumor samples representative of the tumor. In the appropriate
clinical setting, the presence of three characteristic
patterns of nephroblastoma with positive WT1 stain-
ing differentiates this tumor from others with similar
Areas of an admixture of two or all three components morphology.
are present. Anaplasia can be diagnosed using the same
criteria as those for histopathologic specimens. The
presence of very large, hyperchromatic nuclei and
multipolar mitotic figures confirms the diagnosis. RHABDOID TUMOR

CLINICAL FEATURES
ANCILLARY STUDIES
Rhabdoid tumor is a highly malignant, rare renal neo-
Immunohistochemical reactions vary according to the plasm (2.5% of pediatric renal tumors), 95% of which
differentiation. Blastemal cells are usually positive for occur in children under 3 years of age. Genetic

Ch010-F06731.indd 318 10/26/2006 10:33:51 AM


CHAPTER 10 Kidney and Adrenal Gland 319

RHABDOID TUMOR – DISEASE FACT SHEET RHABDOID TUMOR – PATHOLOGIC FEATURES

Incidence and Age Distribution Cytopathologic Findings


៉ Rare, highly malignant neoplasm ៉ Hypercellular aspirates
៉ Comprises ~2.5% of pediatric renal tumors ៉ Monotonous atypical tumor cells with abundant eosinophilic
៉ 95% in children under 3 years of age cytoplasm, eccentric large nuclei, centrally located prominent
nucleoli
Genetic Abnormalities ៉ Necrosis and frequent mitotic figures
៉ Genetic abnormalities involving hSNF5/IN11 gene located at
chromosome 22q11 Ancillary Studies
៉ Immunohistochemistry:
៉ Intense reactivity for vimentin
៉ Scattered cells showing strong immunoreactivity for
cytokeratins and EMA in a background of non-reactive tumor
abnormalities involving the hSNF5/INI1 gene located cells
at chromosome 22q11 are present in both renal and ៉ Electron microscopy: intracytoplasmic whorls of thick
extrarenal rhabdoid tumors of infancy. intermediate filaments

Differential Diagnosis and Pitfalls


៉ Neuroblastoma, nephroblastoma, congenital mesoblastic
CYTOPATHOLOGIC FEATURES nephroma, and clear cell sarcoma may have foci similar to
rhabdoid tumor
៉ Differential diagnosis may be difficult, if not impossible, in
FNA specimens are usually very cellular and composed limited samples
of monotonous, atypical tumor cells with abundant
eosinophilic cytoplasm and eccentric large nuclei with
centrally located prominent nucleoli (Fig. 10-27).
Necrosis and frequent mitotic figures are present.

DIFFERENTIAL DIAGNOSIS AND PITFALLS


ANCILLARY STUDIES
Neuroblastoma, nephroblastoma, congenital meso-
Intense reactivity for vimentin and the presence of blastic nephroma, and clear cell sarcoma of kidney
strong immunoreactivity for cytokeratins and EMA in may have foci similar to rhabdoid tumor, making the
a background of non-reactive tumor cells are character- differential diagnosis difficult in limited samples.
istic of these tumors. Examination of the entire tumor with ample sampl-
Electron microscopy shows intracytoplasmic whorls ing may be required to find areas characteristic of
of thick intermediate filaments, which are characteristic other types of tumors to differentiate them from
of this tumor (Fig. 10-28). rhabdoid.

FIGURE 10-27
Rhabdoid tumor. Large cells with
eccentric nuclei and macronucleoli.
High power. Courtesy of Paul E
Wakely, Jr, MD.

Ch010-F06731.indd 319 10/26/2006 10:33:52 AM


320 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-28
Rhabdoid tumor. Electron micro-
graph showing whorled intermedi-
ate filaments. They can also be
found in other diseases. Courtesy of
Paul E Wakely, Jr, MD.

ANGIOMYOLIPOMA CYTOPATHOLOGIC FEATURES

CLINICAL FEATURES FNA usually provides the diagnosis by allowing iden-


tification of adipose tissue and smooth muscle cells
(Fig. 10-29). Thick-walled blood vessels are seen both
Angiomyolipoma is a benign mesenchymal neoplasm in cell blocks and in core biopsies. Smooth muscle cells
composed of a mixture of adipose tissue, smooth muscle show varying degrees of nuclear atypia. In some cases,
cells, and thick-walled blood vessels. The tumor has a significant nuclear atypia may suggest a malignant neo-
strong association with tuberous sclerosis, which is a plasm; but the presence of fat tissue and more differen-
genetic disorder caused by mutation of either the TSC1 tiated elements of smooth muscle help to establish the
or TSC2 gene. While about 80% of patients with tuber- correct diagnosis.
ous sclerosis develop angiomyolipomas, less than half
of patients with angiomyolipomas have tuberous scle-
rosis. Other hereditary disorders associated with angio- ANCILLARY STUDIES
myolipoma include von Hippel–Lindau syndrome, von
Recklinghausen disease, and autosomal dominant poly-
cystic kidney disease. Angiomyolipomas comprise up Muscle markers (desmin, smooth muscle actin, muscle-
to 2% of renal tumors, and the female to male ratio is specific actin) and vimentin are consistently positive.
2 : 1. These tumors can be distinguished radiologically
from other renal neoplasms.
ANGIOMYOLIPOMA – PATHOLOGIC FEATURES

Cytopathologic Findings
ANGIOMYOLIPOMA – DISEASE FACT SHEET ៉ Varying proportion of adipose tissue and smooth muscle cells
៉ Thick-walled blood vessels seen in cell blocks and core biopsies
Incidence ៉ Varying degrees of nuclear atypia in smooth muscle cells
៉ Up to 2% of renal tumors
Ancillary Studies
Gender Distribution ៉ Muscle markers and vimentin consistently positive
៉ Female to male ratio is 2 : 1 ៉ HMB-45 is usually expressed
៉ c-kit (CD117) is positive in all cases
Clinical Features
៉ Strong association with tuberous sclerosis Differential Diagnosis and Pitfalls
៉ About 80% of tuberous sclerosis patients develop angiomyolipoma ៉ Rarely, atypical smooth muscle cells may be confused with
៉ Other hereditary disorders associated with angiomyolipoma are cancer
von Hippel–Lindau syndrome, von Recklinghausen disease, and ៉ Presence of adipose tissue and thick-walled blood vessels
autosomal dominant polycystic kidney disease confirms the diagnosis

Ch010-F06731.indd 320 10/26/2006 10:33:52 AM


CHAPTER 10 Kidney and Adrenal Gland 321

B
A

C D

FIGURE 10-29
Angiomyolipoma. Mixture of adipose tissue and large cells with atypical smooth muscle cells with large nuclei, one (D) with intranuclear pseudo-
inclusion. A, Diff-Quik stain, low power; B, Diff-Quik stain, high power; C,D, Papanicolaou stain, high power.

Melanocytic markers, specifically HMB-45, are usually


expressed; and c-kit (CD117) is positive in all cases. CYSTIC LESIONS OF KIDNEY – DISEASE FACT SHEET

Clinical Features
៉ Common in kidney
DIFFERENTIAL DIAGNOSIS AND PITFALLS ៉ Most are benign, simple cysts
៉ FNA is indicated in multilocular complex cysts
៉ Multiloculated complex cysts are considered atypical
In general, the cytopathologic findings are sufficient to
establish a definitive diagnosis of this tumor. The pres-
ence of adipose tissue and characteristic blood vessels
helps to differentiate it from benign and malignant
smooth muscle tumors and sarcomatous RCC. Some (Figs 10-30 & 10-31). In addition to the renal neoplasms
angiomyolipomas with epithelioid features can be dif- with cystic growth patterns, up to 15% of RCCs have
ferentiated from melanoma with the help of immuno- degenerative cystic changes.
histochemistry, with positive staining for smooth
muscle markers.

BENIGN CYSTS
CYSTIC LESIONS OF KIDNEY
CYTOPATHOLOGIC FEATURES
Cystic lesions of kidney are common. Most are benign,
simple cysts; however, multiloculated complex cysts are The predominant cell component of benign cysts is
considered atypical, and FNA is indicated to rule out macrophages. In aspirates, they appear as single cells
malignancy. In the aspirates of cystic fluid, exfoliated or loose aggregates. They have eccentric or centrally
cells of benign epithelium may show significant atypia located nuclei with bland chromatin patterns and

Ch010-F06731.indd 321 10/26/2006 10:33:53 AM


322 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-30
Cystic lesions. Rounded tissue frag-
ment of atypical epithelial cells.
This patient had a benign cyst with
atypical epithelial lining. Papanico-
laou stain, high power.

FIGURE 10-31
Cystic lesions. Two groups of
atypical, but markedly degenerated
cells, which may be of epithelial or
histiocytic origin. Note that these
are not true tissue fragments, but
rather aggregates of cells with
degenerative changes. Papanico-
laou stain, high power.

moderate amounts of cytoplasm, which may contain


BENIGN CYSTS – PATHOLOGIC FEATURES engulfed particles, usually hemosiderin. Nucleoli, when
present, are usually small, but prominent nucleoli
Cytopathologic Findings may be seen. Ring-shaped structures with double walls
៉ Macrophages are the predominant cell population in aspirates (Liesegang rings) may be found in the cyst fluid (Fig.
៉ Appear singly or in loose aggregates 10-32) Epithelial elements include fragments of renal
៉ Eccentric or centrally located nuclei, bland chromatin pattern tubular epithelium and cyst-lining epithelium.
៉ Cytoplasm containing engulfed particles, usually hemosiderin
៉ Prominent nucleoli may be present
៉ Epithelial cells may represent renal tubular epithelium or
cyst-lining epithelium
DIFFERENTIAL DIAGNOSIS AND PITFALLS
Differential Diagnosis and Pitfalls
៉ Should be differentiated from malignant neoplasms
៉ Liesegang rings may be confused with cysts of ova of parasites
Distinction from malignant cystic neoplasms can
be difficult in FNA specimens, especially in aspirates
with scant cellularity. The differential diagnosis

Ch010-F06731.indd 322 10/26/2006 10:33:55 AM


CHAPTER 10 Kidney and Adrenal Gland 323

A B

FIGURE 10-32
Cystic lesions. Double-walled, round structures (Liesegang rings) in an infl ammatory background. A, Papanicolaou stain, low power; B, cell block,
medium power.

will be discussed in detail under malignant cystic


lesions. MALIGNANT CYSTIC LESIONS – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Tumor cells present singly, as small aggregates, or tissue
fragments
MALIGNANT CYSTIC LESIONS ៉ Necrotic debris and hemosiderin-laden macrophages may be
present

The majority of malignant cystic lesions in kidney are Differential Diagnosis and Pitfalls
cystic degeneration of various types of RCCs. Papillary ៉ Differential diagnosis between benign cyst and RCC with cystic
RCCs usually present as a cystic lesion with a thick degeneration can be difficult
capsule. Multilocular cystic RCC is primarily a cystic ៉ Tissue fragments with cytologic atypia, when present, are
malignant renal neoplasm considered to be a subtype indicative of malignancy
of clear cell RCC. ៉ Immunohistochemistry is of limited value in differential
diagnosis
៉ Macrophage and epithelial markers would help in differential
diagnosis of macrophages from RCC

CYTOPATHOLOGIC FEATURES

Tumor cells are present singly or in small aggregates or


tissue fragments. Cell morphology varies according to DIFFERENTIAL DIAGNOSIS AND PITFALLS
the tumor type and nuclear grade. In RCCs with degen-
erative changes, necrotic debris and hemosiderin-laden The differential diagnosis between clear cell RCC and
macrophages can be present. multilocular cystic RCC cannot be made in fine needle
aspirates. The differential diagnosis between benign
cysts and RCCs with cystic degeneration can also be
difficult in cytologic samples. In benign cysts, macro-
phages and epithelial lining cells are present. Both may
MALIGNANT CYSTIC LESIONS – DISEASE FACT SHEET have vacuolated cytoplasm and prominent nucleoli.
Intracytoplasmic hemosiderin pigment may occur in
Clinical Features
both macrophages and the papillary variant of RCC.
៉ Majority of malignant cystic lesions are cystic degeneration of
Tissue fragments, when present, are a reliable feature
RCCs
for differentiating neoplastic cells from macrophages,
៉ Papillary RCCs present as a cystic lesion with a thick capsule
៉ Multilocular cystic RCC is a subtype of clear cell RCC
which usually occur as single cells or loose aggre-
gates. Although prominent nucleoli may be seen in
macrophages and benign epithelial cells, the single

Ch010-F06731.indd 323 10/26/2006 10:33:56 AM


324 FINE NEEDLE ASPIRATION CYTOLOGY

macronucleoli, which are frequently present in RCCs, CYTOPATHOLOGIC FEATURES


are not usually present in benign cyst-lining epithelium
or in macrophages. In the absence of well-preserved
tumor cells with macronucleoli or tissue fragments The majority of urothelial tumors of kidney pelvis are
composed of atypical cells, the diagnosis of RCC should high grade. FNA specimens are usually cellular, com-
be made with great caution. prised predominantly of loose aggregates, single cells,
Immunohistochemistry has limited use in the differ- and some tissue fragments. Tumor cells have pleomor-
ential diagnosis. Macrophage markers (e.g. CD68 and phic nuclei and a moderate to high nuclear to cytoplas-
HAM 56) and AE1/AE3 would help in the differential mic (N/C) ratio. Tumor nuclei show uneven chromatin
diagnosis of macrophages from RCC. clumping, irregular nuclear borders, and prominent
nucleoli (Fig. 10-33).

UROTHELIAL CARCINOMA OF RENAL PELVIS

CLINICAL FEATURES
UROTHELIAL CARCINOMA OF RENAL PELVIS –
PATHOLOGIC FEATURES
Urothelial carcinomas of renal pelvis are rare. They
occur in older populations (mean age, 67 years) and Cytopathologic Findings
are more common in men. Approximately half of the ៉ Majority of these tumors are high grade
patients also have lower urinary tract neoplasms. ៉ Cellular specimen, predominantly loose aggregates, single cells,
and some tissue fragments
៉ Moderate to high N/C ratio
៉ Uneven chromatin clumping, irregular nuclear borders, and
UROTHELIAL CARCINOMA OF RENAL PELVIS – prominent nucleoli
DISEASE FACT SHEET
Ancillary Studies
Incidence
៉ Immunostains are positive for CK7 and CK20, thrombomodulin,
៉ Rare CK903, and CEA
៉ Uroplakin III is more specific, but not sensitive
Gender and Age Distribution
៉ Occurs in older population, mostly in men Differential Diagnosis and Pitfalls
៉ Majority of RCCs have characteristic cytologic features
Clinical Features differentiating them from urothelial carcinoma
៉ Approximately half of patients also have lower urinary tract ៉ Poorly differentiated RCCs, however, may not be differentiated
neoplasms from high-grade carcinomas

FIGURE 10-33
Urothelial carcinoma of renal pelvis.
Poorly differentiated carcinoma
with pleomorphic nuclei. Although
these cytopathologic features are
unusual for RCC, the primary site
cannot be determined on morpho-
logic basis alone. Papanicolaou
stain, high power.

Ch010-F06731.indd 324 10/26/2006 10:33:57 AM


CHAPTER 10 Kidney and Adrenal Gland 325

ANCILLARY STUDIES
LYMPHOMA – PATHOLOGIC FEATURES

Tumor cells react to antibodies for CK7 and CK20, Cytopathologic Findings
thrombomodulin, high molecular weight cytokeratin ៉ Primary lymphomas are usually diffuse, large B-cell type
(CK903), and CEA; however, these antibodies are not ៉ FNA specimens contain uniform, round, single cells
specific. Uroplakin III is more specific, but not ៉ Morphology differs according to cell type
៉ In addition to cytopathologic examination, ancillary techniques
sensitive.
are necessary to establish a diagnosis

Ancillary Studies
DIFFERENTIAL DIAGNOSIS AND PITFALLS ៉ Flow cytometry from rinses of FNA samples, or
immunohistochemistry performed on cell blocks, core biopsies,
or rinses is necessary for specific diagnosis
The majority of RCCs have characteristic cytologic fea-
tures which differentiate them from urothelial carcino- Differential Diagnosis and Pitfalls
mas without difficulty. In limited samples, however, ៉ In adult patients, small cell carcinoma of lung is the main
poorly differentiated, high-grade RCCs may not be dif- tumor in differential diagnosis
៉ Presence of tissue fragments with nuclear molding
ferentiated from high-grade urothelial carcinomas.
differentiates small cell carcinoma from lymphoma, which has
single cells
៉ Immunohistochemistry with neuroendocrine and lymphocytic
markers usually establishes the diagnosis in equivocal cases
LYMPHOMA ៉ In children, differential diagnoses include small blue cell
tumors, which can also be differentiated using lymphocytic
markers
Kidneys are commonly involved in the late stages of
lymphomas. Primary lymphomas of kidney are rare
and comprise less than 1% of primary extranodal lym-
phomas. Both kidneys are involved in more than 40%
of the cases. FNA usually provides the diagnosis by
cytopathologic examination and flow cytometry
analysis.
ANCILLARY STUDIES

A specific diagnosis of lymphoma requires phenotyping


CYTOPATHOLOGIC FEATURES studies by flow cytometry from rinses of FNA samples
or immunohistochemistry performed on cell blocks,
Primary renal cell lymphomas are usually diffuse, large core biopsies, or rinses. Detailed discussion of lympho-
B-cell type, but other subtypes may occur. FNAs reveal mas is covered in Chapter 3.
a uniform, round, single cell population (Fig. 10-34).
Lymphomas should be considered in any cytologic
sample composed of uniform, single cells, with high
N/C ratio. Cytomorphology differs according to the cell DIFFERENTIAL DIAGNOSIS AND PITFALLS
type, ranging from normal-appearing lymphocytes (e.g.
MALT) to undifferentiated malignant tumor cells (e.g. In adult patients, the main differential diagnosis is
anaplastic lymphomas). Ancillary techniques (i.e. flow from metastatic small cell carcinoma of lung. The pres-
cytometry or immunohistochemistry), in addition to ence of tissue fragments with nuclear molding in small
cytopathologic examination, are necessary to deter- cell carcinoma versus single cells in lymphoma is a reli-
mine the lymphoid nature and clonality of the tumor able differential diagnostic feature for both low- and
cells to establish a definitive diagnosis. high-grade lymphomas. It should be kept in mind that
in air-dried, Diff-Quik-stained specimens, and in some
alcohol-fi xed smears, tight cellular aggregates of lym-
phoma may give the impression of molding. Although
the evenly dispersed, granular chromatin pattern (‘salt
LYMPHOMA – DISEASE FACT SHEET
and pepper’) characteristic for neuroendocrine neo-
plasms is helpful in the differential diagnosis, it is not
Clinical Features
always present; and, depending on cell preservation,
៉ Lymphomas in late stages commonly involve kidneys
nuclei of small cell carcinomas can have irregular chro-
៉ Primary lymphomas are rare (less than 1% of primary extranodal
lymphomas)
matin clumping or diffuse hyperchromasia. Immuno-
៉ Both kidneys are involved in more than 40% of cases
histochemistry with neuroendocrine and lymphocytic
markers usually establishes the diagnosis in morpho-
logically equivocal cases. Among primary small round

Ch010-F06731.indd 325 10/26/2006 10:33:57 AM


326 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-34
Large B-cell lymphoma. Single round
cells with prominent nucleoli.

cell tumors of kidney, immunohistochemical staining CYTOPATHOLOGIC FEATURES


characteristics separate primitive neuroectodermal
tumor (PNET) and nephroblastoma from lymphomas.
PNET/Ewing sarcomas are characterized by MIC2/ Cytopathologic findings vary according to the tumor.
CD99 expression. Immunostains show membranous
staining.

ANCILLARY STUDIES
METASTATIC NEOPLASMS TO KIDNEY
Cytokeratins help to determine the epithelial nature of
poorly differentiated malignant neoplasms. Organ- or
In late stages, malignant neoplasms from other sites tumor-specific antibodies, such as those for thyro-
may metastasize to the kidneys. Carcinomas of lung, globulin, prostate-specific antigen (PSA), thyroid
breast, and gastrointestinal tract, malignant melanoma,
as well as RCC of the contralateral kidney are among
the most common metastatic neoplasms to kidney.
Metastases into the RCC may also occur. In most cases,
the primary tumor is clinically known. METASTATIC NEOPLASMS TO KIDNEY – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Vary according to the tumor

Ancillary Studies
៉ Cytokeratins help to identify the epithelial origin of poorly
differentiated neoplasms
៉ Organ- or tumor-specific antibodies, along with morphology,
METASTATIC NEOPLASMS TO KIDNEY – DISEASE FACT SHEET determine the primary site

Clinical Features Differential Diagnosis and Pitfalls


៉ Most common metastatic tumors are carcinomas of lung, breast, ៉ Majority of RCCs can be differentiated from metastatic tumors
and gastrointestinal tract, and malignant melanoma by their distinct morphology

Ch010-F06731.indd 326 10/26/2006 10:33:58 AM


CHAPTER 10 Kidney and Adrenal Gland 327

transcription factor-1 (TTF-1), and HepPar 1, are left adrenal masses. The adequacy rate is 100% in most
helpful in determining the primary site of neoplasms series, with a very high accuracy in diagnosis. Complica-
of unknown primary site or in the differential diagno- tions of FNA of the adrenal gland are rare. Pneumotho-
sis of tumors with similar morphology. rax, hemorrhage, abscess, and tumor implant in needle
tract have been reported.

DIFFERENTIAL DIAGNOSIS AND PITFALLS


ADRENAL CYSTS
Differential diagnosis is usually not difficult. The
majority of RCCs have distinct morphology which dif- CLINICAL FEATURES
ferentiates them from metastatic tumors. In addition to
morphology, immunohistochemistry can be helpful for
determining or confirming the specific type of the neo- Cystic lesions of adrenal glands are rare. They are clas-
plasm. Poorly differentiated RCCs, including collecting sified as pseudocysts, endothelial cysts (angiomatous
duct type and medullary type, can be difficult to dif- and lymphangiomatous), epithelial cysts, and parasitic
ferentiate from metastatic poorly differentiated neo- cysts. Pseudocysts are the most common type (56%)
plasms on morphologic bases alone. In these cases, followed by endothelial cysts (24%). True epithelial
immunohistochemistry has limited value because of cysts are very rare, comprising about 6% of the cystic
overlapping results. Core biopsies, as well as clinical lesions. Parasitic cysts, which are usually caused by
and radiologic correlations, are helpful in establishing Echinococcus, occur in areas endemic for this parasite.
the diagnosis. The majority of the cysts range between 5 and 10 cm
(mean size, 9.6 cm), but very large cysts, over 20 cm,
may occur (the largest reported was 50 cm). The female
to male ratio is 2 : 1. The age distribution has two peaks:
one at <1 month; the other in the fourth and fifth
ADRENAL GLAND decades. Adrenal cysts may be either symptomatic
or discovered incidentally (34%). Association with
adrenal cortical neoplasms and pheochromocytomas
has been reported. Metastatic carcinomas from various
Fine needle aspirations of adrenal glands are performed sites may form cystic lesions on rare occasions. Aspira-
on nodular lesions or diffusely enlarged glands to deter- tion of the cysts is recommended for both diagnostic
mine the nature of a lesion which is clinically and and therapeutic purposes. If the cyst fluid is bloody,
radiologically unclear. Nodular lesions may be symp-
tomatic or asymptomatic and benign or malignant. The
adrenal cortex secretes corticosteroids, aldosterone,
cortisol, and sex steroids, and the medulla secretes cat-
echolamines, epinephrine, and norepinephrine. Hyper-
secretory neoplasms or hyperplastic lesions have ADRENAL CYSTS – DISEASE FACT SHEET
clinical symptoms caused by hypersecretion of one of
Incidence
these products. An increasing number of clinically
៉ Cystic lesions of adrenal glands are rare
occult neoplasms or hyperplastic nodules are inciden-
៉ Classified as pseudocysts, endothelial, epithelial, and parasitic
tally detected by abdominal imaging studies (i.e. CT,
cysts
MRI, or US) performed for other reasons. These lesions ៉ Pseudocysts are the most common (56%), followed by endothelial
are called ‘incidentaloma’ in the literature. Clinically cysts (24%)
symptomatic, functional adrenal masses are usually ៉ True epithelial cysts are very rare (6%)
removed without prior FNA. The majority of inciden- ៉ Parasitic cysts, usually caused by Echinococcus, occur only in
talomas are benign, non-functioning adenomas; malig- areas where this parasite is endemic
nancy rates vary from 4% to 35% in published series.
Larger masses (over 4 cm) have a higher incidence of Gender and Age Distribution
malignancy, and they are usually operated on without ៉ Male to female ratio is 2 : 1
prior FNA. Malignant neoplasms, however, have been ៉ Age distribution has two peaks: <1 month, and 4th and 5th
reported in up to about 18% of nodules measuring less decades
than 2 cm. Correlation of FNA and MRI is recom-
mended as a cost-effective approach to the diagnosis of Clinical Features
these tumors. ៉ Majority of the cysts range between 5 and 10 cm, but cysts more
In general, FNA has a high sensitivity (about 85%) 20 cm may occur
៉ About one-third are discovered incidentally
and specificity (100%) in the diagnosis of mass lesions
៉ FNA recommended for both diagnostic and therapeutic purposes
of the adrenal glands. Higher inadequacy rates (28–
៉ Surgery is indicated if fluid is bloody, cystogram is irregular, or
33.3%) have been reported in some series of CT- or pheochromocytoma or malignancy is suspected
US-guided FNAs. In recent years, endoscopic ultra-
sound (EUS) has been used successfully for sampling

Ch010-F06731.indd 327 10/26/2006 10:33:58 AM


328 FINE NEEDLE ASPIRATION CYTOLOGY

ADRENAL CYSTS – PATHOLOGIC FEATURES MYELOLIPOMA – DISEASE FACT SHEET

Cytopathologic Findings Incidence


៉ Specimen is usually hypocellular ៉ Rare benign neoplasm of adrenal gland
៉ Bloody specimens may be seen in malignancy
៉ Hemosiderin- or lipid-laden macrophages may be present Gender and Age Distribution
៉ Roughly equally distributed between males and females
Differential Diagnosis and Pitfalls ៉ Average age is around 50 years
៉ Bloody fl uids should be examined carefully to rule out
malignancy Clinical Features
៉ Cortical adenomas can be difficult to differentiate from lipid- ៉ Generally asymptomatic and incidentally found by imaging studies
laden macrophages in hypocellular specimens or at autopsy
៉ Usually occurs unilaterally
៉ Tumor size varies greatly, ranging from <1 cm to >30 cm
៉ Because of their typical radiologic features (on CT and MRI), FNA
is rarely performed for diagnosis

cystogram is irregular, or pheochromocytoma or malig-


nancy is suspected, surgery is indicated.
radiologic features, specifically on CT and MRI, FNA
is rarely performed for diagnosis.
CYTOPATHOLOGIC FEATURES

Cyst fluid could be clear, turbid, or bloody. The latter CYTOPATHOLOGIC FEATURES
may be seen in malignancy, but it also occurs in pseu-
docysts. The specimen is usually hypocellular. Eryth- These are usually cellular specimens composed of a
rocytes, leukocytes, and hemosiderin- or lipid-laden mixture of adipose tissue and hematopoietic cells in
macrophages may be present. varying proportions (Fig. 10-35). Hematopoietic cells
include myeloid, lymphoid, and erythroid elements in
various stages of maturation, and megakaryocytes.
DIFFERENTIAL DIAGNOSIS AND PITFALLS

In bloody cyst fluids, the specimen should be examined DIFFERENTIAL DIAGNOSIS AND PITFALLS
carefully to rule out malignancy. Benign neoplasms,
such as cortical adenomas, can be difficult to differenti- Combined with the radiologic findings, FNA diagnosis
ate from lipid-laden macrophages in hypocellular of myelolipoma is definitive. In cases with predomi-
specimens. nantly hematopoietic cells and minimal adipose tissue,
extramedullary hematopiesis may be considered in the
differential diagnosis. The latter usually involves mul-
MYELOLIPOMA tiple organs, in contrast to adrenal myelolipoma.

CLINICAL FEATURES
MYELOLIPOMA – PATHOLOGIC FEATURES

Myelolipoma is a rare, benign neoplasm of adrenal Cytopathologic Findings


gland. It is generally asymptomatic and diagnosed inci- ៉ Cellular specimens composed of a mixture of adipose tissue and
dentally by imaging studies or at autopsy. With the hematopoietic cells in varying proportions
advancement of imaging techniques, an increasing ៉ Hematopoietic cells include myeloid, lymphoid, and erythroid
number of myelolipomas are detected. The tumor is elements, and megakaryocytes
roughly equally distributed between males and females,
the average age being around 50 years. Myelolipoma Differential Diagnosis and Pitfalls
usually occurs unilaterally; bilateral occurrence is very ៉ Combined with radiological findings, FNA diagnosis of
rare. Tumor size varies greatly, ranging from less than myelolipoma is definitive
៉ Cases with predominantly hematopoietic cells may be mistaken
1 cm to more than 30 cm. Rare ‘giant myelolipomas’,
for extramedullary hematopoiesis; the latter usually involves
over 4000 g (the largest, 6000 g), have been reported. multiple organs
These tumors may be associated with endocrine dis-
orders. Because most of these tumors have typical

Ch010-F06731.indd 328 10/26/2006 10:33:58 AM


CHAPTER 10 Kidney and Adrenal Gland 329

FIGURE 10-35
Myelolipoma. Low power: mixture of
adipose tissue and myeloid and lym-
phoid elements. High power: mega-
karyocytes. Papanicolaou stain.

NODULAR CORTICAL HYPERPLASIA AND abdominal CT scanning, 1.3–3.4% of patients may be


CORTICAL ADENOMA found to have clinically occult adrenal masses more
than 1 cm in size. Adrenal cortical hyperplasia and
adenoma may be hyperfunctional and accompanied by
CLINICAL FEATURES clinical syndromes such as Cushing (excess cortisol
secretion), Conn (hyperaldosteronism), and viriliza-
tion or feminization (excess testosterone or estradiol).
Cortical nodules in adrenal glands are not uncommon.
They are found incidentally in 2–9% of autopsies. With

CYTOPATHOLOGIC FEATURES
NODULAR CORTICAL HYPERPLASIA AND CORTICAL ADENOMA –
DISEASE FACT SHEET Cortical hyperplastic nodules and adenomas have
similar cytomorphology and cannot be distinguished in
Incidence
FNA specimens. Functional hyperactivity also cannot
៉ Cortical nodules in adrenal gland are not uncommon be determined by morphology. FNA usually yields
៉ Found incidentally in 2–9% of autopsies
cellular material. The cells are arranged mostly singly,
៉ With abdominal CT scanning, 1.3% to 3.4% of patients may be
found to have occult adrenal masses larger than 1 cm
and have round, uniform nuclei and vacuolated cyto-
plasm which is best seen in Romanowsky-based stains
Clinical Features (e.g. Diff-Quik). Anisonucleosis and intranuclear pseu-
៉ Hyperfunctional nodules accompanied by clinical syndromes (e.g.,
doinclusions may be present. Because of the delicate
Cushing’s, Conn’s, virilization of feminization) cytoplasm, some cells are stripped of their cytoplasm,
leaving bare nuclei in a ‘bubbly’ background (Fig.
10-36).

Ch010-F06731.indd 329 10/26/2006 10:33:58 AM


330 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-36
Nodular cortical hyperplasia and
cortical adenoma. Benign adrenal
tissue. Cortical hyperplasia and
cortical adenoma have the same
morphology, mostly bare nuclei in
a ‘bubbly’ background. Diff-Quik
stain, medium power.

distinction from metastatic carcinomas with clear


NODULAR CORTICAL HYPERPLASIA AND CORTICAL ADENOMA – cell features. Adrenal cortical cells, neoplastic or
PATHOLOGIC FEATURES non-neoplastic, react to anti-A103 (Melan A) and
anti-α-inhibin.
Cytopathologic Findings
៉ Cortical hyperplastic nodules and adenomas have similar
cytomorphology
៉ FNA usually yields cellular material DIFFERENTIAL DIAGNOSIS AND PITFALLS
៉ Mostly single cells with round, uniform nuclei and vacuolated
cytoplasm
៉ Anisonucleosis and intranuclear pseudoinclusions may be The differential diagnosis of adrenal cortical adenoma
present (ACA) usually involves adrenal cortical carcinoma
៉ Some cells, striped of their cytoplasm, appear as bare nuclei in (ACC) and metastatic tumors with clear cell features,
a ‘bubbly’ background
such as renal cell carcinoma (RCC) and, rarely, hepato-
cellular carcinoma (HCC). In rare instances, small clus-
Ancillary Studies
ters of bare nuclei of cortical cells may mimic metastatic
៉ Neoplastic or non-neoplastic adrenal cortical cells react to anti-
small cell carcinoma of lung.
A103 (Melan A) and anti-α-inhibin
Distinguishing between ACA and well-differentiated
Differential Diagnosis and Pitfalls
ACC can be very difficult without examining the entire
tumor. Tumor size, invasion, and number of mitotic
៉ Differential diagnosis of adrenal cortical adenoma (ACA)
includes adrenal cortical carcinoma (ACC), renal cell carcinoma
figures have been used as criteria for malignancy. Some
(RCC), and, rarely, hepatocellular carcinoma cortical adenomas, however, can be very large; and inva-
៉ Very rarely, small clusters of bare nuclei of cortical cells may sion and mitotic count cannot be assessed in FNA speci-
mimic small cell carcinoma mens. Certain cytopathologic features, such as larger
៉ Differential diagnosis between ACA and well-differentiated ACC nuclei with scattered bizarre forms, may raise the
cannot be made on FNA suspicion of ACC. Immunohistochemical stains in cell
៉ Immunohistochemical stains in cell blocks are helpful to blocks are helpful in the distinction from RCC and HCC.
differentiate ACA from HCC and RCC Immunostains for Melan A and inhibin are positive in
ACA, but negative in RCC and HCC. Furthermore,
CD10, which is positive in RCC, is negative in ACA.
Characteristic canalicular-type staining with antibodies
ANCILLARY STUDIES to polyclonal CEA and CD10 differentiates the HCC
from ACA. Bare nuclei mimicking small cell carcinoma
rarely causes a problem. The nuclei of ACA are round
Diagnosis in FNA specimens is usually made on routine with small nucleoli, and they do not mold. Small cell
preparations stained by Papanicolaou and Diff-Quik carcinomas show molding and often irregular nuclear
stains. Immunohistochemistry can be helpful in the borders, and they lack nucleoli. The neuroendocrine

Ch010-F06731.indd 330 10/26/2006 10:34:00 AM


CHAPTER 10 Kidney and Adrenal Gland 331

chromatin pattern in small cell carcinoma, if present, is CYTOPATHOLOGIC FEATURES


helpful, but due to degenerative changes, diffuse hyper-
chromasia or irregular chromatin distribution may also
be seen in small cell carcinoma. Fine needle aspirates of ACC are very cellular with
predominantly single cells and loose cellular groups;
capillary vasculature traversing the groups has been
described. Tumor cells have uniform to markedly pleo-
ADRENOCORTICAL CARCINOMA (ACC) morphic nuclei with coarse chromatin, depending upon
the differentiation of tumor (Figs 10-37 & 10-38).
Nucleoli are usually prominent. Multinucleation and
CLINICAL FEATURES bizarre nuclear forms are common in poorly differenti-
ated ACCs (Fig. 10-39). Cytoplasm is usually dense
Adrenocortical carcinoma is a rare tumor. In the litera- granular. Intranuclear pseudoinclusions may be present.
ture, an incidence of 1–4 per million is cited. It occurs Finely vacuolated (lipid-rich) cytoplasm may be present
predominantly in the fourth and fi fth decades of life in some tumors, but is rarely observed in FNA speci-
with another smaller peak in the first decade, and it is mens. Frequent mitotic figures, including atypical ones,
slightly more common (58.6%) in women. Approxi- and necrosis are commonly seen in poorly differenti-
mately 60% of ACCs are functional hypercortisolism ated tumors; but they are less frequently observed in
(e.g. Cushing syndrome) or Cushing syndrome with fine needle aspirates. Another feature, intracytoplas-
virilization. Hyperaldosteronism is very rare. The mic hyaline inclusions described in some of the ACCs,
tumors are usually very large: an average weight ranging is not seen in fine needle aspirates.
from 510 to 1200 g and an average size from 12.0 to
16.6 cm have been reported. Although the size of the
tumor is considered one of the indications of malig-
nancy, especially those larger than 6 cm, very large
ANCILLARY STUDIES
adenomas and small carcinomas may occur. The prog-
nosis of ACC is poor. The mortality rate in adults is ACCs react to antibodies for inhibin, Melan A, and
50–90%, with the majority of deaths occurring within calretinin. Immunostains for synaptophysin and CD56
2 years of diagnosis. Under 20 years of age, the worst are positive, but stain for chromogranin is negative.
survival rates occur in adolescents compared with
children under 5 years old. Metastases usually develop
within 10 years, the most common sites being liver,
lungs, and intra-abdominal or intrathoracic lymph
nodes.
ADRENOCORTICAL CARCINOMA (ACC) – PATHOLOGIC FEATURES

Cytopathologic Findings
៉ Very cellular specimens, predominantly single cells and loose
cellular groups
៉ Uniform to markedly pleomorphic nuclei with coarse chromatin,
usually prominent nucleoli; multinucleated bizarre nuclei are
ADRENOCORTICAL CARCINOMA (ACC) – DISEASE FACT SHEET common in poorly differentiated ACC
៉ Intranuclear pseudoinclusions may be present
Incidence ៉ Dense granular cytoplasm
៉ ACC is a rare tumor (incidence of 1 to 4 per million) ៉ Frequent mitotic figures, including atypical ones

Gender and Age Distribution Ancillary Studies


៉ Slightly more common (58.6%) in women ៉ ACC reacts to antibodies to inhibin, Melan A, and calretinin
៉ Occurs predominantly in the 4th and 5th decades of life, with ៉ Immunostains for synaptophysin and CD56 are positive, but
another smaller peak in the 1st decade stain for chromogranin is negative

Clinical Features Differential Diagnosis and Pitfalls


៉ Approximately 60% of ACCs are functional hypercortisolism or ៉ Differential diagnosis between ACC and ACA cannot be made in
Cushing syndrome with virilization FNA specimens alone
៉ Usually very large tumors: average weight, 510–1200 g; average ៉ Pheochromocytoma vs ACC morphologically can be similar
size, 12–16.6 cm ៉ Immunostains for inhibin, Melan A, calretinin, synaptophysin,
៉ Although the larger size is one of the indications of malignancy, chromogranin, and S-100 are helpful (see Table 10-4)
very large adenomas and small carcinomas may occur ៉ Among metastatic neoplasms, renal cell carcinoma (RCC) and
៉ Prognosis is poor; mortality in adults is 50–90% hepatocellular carcinoma (HCC) should be considered in the
៉ Metastases usually develop within 10 years differential diagnosis
៉ Most common sites of metastases are liver, lungs, and intra- ៉ Immunohistochemistry again is helpful in morphologically
abdominal or intrathoracic lymph nodes similar cases (see Tables 10-5 and 10-6)

Ch010-F06731.indd 331 10/26/2006 10:34:00 AM


332 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-37
Adrenocortical carcinoma. Large,
relatively uniform nuclei. Better
differentiated part of ACC. Papani-
colaou stain, medium power.

FIGURE 10-38
Adrenocortical carcinoma. Pleomor-
phic tumor cells with multinucle-
ated forms. Although there is
significant atypia, this still could
be a benign adenoma. Papanicolaou
stain, medium power.

DIFFERENTIAL DIAGNOSIS AND PITFALLS these features, perhaps with the exception of very
poorly differentiated ACCs, can be accurately assessed
in cytopathologic specimens.
The distinction between ACC and ACA cannot be made Among other primary adrenal neoplasms, pheochro-
with certainty in FNA specimens alone. Some ACAs mocytoma should be considered. Nuclear pleomorphism
may have significant cellular atypia, and some well- and intranuclear inclusions may be seen in pheochro-
differentiated ACCs could have the same cytomorphol- mocytomas. Immunohistochemistry in cell blocks is
ogy as ACAs. The exact nature of some of the cortical helpful in the differential diagnosis (Table 10-4). Immu-
neoplasms may not even be determined after examina- nostains for inhibin, Melan A (A103), and calretinin
tion of the resected tumor. The presence of metastasis are positive in ACC but negative in pheochromocyto-
and venous and/or capsular invasion is considered mas. Synaptophysin and CD56 are positive in both ACC
definitive evidence of malignancy. The number of and pheochromocytoma, but chromogranin is positive
mitoses, presence of atypical mitosis, and necrosis are in pheochromocytoma and negative in ACC. p53 also
stated as evidence of ACC in the literature. None of is positive in pheochromocytoma but negative in ACC.

Ch010-F06731.indd 332 10/26/2006 10:34:00 AM


CHAPTER 10 Kidney and Adrenal Gland 333

FIGURE 10-39
Adrenocortical carcinoma. Bizarre
nucleus in adrenocortical carci-
noma. Diff-Quik stain, high power.

TABLE 10-4
Immunohistochemistry of ACN and Pheochromocytoma

Inhibin A Melan A (A103) Calretinin Ad4BP Chromogranin P53 S-100

ACN + + + + − − −
PHEO. − − − − + + +*

ACN, adrenocortical neoplasms; PHEO., pheochromocytoma.


*Stains sustentacular cells.

TABLE 10-5
Immunohistochemistry of ACN and RCC

CD10 Melan A (A103) Inhibin A Calretinin Ad4BP EMA AE1/AE3 RCCa

ACN − + + + + − − −
RCC + − − − − + + +

ACN, adrenocortical neoplasms; RCC, renal cell carcinoma; RCCa, antibody for renal cell carcinoma.

S-100 stains sustentacular cells in pheochromocytomas, and EMA, and does not react to Melan A (A103),
but it is negative in ACC. Among metastatic neoplasms, inhibin, calretinin, and Ad4BP. Reactions to those anti-
renal cell carcinoma, hepatocellular carcinoma, and, bodies are reverse in ACC (Table 10-5).
rarely, other carcinomas may be considered in the Hepatocellular carcinoma may have similar cellular
differential diagnosis of ACC. The cytomorphology of morphology to ACC with acidophilic, granular or finely
renal cell carcinomas (e.g. grade 3 RCCs) may resemble vacuolated, lipid-rich cytoplasm and intranuclear
ACC. The latter, however, rarely have clear cell features inclusions. When present, well-defined tissue fragments
and frequently have focal marked atypia. Immunohisto- of atypical hepatocytes surrounded with sinusoidal
chemistry is helpful in the differential diagnosis. RCC endothelial cells, are characteristic for HCC, differen-
reacts to antibodies for CD10, RCC marker, AE1/AE3, tiating it from ACC. Most HCCs, usually moderately

Ch010-F06731.indd 333 10/26/2006 10:34:01 AM


334 FINE NEEDLE ASPIRATION CYTOLOGY

TABLE 10-6
Immunohistochemistry of ACN and HCC

Melan A (A103) Ad4BP Calretinin Inhibin CAM 5.2 HepPar-1 pCEA CD10

ACN + + + + − − ± −
HCC − − − − + + +* +*

ACN, adrenocortical neoplasms; HCC, hepatocellular carcinoma.


*Staining in canalicular pattern.

differentiated ones, tend to show uniform atypicality is incidentally discovered by imaging studies performed
with large round nuclei and single macronucleoli, on patients without any clinical evidence of cancer
without markedly atypical single cells or groups of cells (incidentalomas).
as seen focally in ACC. Immunohistochemistry is also
helpful (Table 10-6). Immunostains for Melan A,
Ad4BP, calretinin, and inhibin are positive for ACC but
negative for HCC. Immunostains for polyclonal CEA CYTOPATHOLOGIC FEATURES
and CD10 stain HCCs in a canalicular pattern, and
CD34 stains the sinusoidal endothelial cells surround-
ing tissue fragments (thick, abnormal trabeculae of the Most of the metastatic carcinomas are adenocarcino-
tumor). mas with varying degree of differentiation (Fig. 10-40).
Squamous cell carcinoma (Fig. 10-41), adenocarcinoma,
and small cell carcinoma (Fig. 10-42) usually have the
characteristic features seen in the primary site. Lym-
METASTATIC NEOPLASMS phomas secondarily involve adrenal glands (Fig. 10-
43). They are found in 18% to 25% of patients with
disseminated lymphomas at autopsy. The specific diag-
CLINICAL FEATURES nosis of most of the metastatic tumors can be made by
correlating clinical and microscopic findings, in certain
cases, with the help of ancillary techniques.
The majority of the malignant neoplasms of adrenal
gland are metastatic tumors. The adrenal is the fourth
most common site of metastases after lung, liver, and
bone. About one-third of FNAs of adrenal gland are DIFFERENTIAL DIAGNOSIS AND PITFALLS
metastatic carcinomas. Lung and breast are reported to
be the most common primary sites. Data published in
recent years indicate that lung carcinoma is the leading One of the major diagnostic challenges is distinguish-
type (up to 67.9%) of metastatic cancer diagnosed by ing metastatic clear cell carcinomas from primary
FNAs, and non-small cell carcinomas, specifically adrenal cortical hyperplasia or neoplasms. Renal cell
adenocarcinoma, are the most common type. Other carcinomas are the most common type of clear cell
primary sites include kidney, gastrointestinal tract,
pancreas, liver, and ovary. Bilateral involvement is
common (30% to 49% in recently reported series). In
most cases, patients have known cancers of the other METASTATIC NEOPLASMS – PATHOLOGIC FEATURES
sites; in rare instances, however, the metastatic tumor
Cytopathologic Findings
៉ Most metastatic carcinomas are adenocarcinomas
៉ Metastatic tumors present varying cytomorphology depending
on the type of the tumor
METASTATIC NEOPLASMS – DISEASE FACT SHEET
Differential Diagnosis and Pitfalls
Clinical Features ៉ Most of the metastatic tumors can be differentiated from
៉ Majority of malignant neoplasms of adrenal gland are metastatic primary adrenal neoplasms without difficulty
tumors ៉ In morphologically similar cases of primary neoplasms of
៉ Lung and breast are the most common primary sites adrenal glands, immunohistochemistry helps in the differential
៉ Bilateral involvement is common diagnosis (see Tables 10-4–10-6)

Ch010-F06731.indd 334 10/26/2006 10:34:02 AM


CHAPTER 10 Kidney and Adrenal Gland 335

FIGURE 10-40
Metastatic neoplasms: adenocarci-
noma from lung. Fragment of tumor
with large nuclei, macronucleoli,
and cytoplasmic vacuoles charac-
teristic of adenocarcinoma. Papani-
colaou stain, medium power.

FIGURE 10-41
Metastatic neoplasms: squamous
cell carcinoma from esophagus.
Atypical keratinized squamous cells
with hyperchromatic nuclei. One
cell shows sharp angulation of the
nucleus, which is characteristic of
malignancy. Papanicolaou stain,
medium power.

carcinoma. Well-differentiated RCCs (grades 1 and 2) A, inhibin A, and Ad4BP. Metastatic hepatocellular
are morphologically indistinguishable from adrenal carcinomas to adrenal gland have the typical canalicular
cortical hyperplasia or cortical adenomas. Well- pattern with pCEA and CD10. Metastatic adenocarcino-
differentiated ACCs have uniform cell populations mas are usually identifiable by their characteristic cyto-
with eccentric nuclei and scattered bizarre cells, with morphology. Unlike adrenal cortical neoplasms, they
large pleomorphic, single or multiple nucleoli. Clear cell react to antibodies for cytokeratin and EMA. In cases
RCC has a uniform cell population with usually cen- of unknown primary sites, CK7, CK20, and TTF-1 are
trally located nuclei and finely vacuolated cytoplasm. helpful in suggesting the primary sites, specifically lung
Immunohistochemistry is helpful in differentiating or gastrointestinal tract, the carcinomas of which are
adrenal cortical neoplasms from RCC (Table 10-5). likely to metastasize to the adrenal gland.
Adrenal cortical neoplasms react to monoclonal Melanomas may mimic adrenal cortical carcinomas
antibody A103 (Melan A) and anti-α-inhibin, but do not and both react to Melan A. Other melanoma markers,
react to CD10. Clear cell RCCs react to antibodies for HMB-45 and S-100, however, are negative in adrenal
CD10, EMA, and AE1/AE3, but do not react to Melan cortical carcinomas (Figs 10-44 & 10-45).

Ch010-F06731.indd 335 10/26/2006 10:34:02 AM


336 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-42
Metastatic neoplasms: small cell
carcinoma from lung. Undifferenti-
ated malignant tumor cells with
very high N/C ratio and nuclear
molding. Some nuclei show a stip-
pled chromatin pattern, which is
typical of neuroendocrine differen-
tiation. Papanicolaou stain, high
power.

FIGURE 10-43
Metastatic neoplasms: large B cell
lymphoma. Large, round, single
cells. This was confirmed by pheno-
typing studies. Diff-Quik stain,
medium power.

PHEOCHROMOCYTOMA in both sexes. The peak age is at the fi fth decade, but
it may occur at any age. More than 90% of the pheo-
chromocytomas are sporadic. Between 5% and 10% of
CLINICAL FEATURES these tumors occur at extramedullary sites. About 5%
of sporadic pheochromocytomas and 50% of familial
pheochromocytomas are bilateral. In the pediatric age
Pheochromocytoma is a rare tumor with an estimated group, an increased number of cases occur bilaterally
incidence in the United States of 8 per 1 million person- and at extramedullary sites. Approximately 5–10% of
years. Studies in other countries have shown an inci- pheochromocytomas are familial. They are usually a
dence of about 2 per million. It occurs roughly equally part of one of the multiple endocrine neoplasia (MEN)

Ch010-F06731.indd 336 10/26/2006 10:34:03 AM


CHAPTER 10 Kidney and Adrenal Gland 337

FIGURE 10-44
Metastatic neoplasms: melanoma.
This is the characteristic appear-
ance of melanoma. Predominantly
single cells, some with eccentrically
located nuclei. Other cells have
cytoplasmic extensions. Diff-Quik
stain, medium power.

FIGURE 10-45
Metastatic neoplasms: melanoma.
Positive immunostaining with
HMB-45. High power.

syndromes, MEN IIA or MEN IIB. MEN IIA (Sipple detected in urine and plasma. The majority of pheo-
syndrome) includes varying combinations of pheochro- chromocytomas can be diagnosed based on combined
mocytoma, medullary carcinoma of thyroid, and para- clinical, radiologic, and biochemical studies. FNA is not
thyroid hyperplasia. Pheochromocytomas occur in performed in cases of clinically diagnosed or suspected
50% of the cases with this syndrome. MEN IIB includes pheochromocytomas, because of potentially serious
pheochromocytoma, medullary thyroid carcinoma, complications.
mucosal neuromas involving lips, tongue, and other Clinically malignant pheochromocytomas are rare.
mucosal surfaces, and ganglioneuromatosis of the gas- The incidence of malignancy varies from 2.4% to 14%
trointestinal tract. More than one-third of pheochro- in published series. The 5-year survival rate in two
mocytomas are clinically unsuspected. In the great series was reported as 44% and 53%. Hematogenous
majority of cases, there is an increased level of vanil- and lymphatic metastasis may occur. Liver, lymph
lylmandelic acid (VMA) and catecholamines can be nodes, and bone are the common sites for metastasis.

Ch010-F06731.indd 337 10/26/2006 10:34:04 AM


338 FINE NEEDLE ASPIRATION CYTOLOGY

PHEOCHROMOCYTOMA – DISEASE FACT SHEET PHEOCHROMOCYTOMA – PATHOLOGIC FEATURES

Incidence Cytopathologic Findings


៉ Pheochromocytoma is a rare tumor ៉ Hypercellular specimens with loose groups or single cells
៉ Estimated incidence in the United States is 8 per 1 million ៉ Tumor cells vary in size and shape, usually have large, ill-
person-years defined cytoplasm
៉ Binucleated and multinucleated cells are present
Gender and Age Distribution ៉ Smaller and more uniform tumor cells and rare intranuclear
៉ Occurs roughly equally in both sexes pseudoinclusions may be seen
៉ Peak age is at the 5th decade, but may occur at any age
Ancillary Studies
Clinical Features ៉ Immunohistochemistry:
៉ More than 90% are sporadic ៉ Synaptophysin, chromogranin, and NSE are positive
៉ 5–10% occur at extramedullary sites ៉ S-100 stains the sustentacular cells
៉ About 5% of sporadic pheochromocytomas and 50% of familial ៉ Electron microscopy: numerous electron-dense neurosecretory-
pheochromocytomas are bilateral type granules present
៉ Approximately 5–10% are familial and usually a part of the
multiple endocrine neoplasia syndromes (MEN IIA or MEN IIB) Differential Diagnosis and Pitfalls
៉ More than one-third of pheochromocytomas are clinically ៉ Differential diagnosis includes adrenocortical carcinoma (ACC),
unsuspected sarcomatoid renal cell carcinoma, and, rarely, retroperitoneal
៉ Clinically malignant pheochromocytomas are rare; incidence of sarcomas
malignancy varies from 2.4% to 14% ៉ Pheochromocytoma and ACC have some common
៉ Common metastatic sites are liver, lymph nodes, and bone cytomorphologic features
៉ Immunohistochemistry is helpful in the differential diagnosis
(see Table 10-4)

CYTOPATHOLOGIC FEATURES

FNA specimens are hypercellular with loose groups or cytoma and ACC have some common cytopathologic
single cells (Fig. 10-46). Tumor cells show a broad spec- features, such as nuclear pleomorphism and intra-
trum of changes ranging from large cells with multiple nuclear pseudoinclusions, and differential diagnosis
nucleoli to smaller, more monotonous cellular compo- can be difficult based on morphology alone. Immuno-
nent. The cells vary in size and shape, and usually have histochemistry is helpful in these cases (Table 10-6).
large polygonal or ill-defined cytoplasm. Binucleated Although both ACC and pheochromocytoma react to
or multinucleated cells are present. Smaller and more antibodies to synaptophysin, immunostain for chromo-
uniform tumor cells and rare intranuclear pseudoinclu- granin is positive only in pheochromocytomas. Fur-
sions may also be seen. Intracytoplasmic hyalin inclu- thermore, stains for inhibin and Melan A are positive
sions found in histologic specimens are not usually in ACC and negative in pheochromocytes. Positive
observed in FNA specimens. Spindle cell forms can be staining for neuroendocrine markers differentiates
seen. Some tumor cells have a bland chromatin pattern pheochromocytoma from sarcomatoid RCC and retro-
with prominent nucleoli. Rare pigmented (lipofuscin or peritoneal sarcomas.
melanin) forms have been reported. The spectrum of
the cytomorphology of pheochromocytoma can be seen
in Figures 10-47 to 10-49. NEUROBLASTOMA AND
GANGLIONEUROBLASTOMA
ANCILLARY STUDIES
CLINICAL FEATURES
Immunohistochemical stains for synaptophysin, chro-
mogranin (Fig. 10-50), and NSE are positive. S-100 Neuroblastoma is the fourth most common malignant
stains the sustentacular cells. neoplasm of the pediatric age group, following leuke-
With electron microscopy, numerous electron-dense, mias, lymphomas, and brain tumors. The estimated
neurosecretory-type granules are seen. incidence of neuroblastoma is 8.7 per 1 million; and for
ganglioneuroblastoma, it is 6.8 per 1 million. Most neu-
roblastomas occur in children under the age of 5 years.
DIFFERENTIAL DIAGNOSIS AND PITFALLS Occurrence in adulthood is rare. Adrenal is the most
common site for neuroblastoma, followed by abdomen
and thoracic cavity. The tumor is almost always soli-
Differential diagnosis includes ACC, sarcomatoid RCC, tary. Age and stage are the most significant prognostic
and, rarely, retroperitoneal sarcomas. Pheochromo- factors. However, some histologic grading systems

Ch010-F06731.indd 338 10/26/2006 10:34:05 AM


CHAPTER 10 Kidney and Adrenal Gland 339

FIGURE 10-46
Pheochromocytoma. Tumor cells
occur singly or in loose groups.
Scattered multinucleated cells are
also present. Diff-Quik stain, low
power.

FIGURE 10-47
Pheochromocytoma. Large cells with
single or multiple nuclei. Cytomor-
phology characteristic of pheochro-
mocytoma. Diff-Quik stain, medium
power.

Ch010-F06731.indd 339 10/26/2006 10:34:05 AM


340 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-48
Pheochromocytoma. Larger cells
with single or double nuclei and
prominent nucleoli represent gan-
glion cell differentiation. The fusi-
form nuclei in the background are
probably sustentacular cells. Papa-
nicolaou stain, high power.

FIGURE 10-49
Pheochromocytoma. Smaller cells
with eccentric nuclei and scattered
larger cells. There is a similarity
between this cytomorphology and
that of melanoma. Papanicolaou
stain, medium power.

Ch010-F06731.indd 340 10/26/2006 10:34:07 AM


CHAPTER 10 Kidney and Adrenal Gland 341

FIGURE 10-50
Pheochromocytoma. Immunohis-
tochemical stains of cell block –
positive for chromogranin. Medium
power.

distributed. Nucleoli are inconspicuous or absent.


NEUROBLASTOMA AND GANGLIONEUROBLASTOMA – Occasional cells may show unipolar extension of cyto-
DISEASE FACT SHEET plasm (Fig. 10-51B). A fibrillar background resembling
neutrophils, present in histologic preparations, can
Incidence also be seen in both Diff-Quik- and Papanicolaou-
៉ Neuroblastoma is the fourth most common malignant neoplasm of stained cytologic preparations. In some cases, neoplas-
the pediatric age group tic cells form Homer Wright rosettes surrounding
៉ Estimated incidence of neuroblastoma is 8.7 per 1 million, and round areas of fibrillary material. The fibrillary back-
for ganglioneuroblastoma is 6.8 per 1 million
ground is characteristic for neuroblastomas and helpful
in the differential diagnosis. Ganglion cell differentia-
Clinical Features
tion is characterized by larger and denser cytoplasm
៉ Most tumors occur in children under the age of 5 years
and large nuclei with prominent nucleoli. Ganglioneu-
៉ Adrenal is the most common site, followed by abdomen and
thoracic cavity
roblastomas are composed of predominantly neoplastic
៉ Tumor is almost always solitary ganglion cells with large, dense cytoplasm and multiple
៉ Age and stage are the most significant prognostic factors; some nuclei with prominent nucleoli.
histologic grading systems linked to age have been used

ANCILLARY STUDIES
linked to age have been shown to be effective in predict-
ing the prognosis. Immunostain for NSE is consistently positive (Fig.
10-52). Neuroblastomas and ganglioneuroblastomas
also react to stains for neurofi lament proteins and
microtubule-associated proteins (MAP1 and MAP2),
CYTOPATHOLOGIC FEATURES and α and β tubulin.

FNA specimens usually are hypercellular and com-


posed of small cells with high N/C ratio. Tumor cells DIFFERENTIAL DIAGNOSIS AND PITFALLS
appear singly or in groups. Areas of nuclear molding
may be seen. The nuclei vary slightly in size and are
round, oval, or slightly irregular in shape (Fig. 10-51A). Distinguishing neuroblastoma from other small round
Nuclear chromatin is finely granular and evenly cell tumors of the pediatric age group – specifically,

Ch010-F06731.indd 341 10/26/2006 10:34:08 AM


342 FINE NEEDLE ASPIRATION CYTOLOGY

A B

FIGURE 10-51
Neuroblastoma. Hypercellular specimen with predominantly single small cells and high N/C ratio. Occasional cells with unipolar cytoplasmic exten-
sions are present. A, Diff-Quik stain, low power; B, Papanicolaou stain, high power.

NEUROBLASTOMA AND GANGLIONEUROBLASTOMA –


PATHOLOGIC FEATURES

Cytopathologic Findings
៉ FNA samples show small cells with high N/C ratio, appearing
singly or in groups
៉ Areas of nuclear molding are present
៉ Nuclei are round, oval, or slightly irregular in shape and vary in
size
៉ Nuclear chromatin is finely granular and evenly distributed
៉ Nucleoli are inconspicuous or absent
៉ Occasional cells may show unipolar cytoplasmic extensions
៉ A fibrillar background resembling neutrophils can be seen
៉ Homer Wright rosettes may be present
៉ Ganglion cell differentiation characterized by larger and denser
cytoplasm and large nuclei with prominent nucleoli may be
present in varying proportions
៉ Ganglioneuroblastomas are composed predominantly of
neoplastic ganglion cells FIGURE 10-52
Neuroblastoma. Immunostain of cell block – positive for NSE. High
Ancillary Studies power.
៉ Immunostains for NSE consistently positive

Differential Diagnosis and Pitfalls


៉ Differential diagnoses include other small round cell tumors of
the pediatric age group nephroblastoma, lymphomas, PNET/Ewing sarcoma,
៉ Additional aspirations for flow cytometry and cell blocks, as and rhabdomyosarcoma – often requires the use of
well as a core biopsy are usually needed ancillary techniques. Additional aspirations for flow
៉ In some cases, material for electron microscopy and cytogenetic cytometry and cell blocks, as well as a core biopsy, are
studies is also required for diagnosis usually needed. In some cases, material for electron
៉ NSE differentiates neuroblastoma from other tumors except for microscopy and cytogenetic studies is also required.
PNET, which may react to stain for CD99, which is negative in Positive staining for NSE differentiates neuroblastoma
neuroblastoma from others except for PNET. The latter may react to
៉ Lymphomas and rhabdomyosarcoma can be identified with
stain for CD99, which is negative in neuroblastoma.
lymphoma and muscle markers
Lymphomas and rhabdomyosarcoma can be identified
with lymphoma and muscle markers.

Ch010-F06731.indd 342 10/26/2006 10:34:09 AM


CHAPTER 10 Kidney and Adrenal Gland 343

FIGURE 10-53
Ganglioneuroma. Schwann cells.
Papanicolaou stain, medium power.

GANGLIONEUROMA CYTOPATHOLOGIC FEATURES

CLINICAL FEATURES Mature ganglion cells and an abundant spindle cell


(Schwann cells) background are seen (Figs 10-53 &
10-54). The proportion of ganglion cells varies. Some
Ganglioneuroma is a benign tumor, usually occurring tumors have predominantly spindle cells.
in the posterior mediastinum, less frequently in adrenal
glands. The age range is 6 to 20 years, and it occurs
more commonly in females.
DIFFERENTIAL DIAGNOSIS AND PITFALLS

Because of the paucity of ganglion cells in some tumors,


FNA samples contain only Schwann cells and can be
misdiagnosed as neurofibroma.

GANGLIONEUROMA – DISEASE FACT SHEET


GANGLIONEUROMA – PATHOLOGIC FEATURES
Incidence
៉ A benign tumor, usually in the posterior mediastinum, less Cytopathologic Findings
frequently in adrenal glands ៉ Ganglion cells in an abundant Schwann cell background
៉ Proportion of ganglion cells and Schwann cells varies
Gender and Age Distribution
៉ More common in females Differential Diagnosis and Pitfalls
៉ Age range is 6 to 20 years ៉ Due to sampling error, may be misdiagnosed as neurofibroma

Ch010-F06731.indd 343 10/26/2006 10:34:09 AM


344 FINE NEEDLE ASPIRATION CYTOLOGY

FIGURE 10-54
Ganglioneuroma. Ganglion cells.
Diff-Quik stain, high power.

SUGGESTED READINGS Renshaw AA, Lee KR, Madge R, Granter SR. Accuracy of fine needle aspira-
tion in distinguishing subtypes of renal cell carcinoma. Acta Cytol
Kidney 1997;41:987–994.
Simsir A, Chhieng D, Wei XJ, Yee H, Waisman J, Cangiarella J. Utility of
General
CD10 and RCCma in the diagnosis of metastatic conventional renal-cell
Brierly RD, Thomas PJ, Harrison NW, Fletcher MS, Nawrocki JD, Ashton-Key adenocarcinoma by fine needle aspiration biopsy. Diagn Cytopathol
M. Evaluation of fine-needle aspiration cytology for renal masses. BJU 2005;33:3–7.
Int 2000;85:14–18.
Eble JN, Sauter G, Epstein JI, Sesterhenn IA, eds. World Health Organiza- Renal Cell Carcinoma, Papillary Type
tion Classification of Tumors: Pathology and Genetics of Tumors of the
Urinary System and Male Genital Organs. Lyon, France: IARC Press, Dekmezian R, Sneige N, Shabb N. Papillary renal-cell carcinoma: fine-
2004. needle aspiration of 15 cases. Diagn Cytopathol 1991;7:198–203.
Geisinger KR, Stanley MW, Raab SS, Silverman JF, Abati A. Kidney. In: Wang S, Filipowicz EA, Schnadig VJ. Abundant intracytoplasmic hemosid-
Modern Cytopathology. New York: Churchill Livingstone, 2004:529– erin in both histiocytes and neoplastic cells: a diagnostic pitfall in
618. fine-needle aspiration of papillary renal-cell carcinoma. Diagn Cytopa-
Murphy WM, Grignon DJ, Perlman EJ. Tumors of the Kidney, Bladder thol 2001;24:82–85.
and Related Urinary Structures. Atlas of Tumor Pathology, 4th series,
Fascicle 1. Washington, DC: Armed Forces Institute of Pathology,
2004. Renal Cell Carcinoma, Medullary Type
Truong LD, Todd TD, Dhurandhar B, Ramzy I. Fine-needle aspiration of Assad L, Resetkova E, Oliveira VL, et al. Cytologic features of renal medul-
renal masses in adults: analysis of results and diagnostic problems in lary carcinoma. Cancer (Cancer Cytopathol) 2005;105:28–34.
108 cases. Diagn Cytopathol 1999;20:339–349. Larson DM, Gilstad CW, Manson GW, Henry MR. Renal medullary carcinoma:
report of a case with positive urine cytology. Diagn Cytopathol 1998;18:
276–279.
Renal Cell Carcinoma, Clear Cell (Conventional) Type Qi J, Shen PU, Rezuki WN, Currier AA, Westfall PK, Mandavilli SR. Fine
Al Nazer M, Mourad WA. Successful grading of renal-cell carcinoma in needle aspiration cytology diagnosis of renal medullary carcinoma: a
fine-needle aspirates. Diagn Cytopathol 2000;22:223–226. case report. Acta Cytol 2001;45:735–739.
Cheville JC, Lohse CM, Zincke H, Weaver AL, Blute ML. Comparisons of
outcome and prognostic features among histologic subtypes of renal Renal Cell Carcinoma, Chromophobe Cell Type
cell carcinoma. Am J Surg Pathol 2003;27:612–624.
Ficarra V, Martignoni G, Maffei N, et al. Original and reviewed nuclear Grunter SR, Renshaw AA. Fine-needle aspiration of chromophobe renal cell
grading according to the Fuhrman system: a multivariate analysis of carcinoma. Analysis of six cases. Cancer 1997;81:122–128.
388 patients with conventional renal cell carcinoma. Cancer 2005;103: Peyromaure M, Misrai V, Thiounn N, et al. Chromophobe renal cell carci-
68–75. noma: analysis of 61 cases. Cancer 2004;100:1406–1410.
McGregor DK, Khurana KK, Cao C, et al. Diagnosing primary and metastatic Wang HY, Mills SE. KIT and RCC are useful in distinguishing chromophobe
renal cell carcinoma. The use of monoclonal antibody ‘renal cell carci- renal cell carcinoma from the granular variant of clear renal cell carci-
noma marker.’ Am J Surg Pathol 2001;25:1485–1492. noma. Am J Surg Pathol 2005;29:540–546.
Nayar R, Bourtsos E, DeFrias EV. Hyaline globules in renal cell carcinoma Wiatrowska BA, Zakowski MF. Fine-needle aspiration biopsy of chromo-
and hepatocellular carcinoma. A clue or a diagnostic pitfall on fine- phobe renal cell carcinoma and oncocytoma: comparison of cytomor-
needle aspiration? Am J Clin Pathol 2000;114:576–582. phologic features. Cancer 1999;87:161–167.

Ch010-F06731.indd 344 10/26/2006 10:34:10 AM


CHAPTER 10 Kidney and Adrenal Gland 345

Renal Cell Carcinoma, Collecting Duct Type Wadih GE, Raab SS, Silverman JF. Fine needle aspiration cytology of renal
and retroperitoneal angiomyolipoma. Report of two cases with cytologic
Caraway NP, Wojcik EM, Katz RL, Ro RY, Ordonez NG. Cytologic findings findings and clinicopathologic pitfalls in diagnosis. Acta Cytol
of collecting duct carcinoma of kidney. Diagn Cytopathol 1995;13: 1995;39:945–950.
304–309.
Layfield LJ. Fine-needle aspiration biopsy of renal collecting duct carci-
noma. Diagn Cytopathol 1994;48:843–848. Cystic Lesions
Ono K, Nishino E, Nakamine H. Renal collecting duct carcinoma. Report
of a case with cytologic findings on fine needle aspiration. Acta Cytol Horwitz CA, Manivel JC, Inampudi S, Kaye K. Diagnostic difficulties in the
2000;44:380–384. interpretation of needle aspiration material from large renal cysts.
Sarode VR, Islam S, Wooten D, Watamull LM, Molberg K, Ashfaq R. Fine Diagn Cytopathol 1994;11:380–383.
needle aspiration of collecting duct carcinoma of kidney: report of a Katz LB, Ehya H. Liesegang rings in renal cyst fluid. Diagn Cytopathol
case with distinctive features and differential diagnosis. Acta Cytol 1990;6:197–200.
2004;48:843–848. Raso DS, Greene WB, Finley JL, Silverman JF. Morphology and pathogen-
esis of Liesegang rings in cyst aspirates: report of two cases with
ancillary studies. Diagn Cytopathol 1998;19:116–119.
Sarcomatous Renal Cell Carcinoma Todd TD, Dhurandhar B, Mody D, Ramzy I, Truong LD. Fine needle aspira-
tions of the cystic lesions of the kidney. Morphologic spectrum and
Auger M, Katz RL, Sella A, Ordonez RE, Thung SN, Szporn A. Fine needle diagnostic problems in 41 cases. Am J Clin Pathol 1999;111:317–328.
aspiration cytology of sarcomatoid renal cell carcinoma: a morphologic
and immunocytochemical study of 15 cases. Diagn Cytopathol 1993;9:
46–51. Urothelial Carcinoma of Kidney Pelvis
Cheville JC, Lohse CM, Zincke H, et al. Sarcomatoid renal cell carcinoma:
an examination of underlying histologic subtype and an analysis of Olgac S, Mazumdar M, Dalbagni G, Reuter VE. Urothelial carcinoma of the
associations with patient outcome. Am J Surg Pathol 2004;28:435– renal pelvis: a clinicocytopathologic study of 130 cases. Am J Surg
441. Pathol 2004;28:1545–1552.
Wadih GE, Raab SS, Silverman JF. Fine needle aspiration cytology of renal
and retroperitoneal angiomyolipoma. Report of two cases with cytologic
findings and clinico-pathologic pitfalls in diagnosis. Acta Cytol Lymphoma
1995;39:945–950.
Truong LD, Caraway N, Ngo T, Laucirica R, Katz R, Ramzy I. Renal lym-
Wang J, Weiss LM, Hu B, et al. Usefulness of immunohistochemistry in
phoma. The diagnostic and therapeutic roles of fine-needle aspiration.
delineating renal spindle cell tumours. A retrospective study of 31
Am J Clin Pathol 2001;115:18–31.
cases. Histopathology 2004;44:462–471.

Metastatic Neoplasms
Oncocytoma
Gattuso P, Ramzy I, Truong LD, et al. Utilization of fine-needle aspiration
Caputo V, Repetti ML, Bordoni U, Radice F. Preoperative diagnosis of renal in the diagnosis of metastatic tumors to kidney. Diagn Cytopathol
oncocytoma by fine needle aspiration (FNA). Cytopathology 1996;7: 1999;21:35–38.
366–371.
Huo L, Sugimura J, Tretiakova MS, et al. C-kit expression in renal onco-
cytoma and chromophobe renal cell carcinomas. Hum Pathol 2005;
36:262–268. Adrenal Gland
Liu J, Fanning CV. Can renal oncocytoma be distinguished from renal cell General
carcinoma on fine-needle aspiration specimens? A study of conventional
smears in conjunction with ancillary studies. Cancer (Cancer Cytopa- deAugustin P, Lopez-Rios F, Alberti N, Perez-Barrios A. Fine-needle aspira-
thol) 2001;93:390–397. tion biopsy of the adrenal glands: a ten-year experience. Diagn Cyto-
pathol 1999;21:92–97.
Geisinger KR, Stanley MW, Raab SS, Silverman JF, Aboti A. Adrenal gland.
Nephroblastoma (Wilms Tumor) In: Modern Cytopathology. New York: Churchill Livingstone, 2004:
619–624.
Serrano R, Rodriguez-Peralto JL, DeOrbe GG, Melero C, de Agustin P. Harisinghani MG, Maher MM, Gervais DA, et al. Incidence of malignancy
Intrarenal neuroblastoma diagnosis by fine-needle aspirations: a report in complex cystic renal masses (Bosniak category III): should imaging-
of two cases. Diagn Cytopathol 2002;27:294–297. guided biopsy precede surgery? AJR Am J Roentgenol 2003;181:
Sharifah NA. Fine needle aspiration cytology characteristics of renal 1425–1426.
tumors in children. Pathology 1994;26:359–364. Lack EE. The adrenal gland. In Silverberg SC, ed. Principles and Practice
of Surgical Pathology and Cytopathology. New York: Churchill Livings-
ton, 1997:2751–2799.
Rhabdoid Tumor Zaman MB. The adrenals. In Koss LG, ed. Koss’ Diagnostic Cytopathology
and Its Histopathologic Bases, 5th ed. Philadelphia: Lippincott Williams
Barroca HM, Costa MJ, Carvalho JL. Cytologic profile of rhabdoid tumor and Wilkins, 2005:1482–1494.
of kidney. A report of 3 cases. Acta Cytol 2003;47:1055–1058.
Wakely PE Jr, Giacomantonio M. Fine needle aspiration cytology of meta-
static rhabdoid tumor. Acta Cytol 1986;30:533–537. Incidentalomas
Kasperlik-Zeluska AA, Roslonowska E, Slowinska-Srzeclnicka J, et al. Inci-
Angiomyolipoma dentally discovered adrenal mass (incidentaloma): investigation and
management of 208 patients. Clin Endocrinol (Oxf) 1997;46:29–37.
Glento JA, Partoft S. Ultrasound-guided percutaneous aspiration of renal Lumachi F, Borsato S, Tregnaghi A, et al. CT-scan, MRI and image-guided
angiomyolipoma: report of two cases diagnosed by cytology. Acta Cytol FNA cytology of incidental adrenal masses. EJSO 2003;29:689–692.
1984;28:265–268.
Kulkarni B, Desai SB, Dave B, Tongaonkar HB, Kulkarni JN, Chinoy RF.
Renal angiomyolipomas: a study of 18 cases. Indian J Pathol Microbiol Complications of FNA
2005;48:459–463.
Mai KT, Yazdi HM, Perkins DG, Thijssen A. Fine needle aspiration biopsy Mody MK, Kazerooni EA, Korobkin M. Percutaneous CT-guided biopsy of
of epithelioid angiomyolipoma. A case report. Acta Cytol 2001;45: adrenal masses: immediate and delayed complications. J Comput Assist
233–236. Tomogr 1995;19:434–439.

Ch010-F06731.indd 345 10/26/2006 10:34:11 AM


346 FINE NEEDLE ASPIRATION CYTOLOGY

EUS-Guided FNA Saboorian MH, Katz RL, Charsangavec C. Fine needle aspiration cytology
of primary and metastatic lesions of the adrenal gland. A series of 188
Eloubeidi MA, Seewald S, Tamhane A, et al. EUS-guided FNA of the left biopsies with radiologic correlation. Acta Cytol 1995;39:843–851.
adrenal gland in patients with thoracic or GI malignancies. Gastrointest Sharma S, Singh R, Verma K. Cytomorphology of adrenocortical carcinoma
Endosc 2004;59:627–633. and comparison with renal cell carcinoma. Acta Cytol 1997;41:
Jhala NC, Jhala D, Eloubedi MA, et al. Endoscopic ultrasound-guided fine- 385–392.
needle aspiration biopsy of the adrenal glands: analysis of 24 patients. Suen KC, McNeely TB. Adrenal cortical cells mimicking small cell anaplastic
Cancer 2004;102:308–314. carcinoma in a fine needle aspirate. Mod Pathol 1991;4:594–595.
Stelow EB, Debol SM, Stanley MW, Mallery S, Lai R, Bardales RH. Sampling Wadih GE, Nance KV, Silverman JF. Fine-needle aspiration cytology of the
of the adrenal glands by endoscopic ultrasound-guided fine-needle adrenal gland. Fifty biopsies in 48 patients. Arch Pathol Lab Med
aspiration. Diagn Cytopathol 2005;33:26–30. 1992;116:841–846.
Varadarajulu S, Fraig M, Schmulewitz N, et al. Comparison of EUS-guided Walther MM, Keiser HR, Linehan WM. Pheochromocytoma: evaluation,
19-gauge Trucut needle biopsy with EUS-guided fine-needle aspiration. diagnosis, and treatment. World J Urol 1999;17:35–39.
Endoscopy 2004;36:397–401.

Metastatic Neoplasms
Adrenal Cysts
Candel AG, Gattuso P, Reyes CV, Prinz RA, Castelli MJ. Fine-needle aspira-
Akcay MN, Akcay G, Balik AA, Boyuk A. Hydatid cysts of adrenal gland:
tion biopsy of adrenal masses in patients with extraadrenal malignan-
review of nine patients. World J Surg 2004;28:97–99.
cies. Surgery 1993;114:1132–1136.
deBree E, Schoretsanitis G, Melissas J, Christodoulakis M, Tsiftsis D. Cysts
Kocijancic K, Kocijancic I, Guna F. Role of sonographically guided fine-
of the adrenal gland: diagnosis and management. Int Urol Nephrol
needle aspiration biopsy of adrenal masses in patients with lung cancer.
1998;30:369–376.
J Clin Ultrasound 2004;32:12–16.
Ericksen LA, Lloyd RV, Harman R, Thompson G. Cystic adrenal neoplasms.
Lack EE. Tumors metastatic to adrenal glands. In: Tumors of the Adrenal
Cancer 2004;101:1537–1544.
Gland and Extra-adrenal Paraganglia. Atlas of Tumor Pathology. Wash-
Tung GA, Pfister RC, Papanicolaou N, Yoder JC. Adrenal cysts: imaging and
ington, DC: Armed Forces Institute of Pathology; 1997:199–212.
percutaneous aspirations. Radiology 1989;173:107–110.
Lam KY, Lo CY. Metastatic tumors of the adrenal glands: a 30-year
experience in a teaching hospital. Clin Endocrinol (Oxf) 2002;56:95–
101.
Myelolipoma
Meyer A, Behrend M. Presentation and therapy of myelolipoma. Int J Urol
2005;12:239–243. Immunohistochemistry
Settakorn J, Sirivanichai C, Rangdaeng S, Chaivun B. Fine-needle aspira-
tion cytology of adrenal myelolipoma: case report and review of litera- Fetsch PA, Powers CN, Zakowski MF, Abati A. Anti-alpha inhibin: marker
ture. Diagn Cytopathol 1999;21:4009–4012. of choice for the consistent distinction between adrenocortical carci-
noma and renal cell carcinoma in fine-needle aspiration. Cancer (Cancer
Cytopathol) 1999;87:168–172.
Primary Neoplasms Jorda M, De MB, Nadji M. Calretinin and inhibin are useful in separating
adrenocortical neoplasms from pheochromocytoma. Appl Immunohisto-
Handa U, Khullar U, Mohan H. Pigmented pheochromocytoma: report of chem Mol Morphol 2002;10:67–70.
a case with diagnosis by fine needle aspiration. Acta Cytol 2005;49: Sasano H, Shizawa S, Suzuki T, et al. Transcription factor adrenal 4 binding
421–423. protein as a marker of adrenocortical malignancy. Hum Pathol
Lumachi F, Borsato S, Brandes AA, et al. Fine-needle aspiration cytology 1995;26:1154–1156.
of adrenal masses in noncancer patients: clinicoradiologic and histo- Shin SJ, Hoda RS, Ying L, DeLellis RA. Diagnostic utility of the monoclonal
logic correlations in functioning and nonfunctioning tumors. Cancer antibody A103 in fine-needle aspiration biopsies of the adrenal. Am J
2001;93:323–329. Clin Pathol 2000;113;295–302.
Porcaro AB, Novella G, Ficarra V, D’Amico A, Antoniolli SZ, Curti P. Inci- Yang B, Ali SZ, Rosenthal DL. CD10 facilitates the diagnosis of metastatic
dental adrenal pheochromocytoma. Report on 5 operated patients and renal cell carcinoma from primary adrenal cortical neoplasm in adrenal
update of literature. Arch Ital Urol Androl 2003;75:217–225. fine-needle aspiration. Diagn Cytopathol 2002;27:149–152.

Ch010-F06731.indd 346 10/26/2006 10:34:11 AM


Index

Abscess, breast, 127–8, 127f, 127t, 128f clinical features, 113–14t


Acinar cell carcinoma, 274–8 differential diagnosis and pitfalls, 115
ancillary studies, 277 pathologic features, 114–15, 114f, 114t
clinical features, 274–5, 274t Apocrine carcinoma, 148–9, 148t, 149f
cytopathology, 275–7, 275f, 275t, 276f, 277f Aspergillosis, 173–5
differential diagnosis and pitfalls, 277–8 clinical features, 173–4, 173t
radiologic features, 275 cytopathologic features, 174–5, 174f, 175t
Acinic cell carcinoma, 25–7 differential diagnosis and pitfalls, 175
clinical features, 25–6, 26t radiologic features, 174
cytopathologic features, 26–7, 26f, 26t, 27f Autoimmune pancreatitis, 255f, 258f
differential diagnosis and pitfalls, 27 Autoimmune sialadenitis, 4
Actinomycosis, 176–8
clinical features, 176–7, 176t Basal call adenocarcinoma, 18
cytopathologic features, 177f, 177t Basal cell adenoma, 17–18
differential diagnosis and pitfalls, 177–8 clinical features, 17t
radiographic features, 177 cytopathologic features, 17–18, 17f, 18f, 18t, 19f
Adenocarcinoma Benign cystic teratoma, 207, 208f
basal cell, 4 Benign mixed tumor of salivary glands, 7–10
ductal, 160–70 Bile duct adenoma, 229–30, 229t, 230t
foamy gland, 262, 265f Blastomycosis, 167–8
lung, 185–6 clinical features, 167–8, 167t
ancillary studies, 186 cytopathologic features, 168t, 169f
clinical features, 185t differential diagnosis and pitfalls, 168
cytopathologic features, 185–6, 185t, 186f radiologic features, 168
differential diagnosis, 186 Breast, 127–58
radiological features, 185f apocrine carcinoma, 148–9, 148t, 149f
polymorphous low-grade, 21–3, 21t, 22f ductal carcinoma in situ, 140–2, 140t, 141f
signet-ring cell, 267f fi broadenoma, 132–4, 132t, 133f
Adenocortical carcinoma, 331–4 fi brocytic change, 136–40, 136t, 137f, 137t, 138f, 138t
ancillary studies, 331 non-proliferative breast disease, 138, 138f, 139f
clinical features, 331t proliferative breast disease with atypia, 139, 140f
cytopathologic features, 331t, 332f proliferative breast disease without atypia, 138–9, 139f
differential diagnosis and pitfalls, 332–4, 333f, 333t, 334t granular cell tumor, 154f, 154t
Adenoid cystic carcinoma, 19–21 gynecomastia, 154–5, 155f, 155t
clinical features, 19t infi ltrating lobular carcinoma, 149–51, 149t, 150f, 151f
cytopathologic features, 19–21, 19t, 20f, 21f mastitis, abscess and fat necrosis, 127–8, 127f, 127t, 128f
Adenoma medullary carcinoma, 147–8, 147t, 148f, 148t
basal cell, 17–18 metastases, 151–4, 151t, 152f, 152t, 153f
bile duct, 229–30, 229t, 230t mucinous carcinoma, 145–6, 145f, 145t
cortical, 329–31 papillary lesions, 134–6, 134t, 135f, 136f
hepatocellular, 228–9, 228t, 229f primary breast carcinoma, 142–3, 142t, 143f
Adipose tissue tumors, 93–7 secretory, pregnancy and lactational changes, 130–2, 130t, 131f,
lipoma, 93–4 131t, 132f
liposarcoma, 94–7 signet-ring cell carcinoma, 146–7, 146t, 147f
Adrenal gland, 327–44 treatment-induced changes, 128–30, 128t, 129f, 129t, 130f
adenocortical carcinoma, 331–4 tubular carcinoma, 143–5, 144f, 144t
cysts, 327–8 Bronchial cells, 159, 160f
ganglioneuroma, 343–4 Bronchioalveolar carcinoma, 186–9, 186t
metastatic neoplasms, 334–6 cytopathologic features, 187–8, 187f, 187t, 188f
myelolipoma, 328–9 Burkitt lymphoma, 91f
neuroblastoma, 338, 341–2
nodular cortical hyperplasia and cortical adenoma, 329–31 Carcinoid tumor
pheochromocytoma, 336–8 lung, 191–3
Alveolar pneumocytes, 159 ancillary studies, 193
Alveolar soft part sarcoma, 120–1, 120f, 120t, 121t clinical features, 191–2, 192t
Anaplastic carcinoma, 64–7 cytopathologic features, 192–3, 192f, 192t, 193f
ancillary studies, 66 differential diagnosis and pitfalls, 193
clinical features, 64–5, 65t radiologic features, 193
cytopathologic features, 65–6, 65t, 66f, 67f thymic neuroendocrine, 209–10, 211t
differential diagnosis and pitfalls, 67f Carcinosarcoma of salivary glands, 12f
Angiomyolipoma, 320–1, 320t, 321f Cat-scratch disease, 76f
Angiosarcoma Cavernous hemangioma see Hemangioma
liver, 243–4 Cholesterol crystals, 45f
ancillary studies, 244 Chromophobe renal cell carcinoma, 308–11
clinical features, 243–4, 243t ancillary studies, 310
differential diagnosis and pitfalls, 244 clinical features, 308t
pathologic features, 244f, 244t cytopathologic features, 209f, 308–10, 310t, 311f
soft tissue, 113–15, 243–4 differential diagnosis and pitfalls, 310
ancillary studies, 115 Chronic lymphocytic thyroiditis see Hashimoto’s thyroiditis
347

INDEX-F06731.indd 347 10/26/2006 10:35:03 AM


348 INDEX

Chronic pancreatitis, 253–7 Fibrolamellar hepatocellular carcinoma, 237–9


ancillary studies, 254, 256 ancillary studies, 238
clinical features, 253–4, 254t clinical features, 237, 238t
cytopathologic features, 254t, 255f, 256f differential diagnosis and pitfalls, 238
differential diagnosis and pitfalls, 256–7, 257f, 258f pathologic features, 238, 238f, 238t, 239f
radiologic features, 254 Fibromatosis, 98–100
Ciliated hepatic foregut cyst, 225–6, 225t, 226f, 226t ancillary studies, 99
Clear cell renal carcinoma, 302–5 clinical features, 98–9, 98t
ancillary studies, 304 differential diagnosis pitfalls, 100
clinical features, 302–3, 302t pathologic features, 99f, 99t
cytopathologic features, 303–4, 303f, 304f, 405f Fibrosarcoma, 101–2
differential diagnosis and pitfalls, 304 ancillary studies, 101
Clear cell (sugar) tumor, 180–1, 180t, 181f clinical studies, 101t
Coccidioidomycosis, 168–71 differential diagnosis, 101–2
ancillary studies, 170 pathologic features, 101f, 101t
clinical features, 160t, 168–9 Foamy gland adenocarcinoma, 262, 265f
cytopathologic features, 170f, 170t, 171f Focal nodular hyperplasia, 222–4
differential diagnosis and pitfalls, 170–1 ancillary features, 223
radiologic features, 169, 170f clinical features, 222t
Collecting duct renal cell carcinoma, 311–13 differential diagnosis and pitfalls, 224
ancillary studies, 313 pathologic features, 223f, 223t
clinical features, 311t Follicular lymphoma, 83, 84f
cytopathologic features, 311–13, 311t, 312f, 313f Fungal infections
differential diagnosis and pitfalls, 313 lung, 165–76
Cryptococcosis, 171–3 aspergillosis, 173–5
ancillary studies, 173 blastomycosis, 167–8
clinical features, 171–2, 171t coccidioidomycosis, 168–71
cytopathologic features, 172–3, 172f, 173f, 173t cryptococcosis, 171–3
differential diagnosis and pitfalls, 173 histoplasmosis, 166–7
radiographic features, 172 zygomycoses, 175–6
Cystic lesions of kidney, 321–4, 322f, 322t, 323f, 323t
Cystic papillary carcinoma, 46 Ganglioneuroblastoma see Neuroblastoma
Cysts Ganglioneuroma, 344f
adrenal gland, 327–8, 327t, 328t Gastrointestinal stromal tumor, 115–17
ciliated hepatic foregut, 225–6 ancillary studies, 116–17
lymphoepithelial, 260 clinical features, 115t
retention, 4–5, 6f differential diagnosis and pitfalls, 117
salivary glands, 4–5, 6f pathologic features, 115–16, 115t, 116f
Germ cell tumors
Dedifferentiated liposarcoma, 96f mediastinum, 206–9
Dermatofi brosarcoma protuberans, 100–1 ancillary studies, 208–9
ancillary studies, 101 clinical features, 206–7, 206t
clinical features, 100t cytopathologic features, 207–8, 207t, 208f,
differential diagnosis, 101 209f
pathologic features, 100f, 100t differential diagnosis and pitfalls, 209
Ductal adenocarcinoma, 260–70 Glial fi brillary acidic protein, 13
ancillary studies, 269 Goitre see Nodular goitre
clinical features, 260–1, 261t Granular cell tumor, 106–7, 154f, 154t
cytopathologic features, 261–8, 262t, 263f, 264f, 265f, 266f, breast, 154f, 154t
267f, 268f soft tissue
differential diagnosis and pitfalls, 269–70, 269f ancillary studies, 107
radiologic features, 261 clinical features, 106t
Ductal carcinoma in situ, 140–2, 140t, 141f differential diagnosis and pitfalls, 107
pathologic features, 106–7, 106t, 107f
Embryonal carcinoma, 208, 209f Granulomatous lymphadenopathy, 74–7
Entamoeba histolytica, 224t ancillary studies, 76f
Epithelioid hemangioendothelioma, 242–3 clinical features, 74–5, 75t
ancillary studies, 243 cytopathologic features, 75–6, 75t, 76f
clinical features, 242t differential diagnosis and pitfalls, 76–7
differential diagnosis, 243 Granulomatous sialadenitis, 5f
pathologic features, 243f, 243t Gynecomastia, 154–5, 155f, 155t
Epithelioid sarcoma, 118–20
ancillary studies, 119 Hamartoma, 178–80
clinical features, 118–19, 118t clinical features, 178t
differential diagnosis and pitfalls, 120 cytopathologic features, 178–80, 179f, 180t
pathologic features, 119f, 119t differential diagnosis and pitfalls, 180
Epstein-Barr virus, 77 radiologic features, 178f
Hashimoto’s thyroiditis, 37–41
Fat necrosis of breast, 127–8, 127f, 127t, 128f ancillary studies, 39
Fibroadenoma, 132–4, 132t, 133f clinical features, 37–8, 38t
Fibroblastic/fi brohistiocytic tumors, 97–103 cytopathologic features, 39f, 39t, 40f, 41f
dermatofi brosarcoma protuberans, 100–1 differential diagnosis and pitfalls, 39–40
fi bromatosis, 98–100 Hassall’s corpuscles, 205
fi brosarcoma, 101–2 Hemangioma, 226–8
malignant fi brous histiocytoma, 102–3 ancillary studies, 227
nodular fasciitis, 97–8 clinical features, 226–7, 226t

INDEX-F06731.indd 348 10/26/2006 10:35:03 AM


INDEX 349

differential diagnosis and pitfalls, 228 Lactation, breast changes, 130–2, 130t, 131f, 131t, 132f
pathologic features, 227, 227f, 227t, 228f Large cell carcinoma, 190–1, 190t, 191f
Hepatobiliary cystadenoma, 230–1, 230t, 231f Leiomyosarcoma, 112–13
Hepatoblastoma, 241–2, 241t, 242f, 242t ancillary studies, 112–13
Hepatocellular adenoma, 228–9, 228t, 229f clinical features, 112t
Hepatocellular carcinoma, 232–7 differential diagnosis and pitfalls, 113
clinical features, 232–3, 233t pathologic features, 112, 112f, 113f
see also Fibrolamellar hepatocellular carcinoma Lipoma, 93–4
Histiocytoid cells, 57 ancillary studies, 94
ancillary studies, 235, 236f, 237f, 237t clinical features, 93t
differential diagnosis, 237 differential diagnosis and pitfalls, 94
pathologic features, 233–5, 234f, 234t, 235f, 236f pathologic features, 94f, 94t
Histoplasmosis, 166–7 Liposarcoma, 94–7
ancillary studies, 167 ancillary features, 96
clinical features, 166t clinical features, 94–5, 95t
cytopathologic features, 166–7, 167f, 167t dedifferentiated, 96f
differential diagnosis and pitfalls, 167 differential diagnosis and pitfalls, 96
radiologic features, 166 myxoid, 96f
Hodgkin lymphoma, 78–81, 212f pathologic factors, 95–6, 95t, 96f, 97f
ancillary features, 79–80, 80t pleomorphic, 97f
clinical features, 78–9t Liver, 219–50, 220f
cytopathologic features, 79t, 80f, 81f benign tumors, 226–32
differential diagnosis and pitfalls, 80–1 bile duct adenoma, 229–30
Hürthle cell neoplasms, 49–51 hemangioma, 226–8
ancillary studies, 51 hepatobiliary cystadenoma, 230–1
clinical features, 49–50, 49t hepatocellular adenoma, 228–9
cytopathologic features, 50–1, 50f, 50t, 51f inflammatory myofi broblastic tumor, 231–2
differential diagnosis and pitfalls, 51 contraindications to fine needle aspiration, 221t
Hyaline cell myoepithelioma, 13 cystic lesions, 224–6
Hyalinizing trabecular adenoma, 59f ciliated hepatic foregut cyst, 225–6
Hydatid disease, 224–5 hydatid disease, 224–5
clinical features, 224t metastatic tumors, 246–9, 246t, 247f, 247t, 248f, 248t,
differential diagnosis and pitfalls, 225 249f
pathologic features, 224–5, 225f, 225t needle contamination, 220f, 220t
non-neoplastic diseases, 219–24
Infectious mononucleosis, 77–8 focal nodular hyperplasia, 222–4
ancillary studies, 77–8 macroregenerative nodule, 219–22
clinical features, 77t normal cytology, 220t
cytopathologic features, 77t, 78f parasitic cysts, 224t
differential diagnosis and pitfalls, 78 primary malignant epithelial tumors, 232–42
Inflammatory myofi broblastic tumor, 231–2, 231t, 232f, 232t fi brolamellar hepatocellular carcinoma, 237–9
Insular carcinoma, 63–4 hepatoblastoma, 241–2
ancillary studies, 64 hepatocellular carcinoma, 232–7
clinical features, 63t intrahepatic cholangiocarcinoma, 239–41
cytopathologic features, 63–4, 64f, 64t, 65f primary malignant mesenchymal tumors, 242–6
differential diagnosis and pitfalls, 64 angiosarcoma, 243–4
Intraductal papillary mucinous neoplasm, 291–6 epithelioid hemangioendothelioma, 242–3
ancillary studies, 292 primary hepatic lymphoma, 244–6
clinical features, 291t Lung, 159–99
cytopathologic features, 291–2, 291t, 292f, 293f, 294f, 295f benign neoplasms, 178–81
differential diagnosis and pitfalls, 292 clear cell (sugar) tumor, 180–1
Intrahepatic cholangiocarcinoma, 239–41 hamartoma, 178–80
ancillary studies, 240–1 carcinoid tumor, 191–3
clinical features, 239t metastases, 193–8, 194f, 194t, 195f, 196f, 197f, 198f
differential diagnosis and pitfalls, 241 non-neoplastic conditions, 159–78
pathologic features, 239–40, 239t, 240f actinomycosis, 176–8
fungal infections, 165–76
Kidney, 299–327 sarcoidosis, 159–63
angiomyolipoma, 320–1 tuberculosis, 163–5
cystic lesions, 321–4, 322f, 322t, 323f, 323t normal constituents, 159
benign, 321–3 primary malignant tumors, 181–91
malignant, 323–4 adenocarcinoma, 185–6
lymphoma, 325–6 bronchioloalveolar carcinoma, 186–9
metastatic neoplasms, 326–7 large cell carcinoma, 190–1
nephroblastoma (Wilms tumor), 317–18 pleomorphic carcinoma, 190–1
oncocytoma, 316–17 small cell carcinoma, 189–90
renal cell carcinoma, 299–315 squamous cell carcinoma, 181–4
chromophobe type, 308–11 Lymph nodes, 71–92
clear cell type, 302–5 granulomatous lymphadenopathy, 74–7
collecting duct type, 311–13 ancillary studies, 76f
medullary type, 313–14 clinical features, 74–5, 75t
papillary type, 305–6 cytopathologic features, 75–6, 75t, 76f
sarcomatoid type, 314–15 differential diagnosis and pitfalls, 76–7
rhabdoid tumor, 318–20 Hodgkin lymphoma, 78–81
urothelial carcinoma of renal pelvis, 324–5 ancillary features, 79–80, 80t
Kuttner tumor, 2 clinical features, 78–9t

INDEX-F06731.indd 349 10/26/2006 10:35:03 AM


350 INDEX

cytopathologic features, 79t, 80f, 81f Medullary renal cell carcinoma, 313–14
differential diagnosis and pitfalls, 80–1 ancillary studies, 314
infectious mononucleosis, 77–8 clinical features, 313t
ancillary studies, 77–8 cytopathologic features, 314f, 314t
clinical features, 77t differential diagnosis and pitfalls, 314
cytopathologic features, 77t, 78f Mesothelial cells, 159
differential diagnosis and pitfalls, 78 Metastases
non-Hodgkin lymphoma adrenal gland, 334–6, 334t, 335f, 336f, 337f
large-cell, 86–9 breast, 151–4, 151t, 152f, 152t, 153f
pediatric, 89–91 kidney, 326–7
small-cell, 81–6 liver, 246–9, 246t, 247f, 247t, 248f, 248t, 249f
reactive lymphoid hyperplasia, 71–4 lung, 193–8, 194f, 194t, 195f, 196f, 197f, 198f
ancillary studies, 73–4 mediastinum, 215–17
clinical features, 71t Mucinous carcinoma, 145–6, 145f, 145t
cytopathologic features, 71–3, 71t, 72f, 73f Mucinous cystic neoplasm, 285–90
differential diagnosis and pitfalls, 74 ancillary studies, 289
Lymphoepithelial cysts, 260t, 261f clinical features, 285t
Lymphoepithelial sialadenitis, 4 cytopathologic features, 285–9, 286f, 286t, 287f, 288f,
Lymphoma 289f
follicular, 83, 84f differential diagnosis, 289, 290f
hepatic, 244–6 Mucocele, 4–5, 6f
ancillary studies, 246 Mucoepidermoid carcinoma, 9, 23–5
clinical features, 244–5, 245t clinical features, 23–4, 23t
differential diagnosis and pitfalls, 246 cytopathologic features, 24f, 24t, 25f
pathologic features, 245–6, 245f, 245t, 246f differential diagnosis and pitfalls, 24–5
Hodgkin see Hodgkin lymphoma Mycobacterial lymphadenitis, 76f
kidney, 325–6, 325t, 326f Myelolipoma, 328–9, 328t, 329f
mantle, 83, 84f Myoepithelioma, 12–13, 13f, 14f
mediastinal, 210–13 Myxoid liposarcoma, 96f
ancillary studies, 212 Myxoma, 117–18, 117t, 118f
clinical features, 210, 211t
cytopathologic features, 210–12, 212f, 212t, 213f Nasopharyngeal carcinoma, 89f
non-Hodgkin see Non-Hodgkin lymphoma Necrotizing sialometaplasia, 4
salivary glands, 30–1 Nephroblastoma (Wilms tumor), 317–18
clinical features, 30t ancillary studies, 318
cytopathologic features, 30–1, 30f, 31f, 31t clinical features, 317t
differential diagnosis and pitfalls, 31 cytopathologic features, 317–18, 318t
small lymphocytic, 83–4, 84f differential diagnosis and pitfalls, 318
thyroid gland, 68–9, 68t, 69f Neural tumors, 104–9
granular cell tumor, 106–7
Macroregenerative nodule, 219–22 malignant peripheral nerve sheath tumors, 108–9
ancillary studies, 222 neurofi broma and schwannoma, 104–6
clinical features, 219–21, 219t Neuroblastoma, 338, 341–2
differential diagnosis and pitfalls, 222 ancillary studies, 341
pathologic features, 221–2, 221f, 221t, 222f clinical features, 338, 341t
Malignant fi brous histiocytoma, 102–3 cytopathologic features, 341, 342f, 342t
ancillary studies, 103 differential diagnosis and pitfalls, 341–2
clinical features, 102t Neurofi broma, 104–6
differential diagnosis and pitfalls, 103 ancillary studies, 106
pathologic features, 102–3, 103f, 103t clinical features, 104t
Malignant melanoma, 90f differential diagnosis and pitfalls, 106
Malignant mixed tumor of salivary glands, 10–12, 11f pathologic features, 104–6, 104t, 105f
Malignant peripheral nerve sheath tumors, 108–9, 108t, 109f Neurogenic tumors, 213–15
Mantle lymphoma, 83, 84f clinical features, 213t
Marginal zone lymphoma, 84–5, 85f cytologic features, 213–15, 214f, 215t
Masood Cytology Index, 137t, 138f Nodular cortical hyperplasia and cortical adenoma, 329–31
Mastitis, 127–8, 127f, 127t, 128f ancillary studies, 330
Mediastinal lymphomas, 210–13 clinical features, 329t
Mediastinum, 201–18, 202t cytopathologic features, 329–30, 330f, 330t
differential diagnosis, 202t differential diagnosis and pitfalls, 330–1
germ cell tumors, 206–9 Nodular fasciitis, 97–8
location of mediastinal lesions, 202t ancillary studies, 98
mediastinal lymphomas, 210–13 clinical features, 97t
metastatic lesions, 215–17 differential diagnosis and pitfalls, 98
neurogenic tumors, 213–15 pathologic features, 97–8, 97t, 98f
thymic carcinomas, 205–6 Nodular goitre, 41–6
thymic follicular hyperplasia, 206 ancillary studies, 46
thymic neuroendocrine (carcinoid) tumors, 209–10 clinical features, 41t
thymoma, 203–5 cytopathologic features, 41–2, 42f, 42t, 43f, 44f, 45f, 46f
Medullary carcinoma, 60–3, 147–8, 147t, 148f, 148t differential diagnosis and pitfalls, 46
breast, 147–8, 147t, 148f, 148t Non-Hodgkin lymphoma, 212f, 213f
thyroid gland large-cell, 86–9
ancillary studies, 61–2 ancillary studies, 88
clinical features, 60t clinical features, 86–7, 86t, 87t
cytopathologic features, 60–1, 60t, 61f, 62f, 63f cytopathologic features, 87–8, 87t, 88f
differential diagnosis and pitfalls, 62–3 differential diagnosis, 86–7, 87f, 88f, 89f, 90f

INDEX-F06731.indd 350 10/26/2006 10:35:04 AM


INDEX 351

pediatric, 89–91 clinical features, 71t


ancillar studies, 91 cytopathologic features, 71–3, 71t, 72f, 73f
clinical features, 89–90, 90t differential diagnosis and pitfalls, 74
cytopathologic features, 90–1, 91f Reed-Sternberg cells, 80f, 81f, 82f
differential diagnosis and pitfalls, 91 Renal cell carcinoma, 299–315, 300f
small-cell, 81–6 chromophobe type, 308–11
ancillary studies, 85 classification, 299, 300t
clinical features, 81–3, 82t clear cell type, 302–5
cytopathologic features, 83–5, 83t, 84f, 85f, 85t clinical features, 299t
differential diagnosis and pitfalls, 85–6, 86f collecting duct type, 311–13
cytopathologic features, 301–2, 302t
Oncocytic carcinoma, 16 medullary type, 313–14
Oncocytoma, 16, 316–17 nuclear grading, 300–1, 301f, 302f
ancillary studies, 316–17 papillary type, 305–6
clinical features, 316t sarcomatoid type, 314–15
cytopathologic features, 316f, 316t, 317f Retention cyst, 4–5, 6f
differential diagnosis and pitfalls, 317 Rhabdoid tumor, 318–20
Owl’s eye appearance, 269f ancillary studies, 319, 320f
Pancreas, 251–98 clinical features, 318–19, 319t
cystic neoplasms, 279–96 cytopathologic features, 319f, 319t
intraductal papillary mucinous neoplasm, 291–6 differential diagnosis and pitfalls, 319
mucinous cystic neoplasm, 285–90 Rhabdomyosarcoma, 109–11
serous cystadenoma, 279–82 ancillary studies, 111
solid-pseudopapillary neoplasm, 282–5 clinical features, 109–10, 109t
non-epithelial and metastatic tumors, 296 differential diagnosis and pitfalls, 111
non-neoplastic mass lesions, 253–60 pathologic features, 110–11, 110f, 110t, 111f
chronic pancreatitis, 253–7
lymphoepithelial cysts, 260 Salivary duct carcinoma, 28–9
pancreatic pseudocyst, 257–60 clinical features, 28, 28t
normal, 251–3, 252f, 253f cytopathologic features, 28–9, 28t, 29f
solid neoplasms, 260–79 differential diagnosis and pitfalls, 29
acinar cell carcinoma, 274–8 Salivary glands, 1–36, 2f
ductal adenocarcinoma, 260–70 acinic cell carcinoma, 25–7
pancreatic endocrine neoplasm, 270–4 clinical features, 25–6, 26t
pancreatoblastoma, 278–9 cytopathologic features, 26–7, 26f, 26t, 27f
differential diagnosis and pitfalls, 27
Pancreatic endocrine neoplasm, 270–4 adenocarcinoma, 32
ancillary studies, 273 basaloid neoplasms, 17–23
clinical features, 270t adenoid cystic carcinoma, 19–21
cytopathologic features, 270–3, 270f, 271f, 272f, basal cell adenocarcinoma, 18
273f basal cell adenoma, 17–18
differential diagnosis and pitfalls, 273–4, 274f polymorphous low-grade adenocarcinoma, 21–3
Pancreatic pseudocyst, 257–60 benign mixed tumor, 7–10
ancillary features, 258–9 clinical features, 7–8, 7t
clinical features, 257–8, 258t cytopathological features, 7–8, 8f, 9f
cytopathologic features, 258, 259f, 259t differential diagnosis and pitfalls, 8–10, 10f, 11f
differential diagnosis and pitfalls, 259–60 carcinosarcoma, 12f
radiologic features, 258 clear cell carcinoma, 32
Pancreatoblastoma, 278–9, 278t, 279f, 280f lymphoepithelial carcinoma, 32, 33t
Papillary carcinoma, 51–9 malignant lymphoma, 30–1
ancillary studies, 58 malignant mixed tumor, 10–12, 11f
clinical features, 51–2, 51t mucoepidermoid carcinoma, 23–5
cytopathologic features, 52–8, 53f, 54f, 55f, 56f, 57f, 58f clinical features, 23–4, 23t
differential diagnosis and pitfalls, 58–9, 59f cytopathologic features, 24f, 24t, 25f
Papillary renal cell carcinoma, 305–6 differential diagnosis and pitfalls, 24–5
ancillary studies, 305, 308, 309f myoepithelial neoplasms, 12–13, 13f, 14f
clinical features, 305t non-neoplastic cystic lesions, 4–7
cytopatologic features, 305, 306f, 307f, 308t cytopathologic features, 5–6, 6f
differential diagnosis and pitfalls, 308 differential diagnosis and pitfalls, 6–7
Parathyroid cyst, 46 non-neoplastic lesions, 1–7
Pheochromocytoma, 336–8 sialadenitis, 1–4, 2t, 3f, 4f
ancillary studies, 338, 341f sialadenosis, 1
clinical features, 336–8, 338t oncocytic neoplasms, 13–16
cytopathologic features, 338t, 339f, 340f oncocytoma and oncocytic carcinoma, 16
differential diagnosis and pitfalls, 338 Warthin tumor, 13–16
Plasmacytoma, 274f primary small cell carcinoma, 31–2, 32f
Pleomorphic carcinoma, 190–1, 190t, 191f salivary duct carcinoma, 28–9
Pleomorphic liposarcoma, 97f sarcoma, 32
Polymorphous low-grade adenocarcinoma, 21–3, 21t, secondary tumors, 32–4, 34f
22f squamous cell carcinoma, 32, 33t
differential diagnosis and pitfalls, 22–3 ‘Salt and pepper’ chromatin, 211f, 270, 271f
pathologic features, 23t Sarcoidosis, 76f, 159–63
Pregnancy, breast changes, 130–2, 130t, 131f, 131t, 132f ancillary studies, 162
clinical features, 159–61, 160f, 160t, 161f, 161t
Reactive lymphoid hyperplasia, 71–4 cytopathologic features, 161–2, 162f, 162t
ancillary studies, 73–4 differential diagnosis and pitfalls, 162–3

INDEX-F06731.indd 351 10/26/2006 10:35:04 AM


352 INDEX

lung, 159–63 Thymic carcinoma, 205–6, 205f, 206f


radiologic features, 161 Thymic follicular hyperplasia, 206
Sarcoma Thymic neuroendocrine (carcinoid) tumors, 209–10
alveolar soft part, 120–1, 120f, 120t, 121t ancillary features, 210
epithelioid, 118–20 clinical features, 209–10, 209t
salivary gland, 32 cytopathologic features, 210f, 210t, 211f
synovial, 121–3 differential diagnosis and pitfalls, 210, 211f
Sarcomatoid renal cell carcinoma, 314–15, 315f, 315t Thymoma, 203–5
Schistosoma mansoni, 224t ancillary studies, 205
Schwannoma, 104–6, 213f clinical features, 203t
ancillary studies, 106 cytopathologic features, 203–5, 203t, 204f
clinical features, 104t differential diagnosis, 205, 205f
differential diagnosis and pitfalls, 106 Thyroid gland, 37–70, 38f
pathologic features, 104–6, 104t, 105f anaplastic carcinoma, 64–7
Seminoma, 89f, 207 ancillary studies, 66
Serous cystadenoma, 279–82 clinical features, 64–5, 65t
ancillary studies, 281 cytopathologic features, 65–6, 65t, 66f, 67f
clinical features, 279–80, 279t differential diagnosis and pitfalls, 67f
cytopathologic features, 280–1, 280t, 281f follicular neoplasms, 47–9
differential diagnosis and pitfalls, 281–2 ancillary studies, 48–9
radiologic features, 280 clinical features, 47t
Sialadenitis, 1–4 cytopathologic features, 47–8, 47t, 48f, 49f
clinical features, 1–2t differential diagnosis and pitfalls, 49
cytopathologic features, 2–4, 2t, 3f, 4f, 5f Hashimoto’s thyroiditis, 37–41
differential diagnosis and pitfalls, 4 ancillary studies, 39
Sialadenosis, 1 clinical features, 37–8, 38t
Sialolithiasis, 2 cytopathologic features, 39f, 39t, 40f, 41f
Signet-ring cell adenocarcinoma, 267f differential diagnosis and pitfalls, 39–40
Signet-ring cell carcinoma, 146–7, 146t, 147f Hürthle cell neoplasms, 49–51
Sjögren syndrome, 4 ancillary studies, 51
Skeletal muscle tumors, 109–11 clinical features, 49–50, 49t
Small cell carcinoma, 189–90, 189f, 189t, 190f cytopathologic features, 50–1, 50f, 50t, 51f
Small lymphocytic lymphoma, 83–4, 84f differential diagnosis and pitfalls, 51
Smooth muscle tumors, 112–13 insular carcinoma, 63–4
Soft tissue, 93–126 ancillary studies, 64
adipose tissue tumors, 93–7 clinical features, 63t
lipoma, 93–4 cytopathologic features, t, 63–4, 64f, 64t, 65f
liposarcoma, 94–7 differential diagnosis and pitfalls, 64
alveolar soft part sarcoma, 120–1 lymphoma, 68–9, 68t, 69f
epithelioid sarcoma, 118–20 medullary carcinoma, 60–3
fi broblastic/fi brohistiocytic tumors, 97–103 ancillary studies, 61–2
dermatofi brosarcoma protuberans, 100–1 clinical features, 60t
fi bromatosis, 98–100 cytopathologic features, 60–1, 60t, 61f, 62f, 63f
fi brosarcoma, 101–2 differential diagnosis and pitfalls, 62–3
malignant fi brous histiocytoma, 102–3 nodular goitre, 41–6
nodular fasciitis, 97–8 ancillary studies, 46
gastrointestinal stromal tumor, 115–17 clinical features, 41t
myxoma, 117–18 cytopathologic features, 41–2, 42f, 42t, 43f, 44f, 45f, 46f
neural tumors, 104–9 differential diagnosis and pitfalls, 46
granular cell tumor, 106–7 papillary carcinoma, 51–9
malignant peripheral nerve sheath tumors, 108–9 ancillary studies, 58
neurofi broma and schwannoma, 104–6 clinical features, 51–2, 51t
skeletal muscle tumors, 109–11 cytopathologic features, 52–8, 53f, 54f, 55f, 56f, 57f, 58f
smooth muscle tumors, 112–13 differential diagnosis and pitfalls, 58–9, 59f
synovial sarcoma, 121–3 Tuberculosis, 163–5
vascular tumors, 113–15 ancillary studies, 165
Solid-pseudopapillary neoplasm, 282–5 clinical features, 163t
ancillary studies, 285 cytopathologic features, 163–5, 163t, 164f, 165f
clinical features, 282t differential diagnosis and pitfalls, 165
cytopathologic features, 282–4, 282t, 283f, 284f radiologic features, 163
differential diagnosis and pitfalls, 285 Tubular carcinoma, 143–5, 144f, 144t
radiologic features, 282
Squamous cell carcinoma Urothelial carcinoma of renal pelvis, 324–5, 324f, 324t
lung, 181–4
clinical features, 181t Vascular tumors, 113–15
cytopathologic features, 182–3, 182f, 182t, 183f, 183t, Warthin tumor, 4, 13–16
184f clinical features, 13, 14t
differential diagnosis and pitfalls, 183–4 cytopathological features, 14–15, 15f, 16f
radiologic features, 182 differential diagnosis and pitfalls, 15–16
salivary glands, 32, 33t
Squamous pearls, 205 Yolk sac tumor, 207, 208f, 209f
Synovial sarcoma, 121–3
ancillary studies, 122 Zygomycoses, 175–6
clinical features, 121t clinical features, 175t
differential diagnosis and pitfalls, 123 cytopathologic features, 176f, 176t
pathologic features, 121–2, 122f, 122t, 123f radiologic features, 175

INDEX-F06731.indd 352 10/26/2006 10:35:04 AM

You might also like