The Washington Manual of Surgical Pathology
The Washington Manual of Surgical Pathology
The Washington Manual of Surgical Pathology
Editors
Peter A. Humphrey, MD, PhD
Lou is P. Dehner, MD
John D. Pfeifer, MD, PhD
I
®.Wolters Kluwer Lippincott Williams & Wilkins
Health
Philadelphia • Baltimore • New York • London
Buenos Aires • Hong Kong • Sydney • Tokyo
Senior Executive Editor: Jonathan W. Pine, Jr.
Product Managers: Marian Bdlus, Emilie Moyer
Vendor Manager: Bridgett Dougherrty
Senior Manufacturing Manager: Benjamin Rivera
Director of Marketing: Caroline Foote
Design Coordinator: Holly McLaughlin
Production Service: Aptara, Inc.
All rights reserved. This book is protected by copyright. No part of this book may be reproduced in
any form by any means, including photocopying, or utilized by any information storage and retrieval
system without written permission from the copyright owner, except for brief quotations embodied in
critical articles and reviews. Materials appearing in this book prepared by individuals as part of their
official duties as U.S. government employees are not covered by the above-mentioned copyright.
Printed in China
Care has been taken to confirm the accuracy of the information presented and to describe generally
accepted practices. However, the authors, editors, and publisher are not responsible for errors or
omissions or for any consequences from application of the information in this book and make no
warranty, expressed or implied, with respect to the currency, completeness, or accuracy of the contents
of the publication. Application of the information in a particular situation remains the professional
responsibility of the practitioner.
The authors, editors, and publisher have exerted every effort to ensure that drug sdection and
dosage set forth in this text are in accordance with current recommendations and practice at the time
of publication. However, in view of ongoing research, changes in government regulations, and the
constant flow of information relating to drug therapy and drug reactions, the reader is urged to check
the package insert for each drug for any change in indications and dosage and for added warnings
and precautions. This is particularly important when the recommended agent is a new or infrequently
employed drug.
Some drugs and medical devices presented in the publication have Food and Drug Adminis-
tration (FDA) clearance for limited use in restricted research settings. It is the responsibility of the
health care provider to ascertain the FDA status of each drug or device planned for use in their clinical
practice.
To purchase additional copies of this book, call our customer service department at (800) 638-3030
or fax orders to (301) 223-2320. International customers should call (301) 223-2300.
Visit Lippincott WJ.lliams & WJ.lkins on the Internet at LWW.com. Lippincott Williams & Wilkins
customer service representatives are available from 8:30am to 6:00pm, EST.
10 9 8 7 6 5 4 3 2 1
My wife Kay, and children Tom and Jennifer-P.A.H.
For the continued opportunity, joy, and satisfaction of working with great
colleagues and house staff-L.P.D.
P.J., S.M., and L.P.D., for their friendship and support-J.D.P.
Contributors
Ashima Agarwal, MD Danielle H. Carpenter, MD
Instructor Surgical Pathology Fellow
Department of Pathology and Immunology Department of Pathology and Immunology
Washington University School of Medicine Washington University School of Medicine
St. Louis, Missouri St. Louis, Missouri
X
Acknowledgments
The second edition of The Washington Manual of Surgical Pathology would not have
been possible without the participation of our colleagues, who so willingly and gener-
ously provided their time and expertise to the project. All but one of the authors (again,
special thanks to Dr. Michael Klein) are, or were, faculty or house staff in the Depart-
ment of Pathology and Immunology at Washington University School of Medicine,
which emphasizes that surgical pathology at Washington University has always been a
collaborative venture between the faculty and trainees. The Department's Chairman,
Dr. Herbert (Skip) Virgin, has not hesitated to continue to provide the support nec-
essary to maintain the tradition of diagnostic excellence, academic productivity, and
education that forms the foundation of academic surgical pathology at our institution.
We extend special thanks to our administrative assistants, Elease Barnes, Jeannie
Doerr, and Shari Jackson, who handled most of the logistical and secretarial work
required to make the second edition a reality. We also acknowledge the artistic expertise
of the staff at MedPIC (the medical illustration service of Washington University School
of Medicine), especially Marcy Hartstein and Vicki Friedman, who together revised the
figures for the book. Walter Clermont expertly and patiently edited all the electronic
figures found at the associated website. We are lucky to continue to have had the
opportunity to work with several wonderful people at Lippincott William & Wilkins,
including Jonathan W. Pine, Jr. (who enthusiastically suggested at an early stage that
we produce a second edition of the manual), Emilie Moyer, Marian Bellus, and Martha
Cushman.
And our families. Their love, patience, and support made the book possible, and
make it all worthwhile.
Peter A. Humphrey
Louis P. Dehner
John D. Pfeifer
xi
Contents
Contributors v
Preface x
Acknowledgments xt
- . Larynx 19
Iii James S. Lewis Jr.
Ell Nasal Cavity, Paranasal Sinuses, and
. . Nasopharynx 34
Heather N. Wright and James S. Lewis Jr.
El Salivary Glands 81
. . Joshua I. Warrick, Elise L. Krejci, and James S. Lewis Jr.
Cytopathology of the Salivary Glands 95
Brian Collins
The Ear 97
Ill Peter A. Humphrey and Rebecca D. Chernock
II THORAX 104
Ell Lung 104
. . Jon H. Ritter and Hannah R. Krigman
Cytopathology of the Lung 145
Hannah R. Krigman
IV BREAST 298
11:1 Breast Pathology 298
. . Souzan Sanati, Omar Hameed, Joshua I. Warrick,
and Craig Allred
Em Vagina 551
. . Rao Watson, John D. Pfeifer, and Phyllis C. Huettner
E6 Placenta 566
Iiiii Phyllis C. Huettner
VIII SKIN 579
Ern Skin: Nonneoplastic Dermatopathology 579
. . Samuel J. Pruden II, Kimberley G. Crone, and Anne C. Lind
~ NERVOUSSYSTEM 625
HI Central Nervous System: Brain, Spinal Cord,
1M and Meninges 625
Richard J. Perrin, Sushama Patil, and Arie Perry
Cytopathology of the Central Nervous System 671
Souzan Sanati and Lourdes R. Ylagan
~ Spleen 729
1M Mohammad 0. Hussaini and Anjum Hassan
Index 931
SECTION I
Head and Neck
I. NORMAL ANATOMY
A. Oral cavity. The anterior aspect of the oral cavity extends from the mucocu-
taneous junction (vermilion border) of the lips to include the buccal mucosa
(inside of cheek), maxillary and mandibular arches (teeth), retromolar trigone,
anterior two-thirds of the tongue (oral tongue), floor of mouth, and hard palate.
Posteriorly, the oral cavity freely communicates with the oropharynx; the border
between the two is marked by the junction of the hard and the soft palate superi-
orly and the line of circumvallate papillae on the dorsal tongue (border between
the anterior two-thirds and posterior one-third of the tongue) inferiorly.
1. The oral tongue is freely mobile and composed mainly of skeletal muscle. It
has a dorsal (exposed) surface, ventral surface, and tip. The dorsal surface
contains numerous papillae that have specialized taste receptors. The floor
of the mouth lies beneath the tongue and is divided into sides by the midline
frenulum (mucosal fold) of the tongue. It contains ostia of the submandibu-
lar and sublingual salivary glands, whereas the main duct of the parotid
gland (Stensen's duct) enters the oral cavity through the buccal mucosa. The
hard palate forms the roof of the oral cavity and consists of portions of the
maxillary and palatine bones.
2. The oral cavity is lined by stratified squamous mucosa with prominent
mucoserous glands in the submucosa. Most of the mucosa is nonkeratiniz-
ing with the exception of the hard palate, gingiva, and dorsal tongue, which
become keratinized due to the friction of mastication.
B. Oropharynx. The oropharynx is the space posterior to the oral cavity that com-
municates with the nasopharynx superiorly and the larynx and hypopharynx
inferiorly. The soft palate (posterior one-third of the palate) marks the superior
aspect of the oropharynx and is suspended from the posterior aspect of the hard
palate. In contrast to the hard palate, the soft palate is fibromuscular without a
bony skeleton. Whereas the oral aspect is covered with nonkeratinizing stratified
squamous epithelium, the nasal surface is covered by pseudostratified ciliated
columnar (respiratory-type) epithelium. Numerous mucoserous glands lie in its
submucosa. The uvula extends down from the posterior aspect of the soft palate
and has an identical histology. The posterior one-third of the tongue is rich in
lymphoid tissue (known as the lingual tonsil). The palatine and lingual tonsils,
together with the pharyngeal tonsil in the nasopharynx, are collectively known
as Waldeyer's ring.
2 I SECTION 1: HEAD AND NECK
more frequently in women than men. They are diseases that affect mucosal sites
as well as skin, so the oral cavity is sometimes involved as well.
1. Lichen planus. This disorder commonly affects the oral mucosa. Whereas
skin involvement is usually self-limited, oral lichen planus follows a more
protracted waxing and waning course. The oral lesions are typically asymp-
tomatic unless ulceration occurs.
Any oral mucosal surface may be involved, and several patterns can be
seen. The classic pattern is reticular with intersecting white keratotic streaks
(Wickham's striae); the lesions are ill-defined, and the background may be
erythematous due to mucosal atrophy. Some lesions may be mostly erythema-
tous with minimal keratotic streaks, whereas others may show extensive ker-
atinization and/or ulceration or form bullae. Microscopically, a dense submu-
cosal band of lymphocytes is present, which may be less distinct in ulcerated
lesions (e-Fig. 1.1)."' The rete ridges may be hyperplastic (saw-toothed) or
flattened. There is loosening of the basal layer of the epithelium, with degener-
ation of individual keratinocytes that may form eosinophilic colloid (dysker-
atotic, cytoid, or Civatte) bodies (e-Fig. 1.2). The surface may show hyper-
or parakeratosis. Although the histologic findings of lichen planus have been
well defined, they are nonetheless not specific; for example, some oral lesions
with a similar histologic picture may be due to a contact hypersensitivity reac-
tion. In addition, a lichenoid infiltrate may accompany dysplastic lesions.
Asymptomatic patients require no treatment, but steroids (particularly
topical) are often used for erosive or erythematous lesions. Some data sug-
gest that there is an increased risk of malignant transformation in the erythe-
matous, ulcerative, and bullous forms of oral lichen planus. Although this
proposed risk of malignancy is controversial, these lesions at least require
closer clinical follow-up.
2. Pemphigus vulgaris. This is an uncommon disorder that causes superficial
ulceration of the skin and mucous membranes. Involvement of the oral
mucosa may precede the development of skin lesions. The disease is caused
by autoantibodies to desmogleins 1 and 3, cellular transmembrane proteins
involved in the assembly of desmosomes. Cell-to-cell adhesion is impaired
in the suprabasal epithelium, leading to clefting and ulceration. Flaccid bul-
lae that easily rupture to form painful erosions can be seen on any oral
mucosal surface. The lesions heal without scarring. Microscopically, intraep-
ithelial separation with edema and acantholysis, which imparts a "tomb-
stone" appearance to the remaining attached basal cell layer (e-Fig. 1.3),
is seen at the edge of the ulce1:. Acute and chronic inflammation are fre-
quently present in the submucosa. Direct immunofluorescence is positive for
immunoglobulin G (lgG) along cell membranes throughout the epidermis.
Paraneoplastic pemphigus, which is associated with an underlying malig-
nancy, may be distinguished from pemphigus vulgaris by the identification
of a different pattern of antibody staining by direct immunofluorescence.
3. Cicatricial or mucous membrane pemphigoid. This is a rare disease caused by
various antibodies that target the basement membrane of mucous membranes
and occasionally the skin. The oral mucosa is almost always involved, most
commonly the gingiva. In contrast to pemphigus vulgaris, the variably sized
bullae are not flaccid, and ruptured bullae heal with scarring. Microscopi-
cally, there is clefting between the epithelium and the basement membrane,
and the space may be filled with serous fluid containing sparse inflammatory
cells. Direct immunofluorescence shows a linear band of IgG and C3 on the
"'All e-figures are available online via the Solution Site Image Bank.
4 I SECTION 1: HEAD AND NECK
llol,rant-1111111_.. "'-"'ec>llt.lllaleoid>orrta
Squ.-noua ail c:at:homa OlrcmiNI .. pioom«plilc: adenoma
V«ruca:u:c.n:ttoma lillllj'l ..aa.II•JII-
S..IDU oquemout col...,..,,....,... PioornOI!>IIIc adOIIOI!III
Polllla!Y&Qllllm0114 col Cllel1101110 ,.,_illlellomo
Sl>llldlo col ClleiiiOitlO 8&.11!11 01!1!1 t!rthltiOMil
Aoo11111otllle sq........,.oeuc:o-• Olndcu!ar _,.ma
AIJ~LWnOUl t~~~n::!l'd'M lluclal I"PIIbma
<:arcmm• <Uiialil1llm C)oblllo-
Lfmpl\oop!ll\11111 Qll<tloml
SIIUI--
III•U•Ipr-lllln K.epo~l's......,ma
__,. _
~ ..allolla!- Lymplla...,ma
Pa!lllomu EdomOMIIcllyrnai.,_"""YJ'>Id l!.mor
Squtmoua coli P<l>lbna and'""'""' Foc::el«al mv~
wlptla
Oonlllbno awrrlno.u.m
....
Qonpillll.,.rw!or cdlepulll
........,,__
l'll--m.a
Sd\lel)'pnd <1l<tlom ..
""r'ollo ... """"""""'
Follcu!ar t,.o!>llom.a
Earanodol merp,ol...,o -lymphoma
. , . . . , _ _ ljmpllaltlnuo
Aelllle collc:o.....,mo IMAI.ntoD&
"'""""plll&m-dd ......,_ Bultlt t~m!>lloma
T-coll t~m!>lloma (h:b!IIJ!:•••plulle
Adonolll . . "'""'""""'
l'<>tlm<>rph"" ~ a.clonocatdnoma 11'111 Ql/llympl\01'111)
Bt.•l oa!leda'loc::llrdnOI'l'lia
Eji!IIIOIII-<n)OIOpii!IOIIBI-noma
Elllra.....,loly
la'ipfl\eM..II ~It
"'""'"""'me
Clellr col Cllelnomo.IIOI CCI!ono1oo Elilra.....,liiiY ln'Jiliol:l ..rcoma
-11\od Follculllrdondilile c o l l - m «
~~ _!.,.paltno-
Mudnow:.tdlnoa.rdnorna
0nc1>q1t: co rdnoma 1-ltlf-
Sd\lel)'duct oardn"""
Chapter 1 • Oral Cavity and Oropharynx I 9
hyperkeratosis, and the sheets of tumor are composed of bland cells with
abundant eosinophilic to clear cytoplasm, sometimes described as glassy
in appearance. There is no cytologic atypia. The stroma directly beneath
the tumor typically demonstrates prominent chronic inflammation, often
with abundant plasma cells (e-Fig. 1.27).
Pure VC is a tumor type in which the cells have not invaded through
the basement membrane, and as such has an excellent prognosis. Although
the tumor can be large and locally destructive, complete surgical resection
is often curative because the tumor cells cannot spread beyond the local
site. Radiation is also an acceptable treatment, particularly in poor surgical
candidates. Conventional invasive SCC sometimes arises from VC. When
this occurs, the prognosis and behavior are the same as for conventional
SCC. Thus, the lesion must be thoroughly sampled histologically before
a diagnosis of pure VC is made.
d. Spindle cell carcinoma (SpCC} is the head and neck mucosal form of sarco-
matoid carcinoma, a variant of sec consisting of spindled or pleomorphic
tumor cells that simulate a true sarcoma. It is clear from ultrastructural,
immunohistochemical, and molecular studies that the sarcomatoid cells
represent a clone of poorly differentiated, or divergently differentiated,
carcinoma cells. SpCC has demographics similar to those of conventional
sec, occurring in the fifth to sixth decade, showing a strong association
with smoking and alcohol use, and having a very high male to female ratio.
It occurs most commonly in the larynx (particularly the glottis) followed
by the oral cavity, hypopharynx, and nasal cavity. Up to 20% of patients
have a history of previous radiation to the originating site, which is higher
than that for conventional sec.
Grossly, the vast majority of laryngeal and hypopharyngeal SpCC, and
approximately 50% of oral SpCC, have a polypoid growth pattern result-
ing in an exophytic mass with a smooth and extensively ulcerated surface.
Up to 75% of SpCC are biphasic tumors with areas of conventional SCC
admixed with areas of spindled and/or pleomorphic tumor cells (e-Fig.
1.28). The spindled component usually predominates. The conventional
squamous component may take the form of squamous dysplasia, carci-
noma in situ, or invasive carcinoma. Because the tumors are usually exo-
phytic with extensive surface ulceration, the noninvasive component may
be only a focal finding or may be effaced altogether. SpCC has a wide vari-
ety of architectural patterns including fascicular (e-Fig. 1.29), storiform,
lace-like, or myxoid areas of growth. On occasion, the tumor cells may be
widely spaced in an edematous stroma mimicking granulation tissue with
cytologic atypia, which is a major diagnostic pitfall. Approximately 5%
of tumors will have definable heterologous sarcomatous differentiation,
either osteo- or chondrosarcomatous. Immunohistochemistry in the spin-
dle cell component is positive for cytokeratins and/or epithelial membrane
antigen in approximately two-thirds of cases, and positive for p63 in a sim-
ilar percentage. Vimentin is always positive, and a significant minority of
tumors will be positive for smooth muscle actin.
Although any malignant spindle cell lesion of the mucosa of the upper
aerodigestive tract should be considered an SpCC until proven otherwise,
the differential diagnosis includes a true sarcoma, spindle cell melanoma,
nodular fasciitis, and ulcers and granulation tissue with reactive atypia
(particularly after radiation). When a conventional squamous cell carci-
noma component is present intermingled with the spindle cells, the diag-
nosis of SpCC is confirmed without the need for additional studies.
Because SpCC is inherently a carcinoma, current treatment recommenda-
tions are essentially identical to those for conventional sec, and taking all
Chapter 1 • Oral Cavity and Oropharynx I 13
patients together, the prognosis does not appear different from conventional
SCC. However, the prognosis is worse for oral cavity SpCC, and patients
with endophytic tumors do worse than those with exophytic tumors.
e. Papillary SCC is a rare variant of SCC that is defined as more than 50%
of the tumor having an exophytic, papillary growth pattern (e-Figs. 1.30
and 1.31). It is more common in the larynx than in the oral cavity and
oropharynx, and has a good prognosis.
f. Adenosquamous carcinoma is another rare variant consisting of a mixture
of sec and adenocarcinoma with true gland formation, often with mucin
production (e-Fig. 1.32). The oral cavity and oropharynx are less common
sites than the larynx. These tumors are typically more aggressive than
conventional sec.
g. Basaloid squamous cell carcinoma (BSCC) is a variant that is rare in the
oral cavity but is slightly more common in the oropharynx, larynx, and
hypopharynx. It is characterized by molded nests forming a "jigsaw" pat-
tern. The tumor cells are basaloid with hyperchromatic, round to oval
nuclei, and scant cytoplasm (e-Fig. 1.33 ). Extracellular mucoid or hyaline
material may be present. Abrupt squamous differentiation or overlying
squamous dysplasia is seen (e-Fig. 1.34). While generally this tumor is
an aggressive variant with high rates of distant metastasis, BSCC in the
oropharynx is frequently HPV positive and when so, has a better progno-
sis than BSCC at other sites.
B. Melanocytic. Melanoma is not uncommon in the oral cavity. It occurs in adults
with an average age of approximately 60 years, with a very even incidence
from age 20 to 80 years. Unlike cutaneous melanoma, in which sun damage
underlies the development of most cases, no major etiology has been identified
for oral lesions. There is a slight male preponderance and, also unlike cutaneous
melanoma, oral lesions occur relatively equally among numerous races. Oral
melanomas present as incidental pigmented lesions identified by a dentist or
physician, as masses arising in a preexisting pigmented lesion, or most frequently
as a new mass growing over a few months. The most common oral site is the
hard palate ("'40%), followed by the maxillary gingiva ("'25%) (e-Fig. 1.35),
the buccal mucosa, the mandibular gingiva, and the lip. Melanomas of the
oropharynx are rare.
Grossly, most tumors are heavily pigmented and heterogeneous, with a
brown, gray, or black color. There are sometimes satellite lesions without inter-
vening pigmentation. Microscopically, most melanomas are deeply invasive,
but two-thirds retain a surface in situ component as well. Architecturally, the
tumors consist of single cells and ill-defined sheets and nests of either epithelioid
cells, spindle cells, or both. The large epithelioid cells have abundant cytoplasm
(which can be eosinophilic to gray) and round to oval nuclei with a characteristic
single, large, cherry red nucleolus (e-Fig. 1.36). Melanin pigment is commonly
present. Spindle cells are less common and have cigar-shaped nuclei and mod-
erate dear to eosinophilic cytoplasm. Plasmacytoid and rhabdoid cells can also
be seen. Intranuclear cytoplasmic inclusions that are typical of melanomas at
other sites are uncommon in oral melanomas. By immunohistochemistry, oral
melanomas express the same proteins as other melanomas, namely Melan-A,
MART-1, HMB-45, S-100, and tyrosinase.
Treatment consists of radical resection with radiation, with or without
chemotherapy. Neck dissection is performed only for clinically detected dis-
ease. The prognosis is poor. Numerous studies have demonstrated a collective
5-year survival between 15% and 25%. Breslow thickness, unlike cutaneous
lesions, is of very limited prognostic utility. Approximately 40% of patients will
develop cervical lymph node metastases, and distant metastases are common.
Major sites include the lung, the liver, and the brain.
14 I SECTION 1: HEAD AND NECK
C. Neuroendocrine carcinomas are uncommon tumors in the head and neck region
in general, and are very uncommon in the oral cavity and oropharynx. They
are essentially all high grade with a small cell morphology, composed of cells
with scant cytoplasm, crush artifact, granular chromatin without nucleoli, and
extensive necrosis with brisk mitotic activity (i.e., morphologically identical
to small cell carcinomas of the lung and other organs) (e-Fig. 1.37 and e-Fig.
1.38). Some are mixed with a component of SCC. In the oropharynx, recent
studies have shown that roughly one-half of tumors have been shown to have
transcriptionally-active HPV. Carcinoid tumors (low-grade neuroendocrine car-
cinomas) almost never occur in the oral cavity and oropharynx.
The prognosis for high-grade neuroendocrine carcinoma is very poor, with
rapid progression of disease including the development of cervical lymph node
metastases and distant metastatic disease. This seems to still hold true for
patients with HPV-related oropharyngeal neuroendocrine carcinomas as well.
D. Vascular
1. Pyogenic granulomas (or lobular capillary hemangiomas) are benign lesions
of the oral cavity that are usually solitary, and are most common on the
lips, tongue, and gingival and buccal mucosa. They occur in patients of all
ages but have a particular predilection for the gingiva of pregnant women
and, thus, are often referred to as a "pregnancy tumor" in this setting. They
usually present as small (a few centimeters or less) exophytic masses that
bleed easily. They may grow rapidly and cause false clinical concern for a
malignant neoplasm.
Grossly, they are polypoid or pedunculated, pink to red, and have a
smooth surface. Microscopically, they consist of lobules of small capillaries
with plump endothelial cells that have round to oval nuclei and occasional
mitoses (e-Fig. 1.39), with larger central "feeder" vessels. Extensive surface
ulceration with associated fibrin is usually present. The cells are positive for
endothelial immunohistochemical markers such as CD34, CD31, and factor
Vlll-related antigen.
Pyogenic granulomas are benign neoplasms that are cured by simple exci-
sion. A small percentage may recur. Pregnancy-related lesions often regress
postpartum and thus may be hormonally driven.
2. Hemangioma and lymphangioma are benign tumors composed of abundant
blood or lymphatic vessels, respectively. Hemangiomas of the oral cav-
ity usually occur in adults. They occur most commonly on the lip, buccal
mucosa, and lateral tongue borders and present as painless, nodular or well-
circumscribed, red or blue masses (e-Fig. 1.40) measuring <2 em in maxi-
mal dimension. Microscopically, they consist of blood vessels ranging from
small capillaries to large cavernous spaces. The endothelial lining cells can
be plump and may have mitotic activity, a feature more common in children.
There are a number of named histologic variants, most of which have no
clinical significance.
Lymphangiomas (or cystic hygromas) are composed of dilated lymphatic
channels. About 75% occur in the head and neck and, when presenting in the
oral cavity, they are almost always found in children younger than 3 years.
Histologically, they typically consist of very dilated lymphatic channels lined
by bland, inconspicuous endothelial cells, with intraluminal eosinophilic
material, lymphocytes, and occasional red blood cells. They often are inter-
stitial aggregates of lymphocytes.
Both hemangioma and lymphangioma are benign lesions cured by conser-
vative excision. Hemangiomas can be treated by sclerotherapy as well. Large
lymphangiomas are often de bulked, often via serial resections to avoid major
morbidity.
3. Kaposi sarcoma (KS) is a locally aggressive tumor uniformly associated with
human herpesvirus 8 (HHV-8) that predominantly involves the skin but can
Chapter 1 • Oral Cavity and Oropharynx I 15
also involve mucosal sites. When KS involves the oral cavity, usually as a
complication of AIDS, it is most commonly found in the palate, followed by
the gingiva and dorsal tongue. Clinically, KS appears as purple, red-blue, or
brown macules or plaques. Later in the disease course, the lesions become
nodular and may ulcerate.
The three histologic stages of KS (patch, plaque, and nodular; all dis-
cussed in more detail in Chap. 39) can all be associated with oral KS (e-Fig.
1.41). There are frequently associated collections of extravasated red blood
cells and hemosiderin-laden macrophages. A characteristic feature that is
sometimes seen is the pale, eosinophilic hyaline globule, which probably
represents degenerating red blood cells. Immunohistochemistry is positive
for the common endothelial markers CD34 and CD31.
The behavior of oral KS is variable. It is generally indolent in nonim-
munocompromised patients but is more aggressive in patients with AIDS.
However, the mortality related to KS is highly dependent on other comor-
bidities such as opportunistic infections and systemic symptoms.
E. NeuraVneuroectodermal. Granular cell tumors are benign, slow growing tumors
of neural origin that occur at many anatomic sites. Approximately 50% occur
in the head and neck region, and half of these occur in the tongue. They also
occur in the buccal mucosa, floor of mouth, and palate, are twice as common in
women as men, and approximately 10% to 20% are multiple. Grossly, they are
smooth, sessile, and firm with a pink or tan-white color. Microscopically, they
consist of infiltrative nonencapsulated sheets and cords of bland cells with abun-
dant eosinophilic granular cytoplasm and indistinct cell borders (e-Fig. 1.42).
There is usually no significant stromal reaction. The nuclei are small, oval, and
hyperchromatic with minimal atypia and no mitotic activity. A common feature
is pseudoepitheliomatous hyperplasia of the overlying squamous epithelium,
which can closely mimic SCC. Conservative excision is the treatment of choice,
with a risk of recurrence of <10%. Malignant granular cell tumors (as covered
in the Soft Tissue chapter) are very rare but do occur.
F. Mesenchymal
1. Peripheral ossifying fibroma is a reactive proliferation of fibrous tissue on the
gingiva which shows focal bone formation. It occurs over a broad age range,
but young adults are most commonly affected. The lesions range from a few
millimeters up to 2 em in maximal dimension. They are essentially exclusive
to the gingiva, particularly along the incisors, and present as sessile pink
nodules, usually with surface ulceration. Microscopically, they consist of
randomly distributed plump (but not atypical) fibroblasts with foci of miner-
alization ranging from dystrophic calcification, to cementum-like material,
to well-formed bone (e-Fig. 1.43). Rare giant cells can be seen. The lesions
should be excised down to the periosteum but will recur in 15% to 20% of
cases.
2. Peripheral giant cell granuloma is another reactive proliferation of the gingiva,
particularly along the incisors, caused by chronic irritation. It presents over
a wide age range, particularly in middle-aged to older adults, as a solitary
broad-based nodule that is reddish or blue and <2 em in diameter. Micro-
scopically, it consists of a mixture of multinucleated osteoclast-like giant cells
and plump spindled to oval mononuclear cells (e-Fig. 1.44). Hemosiderin,
chronic inflammation, and foci of metaplastic bone are frequent. The differ-
ential diagnosis includes brown tumor of hyperparathyroidism, cherubism,
and central (intraosseous) giant cell granuloma. The lesion is treated by local
excision down to the bone. Recurrence occurs in approximately 10% of
cases.
3. Congenital granular cell epulis is a rare benign mesenchymal tumor that clas-
sically arises from the anterior alveolar ridge of a newborn. Girls are more
frequently affected than boys by a ratio of 9:1. The tumor presents as a
11 I SECTION I. HEAD AHD NECK
'' 1.''! I Ull llle 2010 AlCC hlflll G'Mdeli!IIM for"'- <A 1lle 0111 CD1V.,.. Up
"'-'a-m
P"'-iY"'-<:el'lllll be • - (11()
rw-111 polmuylum0f(11))
eo,..,.,.."' .au8J'M1Mt
1\JmOf :;2 ern In
('lis)
<III\IJIUn <Til
1\JmOf >2 ern but not ><I ern In I l l -dim..- (T2)
TumOf >4 an In JP'OII8II dim- (11!)
-111tt..t.- bcald-(T41)
(Otoi<:O>Ity) Tunvln¥11dao ocljaalnt lllrudum oni)' (....., lhrouJ,!I Cllftlcll - , lnlo lloo9
ISctliMicl ll'oiiOCio of'>nlUO ~"'· hYollooout I>Oiaqjooouo.llld ~t
l!lllllllluy'*'"" ol(l'l of feool
~ 1\Jnvln""'MIIIrtJ<>.I!jl-bolt ..,,,_,• .....,,........, ftoorofmo\1111, Of akin t1
r- (thin or nOM)
Vff'/ ocl¥1ntod lealI d - (T~bl
Tumor k\Widnu~r tpec:o. pla,.,.:fd ~or atul blnoandlorenc:atOS ~ Clln7tld
•"'IY
...,..., .... -Oil
~·~~lymph na~ .. OUinct
rw ...,..., tlml!ll noclo m - · •
be._. . (Nl()
,_..,_,a''""' (NO)
lpalfllloral lymph nodo, ~an
In lll"''fttO!dlnonllon (Nil
,_..,In• '*'ll)olpallidofal lymph noda. >3<m but not >6tm In .,.n.stdmonalon (N:lol
Moll :1>* 1'1 mutilllo ~I ttml!ll nodoo. nM& >6ct0 1'1 -'>Otdlnenololl (~(~!))
Mo.!O' '''* .,_
bltJbrtllll tiOl~ ~ph nodi!~!., I'IOI'IIIl >6em lr. Wll!ltttMtdm..wol1 (N2t!)
Mo.!tu••+ .,_a tjTrtptt nadtt >6 c:rn In S16ft'dt dlrtwd:fon (N3)
ltlbll • 1ntnll (II)
fWdlllllnt mllllll•* (MOl
Dlollnt rn""a ... (Mil
tap""""'''
Tho- f)<l!ldog1c AJCC -~~
1IIINOIMO Oltllla 0)
TIJNOIIII) (Stop U
T2INOifll) (Stop I0
~CS,...IID
TIJNIJIII) ~liD
T21NIJIII) ~liD
T3/NIJIII) 0!\let liD
T4IINOifll) (S1j;p IVA!
T41/NIJIII) (S1j;p IVA!
TIIN2/MO cs,... Ml
T2JN2/III) ~lilA)
TSJtWIII) ~lilA)
T4I/N2/III) (ll1aao IVA)
T4111Any NlloiO (Slap IVBl
Any TINii/MO !SIIill!IVB)
Any T/Any HIMICSII.Io IVCl
·~~~ t!OM c1 bc:nWbx:thacdlet by 11,.,..1 ~tt'feat'Ch:rM Ia: I'd: 8l.l'f'fdernl: ID c:•~t,
6tu/Mf'MT4.
F.,,. £<W& SB, B)<d DR, Cool"""
0C:, .. tl, d . AJCC Clw8r~- 7111 ed. NM Yi>lt,
NV,~~-. 2010. llood olth p e - .
,,, , ..... (!)
Pmwy 11/mor <:~nnot bo -lOCI (11()
No -noaa! prfrneiJtumllf (TO)
a-
Cor<b>mt1 lh o!IU fllll
1\Jmor :;2 «n In a -dmenolon (Til
1\Jmot >2""' blt not ><I""' In.,._ <Imendon ('12)
Tumor :.<1 an In . - < Imendon tD tile lnpiGIIilcooftllt 'I>WOI!Il (T3)
Modnt.lt/ .odol.-~ocot-. (T4a)
Tumor -lila 1o1:1n><. d~ "">do of tonp,o. mod~l JIGt"11!HI.I•ml pelodD. or
mondl>l&
\leiJ-...d ..... dhoooe (T-41>)
1\Jmot-- r<ISI)'IIOid mwdt, P'MYIPd pio1M, ......1nUOpMIJIII. I>' 1\wl b&ot, «
.,.._..RII!d utaly
_, .. ,.,.,..-Oil'
R...,.lljmptl nodot..nnot be • - (NX)
No I'II8Sohll b'rnl>h nodo ~(NO)
Mello*"' lh aolnlloll>illlol.nlljmptl node. ;S3 em In • - dim"'*' !Nll
,...""'"In ul,..lpollftlral tlm!ltl node, >3 em llutnot:.6cm In .,...,.d_n (N2o)
Mollo*•~ ln,..,a-.lo~b'rnl>hnodos, none >Gem ln.,..talldlnlnllon (N:!b)
Mollo*•~ lnbllllotalor...,.-.olf.lmph nodoa,nono >6anln IP""'Mtdlm- (N:!J:)
Mol-In alpnpti!Do >Ocm In .,nootdmenslon (~)
lf.lllnrau•••• II)
Nod.._m_+ (1,10)
.........
Olarrtm!OP''W (Ml)
smooth, nonulcerated mass about 1 em in size arising from the gingiva over
the lateral incisor/canine area of the maxilla, or less commonly, the mandible.
Grossly, it is polypoid and has a homogeneous firm pink or tan cut surface.
Microscopically, it consists of sheets of large cells that have abundant, gran-
ular, eosinophilic cytoplasm and round to oval, bland nuclei. The surface
squamous epithelium is intact and shows no hyperplasia. By immunohisto-
chemistry, the cells are positive only for vimentin, and specifically are negative
for S-100.
Congenital granular cell epulis is not the newborn equivalent of a granular
cell tumor and shows no neural differentiation. The tumor stops growing at
birth and regresses over time, but most cases still require surgical resection.
There is no recurrence, even after incomplete removal.
V. PATHOLOGIC REPORTING OF ORAL CAVITY AND OROPHARYNGEAL MALIGNANCIES
A. Staging. The American Joint Committee on Cancer (AJCC) staging guidelines
for oral cavity and oropharyngeal carcinomas are listed in Tables 1.3 and 1.4.
Staging is extremely important for clinical management and establishing prog-
nosis. These staging guidelines are applicable to all forms of carcinoma. Any
nonepithelial tumor type is excluded. A specific and very important point in stag-
ing involves bone involvement by tumor. To qualify as a T4lesion, the tumor
must erode through the bone cortex. Superficial bone erosion is not sufficient
for classification as T4.
It is important to note that mucosal melanomas of the head and neck, includ-
ing the oral cavity, have their own staging system.
B. Additional pertinent pathologic features. As with carcinomas at all upper aerodi-
gestive tract sites, margin status, tumor differentiation, and the presence or
absence of perineural or lymphovascular space invasion should be reported.
Perineural invasion is particularly common in oral cavity carcinomas and is
correlated with a poorer prognosis. The pattern of infiltration as well as the
presence or absence of a host inflammatory response should also be reported,
because both features have been correlated in many studies with a higher rate of
local recurrence, a poorer prognosis, or both. Some recent studies have devel-
oped formal grading systems for these features that have promise for clinical
implementation, particularly for early stage oral cavity sec, but these are not
ready for active implementation. Depth of invasion, particularly for T1 and T2
tumors, although not reflected in the staging system specifically, is important
for clinical management and prognosis, and so should be reported.
SUGGESTED READINGS
Adelstein DJ, Ridge JA, Gillison ML, et al. Head and neck squamous cell cancer and the human
papillomavirus: summary of a National Cancer Institute State of the Science Meeting, Novem-
ber 9-10,2008. Washington, DC, Head Neck. 2009;31:1393-1422.
Ang KK, Harris J, Wheeler R, et al. Human papillomavirus and survival of patients with oropha-
ryngeal cancer. N Engl] Med. 2010;363:24-35.
Bouquot JE, Muller, S, Nikai H. Lesions of the oral cavity. In: Gnepp DR, ed. Diagnostic Surgical
Pathology of the Head and Neck. 2nd ed. Philadelphia, PA: W.B. Saunders Publishers; 2009.
Chemock RD, El-Mofty SK, Thorstad WL, et al. HPV-related nonkeratinizing squamous cell car-
cinoma of the oropharynx: utility of microscopic features in predicting patient outcome. Head
Neck Pathol. 2009;3:186-194.
Chemock RD, Lewis JS, Jr, Zhang Q, et al. Human papillomavirus-positive basaloid squamous
cell carcinomas of the upper aerodigestive tract: A distinct clinicopathologic and molecular
subtype of basaloid squamous cell carcinoma. Hum Pathol. 2010;41:1016-1023.
Slootweg P, Eveson ]W. Tumours of the oral cavity and oropharynx. In: Barnes L, Eveson JW,
Reichart P, Sidransky, eds. Pathology and Genetics Head and Neck Tumours. Lyon, France:
IARC Press; 2005.
Thompson LDR, ed. Head and Neck Pathology, 1st ed. New York, NY: Churchill Livingstone;
2006.
Larynx
James S. Lewis Jr.
I. NORMAL ANATOMY
A. Macroscopic/gross. The larynx is a unique organ designed to produce phonation
by modulation of the respiratory airstream. It is composed of several cartilagi-
nous structures: the thyroid, cricoid, and arytenoid cartilages and the epiglottis.
The hyoid bone sits above and is connected to the larynx by the thyrohyoid
membrane (Fig. 2.1). The thyroid (and lesser so, cricoid cartilage) ossifies in
adults. In functional terms, the larynx is divided into three subsites. The glot-
tis includes the true vocal folds or cords, below, and the false folds or cords,
above. The space between them is called the ventricle, and its deeper recess,
the saccule. The true cords, with a hypocellular stroma called Reinke's space,
are designed to vibrate for phonation, and the ventricle amplifies this further.
The cords are manipulated by muscles that attach to and move the arytenoid
cartilages, which sit at the posterior aspect of the vocal folds. The larynx can be
divided into three compartments for tumor management and staging purposes:
the supraglottis, glottis, and subglottis. The supraglottis includes the epiglottis,
aryepiglottic folds, false cords, and ventricle. Glottis refers to the vocal cords
from the edge of the ventricle to the free edge of the vocal cord. Subglottis
refers to the area from the free edge of the vocal fold to the inferior border of
the cricoid cartilage but has a slightly different definition for American Joint
Committee on Cancer (AJCC) staging (see later).
B. Microscopic. The larynx is covered by a mixture of squamous and pseudos-
tratified ciliated columnar (respiratory-type) epithelium. In smokers, however,
often the entire endolarynx is covered by squamous epithelium. The true cords
themselves are always covered by squamous epithelium. They contain a lamina
propria area called Reinke's space, which lies between the epithelium and the
vocal ligament and which consists of loose connective tissue with few capillaries,
no lymphatics, and sparse seromucinous glands (e-Fig. 2.1).* The false cords
and ventricle are typically lined by respiratory-type epithelium.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION
A. Endoscopic biopsies. The majority of specimens from this region consist of endo-
scopic forcep biopsies. The tissue samples should be placed immediately into
10% buffered formalin or other appropriate fixative. These should undergo
gross examination and description documenting the exact number of pieces
present and their size, and then be entirely submitted with three levels cut from
each paraffin block for hematoxylin and eosin (H&E) examination.
B. Resections
1. Partial. These are widely variable specimens depending on the location of the
tumor. Standard procedures include vertical hemilaryngectomy and supra-
glottic or supracricoid laryngectomy. As a generalization, these specimens
need to be oriented, the soft tissue margins inked, and margins demonstrated
by shave or radial section followed by sectioning of the tumor relative to car-
tilage/bone and soft tissue margins. The use of either shave or radial sections
depends on the nature of the specimen. If the tumor is relatively distant from
a margin, 1- to 2-mm shave sections are preferred. If the tumor approximates
a margin to < 1 to 2 mm, radial sections are taken.
*All e-figures are available online via the Solution Site Image Bank.
19
20 I SECTION 1: HEAD AND NECK
Pre-epiglottic
space (fa t)
Thyroid
C~~~"c--iJ,_-Cricothyroid cartilage
ligament
Cricothyroid
muscles
Cricoid
cartilage
(anterior)
B
Figure 2.1 Larynx anatomy: (A) anterior view; (B) sagittal cross section.
are taken from deep into the tissue. The pieces should be evaluated grossly for
mucosa-typically shiny and pink-tan on one surface of the tissue; if present,
the specimen should be oriented to demonstrate this surface on one edge of the
section with the submucosa below. Additional sections should be cut if needed
to assure that two or three quality sections are obtained.
The tissue that remains after frozen section is submitted for evaluation by
permanent sections. This process can help resolve a number of issues from frozen
section including freezing and cautery artifact, amount of tumor represented,
and orientation or embedding issues. The margins of the main resection speci-
men are also evaluated throughout its entirety because the separate frozen sec-
tion specimens are small and almost never cover the entire margin of a resection.
The final margin status is then a conglomerate of all three sources: frozen section
slides, permanent slides of the frozen tissue, and the margins of the specimen
itself.
Ill. DIAGNOSTIC FEATURES OF COMMON DISEASES
A. Inflammation and infection. Inflammation of the larynx (laryngitis) is quite com-
mon clinically, and can be divided into acute and chronic forms, which are
variable by age. Laryngitis rarely necessitates tissue biopsy for pathologic eval-
uation. Infections can be caused by a myriad of agents including viruses, bacteria,
fungi, and parasites. The pathologist must be alert to the possibility of infection,
and the immune status of the patient is helpful information, as many of these
patients will be immunocompromised.
Inflammation is essentially never present in the normal larynx, so the pres-
ence of inflammatory cells is a diagnostic clue. Depending on the organism, the
inflammation can take a number of different forms, almost all of which are typ-
ical for the type of organism when it presents in other locations. Examples of
some of the major infections include cytomegalovirus, herpes simplex virus,
tuberculosis, rhinoscleroma (Klebsiella rhinoscleromatis), candidiasis, histo-
plasmosis, blastomycosis, cryptococcosis, coccidiomycosis, or rhinosporidio-
sis (Rhinosporidium seeberi). The resulting inflammation may cause mucosal
ulceration, acute and chronic inflammation, necrosis, or granulomas. Special
stains (such as Gomori•s methenamine silver (GMS), acid-fast bacillus (AFB),
or periodic acid-Schiff (PAS)) should be utilized to look for, and characterize,
organisms.
B. Non-neoplastic lesions
1. Traumatic
a. Vocal cord nodules and polyps. These are non-neoplastic degenerative stro-
mallesions of Reinke•s space that are usually related to trauma due to
misuse or vocal excess. As such, they have been referred to as singer•s or
screamer•s nodules. They are more common in women and are commonly
bilateral, characteristically occurring at the junction of the anterior and
middle one-third of the vocal cord, as this is the point of maximal vibra-
tion during phonation. Macroscopically, they appear as gray or white
broad-based nodules or polyps (e-Fig. 2.3). Microscopically, they consist
of squamous mucosa with or without hyperkeratosis, are only rarely ulcer-
ated, and overlie a sparsely cellular myxoid, edematous, fibrous, fibrinous,
or vascular stroma. The myxoid appearance is most common and it may
demonstrate small cystic spaces (e-Figs. 2.4 and 2.5). So-called vocal cord
polyps are histologically identical but clinically present unilaterally, and
in men and smokers with more regularity.
b. Contact ulcer. Also referred to clinically as contact granuloma or just gran-
uloma, these occur on the vocal process of the arytenoids classically as a
result of forceful vocalization in individuals who must affect a low, deep,
forceful voice. However, they also may result after endotracheal intubation
or as a result of gastroesophageal reflux disease. They are more common
in men, can be unilateral or bilateral, and present as polypoid lesions.
Chapter 2 • Larynx I 23
.........
""""' rt IOINI!y I'J<nd-«ypo 01/'dn..,...
Adonolil~ ..rdnoma
,M.-plllonnold .,.
..TJI>biCII'<IIold corctloma
~loor<b>ld
Smollooll..- a , ,.......-,.~1!6
lllllla .....
Popilorna
lo,_
Comblnod ..,., coli co,_ .. TWJroondcal,.~pe
Popllorn-
SatJIJypnd.typoedonomu
Ploomorpllk: odonom•
On<:l>qlt: paplloiJ eyoolodlnoma
M*"'•-
Ma..,.rn.rmoll
AbtCJM('CI)I'rtit
""""'nl flb- hiiiSoq!Dma
u.,...""'""'
U!lomjullr....,.
""....""'""'
Rho~
Kapaol""""""'
""""''* .,.,_.h...l· - -"'"'"'
s,n..r.l..,..,ma
Bordoo1lne ..,.,..,..., moli,vlllrt potonUOI
--
Hllmmalory m)'ll!lnblullc tumllf
klllntum.,..
-blorna
u-
~lomJO>ma
RlllbOom)O>n\11
Henll/liSOml
_,.....,_
-If-. . .
~pllllliSomo
Ghl'lut.r 0&1 tumor
--
Ch~
,.........,...•.
Chon<t'oma
.......
Cion! oollll.r.,...
......,.....
Ill. .
Squamous papillomas are strongly associated with the low risk HPV
types 6 and 11, which are thought to be transmitted from the mother
to the upper aerodigestive tract (UADT) of the neonate during vaginal
delivery, thus explaining the occurrence in childhood. There is minimal
risk of transformation to invasive carcinoma.
Regardless of the clinical context, squamous papillomas have a typi-
cal morphology. Grossly, they appear as exophytic, granular, and friable
pink, red, or tan lesions. Microscopically, they consist of arborizing, pap-
illary fronds of thickened but maturing squamous epithelium with slight
hyperplasia of the basal layer (e-Fig. 2.9). Cells in the midlayer often have
cytoplasmic clearing, but frank koilocytosis is not regularly observed. The
nuclei are slightly enlarged and irregular but do not appear overtly dys-
plastic, and mitotic activity, while present, is usually limited. Character-
istically, there is minimal surface keratinization (e-Fig. 2.10). Frank dys-
plasia can be seen in some lesions and should be reported and graded as
in nonpapillary squamous mucosa (see Section m.C.2). However, there
is no consistent correlation of overt dysplasia with the development of
subsequent invasive carcinoma.
b. Granular cell tumors are benign, slowly growing tumors of neural origin
that occur in a multitude of anatomic locations. They are particularly
common in the head and neck region with a minority occurring in the lar-
ynx. They are slightly more common in African Americans and typically
present with hoarseness. Grossly, they are smooth, white polypoid tumors
involving the posterior true cords, anterior commissure, false cords, or
subglottis. Microscopically, they consist of infiltrative, nonencapsulated
sheets and cords of bland cells with abundant eosinophilic granular cyto-
plasm and indistinct cell borders. There is usually no significant stromal
reaction. The nuclei are small, oval, and eccentric with minimal atypia and
no mitotic activity. A common feature is pseudoepitheliomatous hyperpla-
sia of the overlying squamous epithelium, which can be quite alarming,
can mimic squamous cell carcinoma, and should prompt a search of the
submucosa for granular cells (e-Fig. 2.11). Conservative endoscopic exci-
sion is the treatment of choice with a less than 10% risk of recurrence.
c. Paragangliomas are tumors recapitulating the paraganglia, specialized
organs of the autonomic nervous system derived from the neural crest.
They occur in numerous locations throughout the body, and their behavior
is largely dependent on site. Laryngeal paragangliomas are benign and are
divided into two groups: superior and inferior. Superior paragangliomas
are much more common and occur in the supraglottic larynx, from the
false cord up into the aryepiglottic fold. They are polypoid submucosal
lesions. Inferior paragangliomas occur along the cricoid cartilage in the
subglottic region and often present as dumbbell-shaped lesions with both
intra- and extralaryngeal components.
Microscopically, these tumors are identical to those arising elsewhere.
They consist of sheets and nests of polygonal to spindled cells with abun-
dant eosinophilic to slightly basophilic cytoplasm, and round to oval
nuclei with a slightly granular chromatin. They are very vascular with
stellate large vessels throughout (e-Fig. 2.12). Nuclear pleomorphism may
be striking, but there is minimal mitotic activity. They are arranged in
nests (classically termed zellballen) (e-Fig. 2.13). By immunohistochem-
istry, they are strongly positive for synaptophysin and chromogranin A,
virtually always negative for epithelial markers such as epithelial mem-
brane antigen (EMA) and cytokeratin, and show the typical sustentacular
(or supporting) cell staining for S-100 around the periphery of the nests
whereas the tumor cells themselves are negative.
21 I SECTION I. HEAD AHD NECK
tumors with raised edges. Microscopically, the term "squamous cell car-
cinoma, keratinizing type" is used to describe the common version of the
cancer and to distinguish it clearly from the numerous variant squamous
cell carcinoma types that exist. Keratinizing-type squamous cell carci-
noma consists of nests and sheets of cells with abundant eosinophilic
cytoplasm and round to oval nuclei, often with prominent nucleoli. Well-
differentiated tumors retain abundant pink or clear cytoplasm and often
show keratin "pearl" formation (e-Fig. 2.9). Moderately differentiated
tumors have more pleomorphism and a higher nucleus to cytoplasm ratio
in many of the cells while still retaining moderate eosinophilic cytoplasm
(e-Fig. 2.10). Poorly differentiated tumors often have single cells or small
nests of cells with more mitotic activity and less cytoplasm (e-Fig. 2.11).
Grading should be performed, although it has not been shown to consis-
tently predict the clinical behavior of individual tumors. The majority of
UADT squamous cell carcinomas are moderately differentiated. Although
frank keratin formation is commonly seen in well- and moderately differ-
entiated carcinomas, its presence or absence does not have any clinical
significance for laryngeal carcinoma.
i. Verrucous carcinoma (VC) is a specific, well-differentiated, and non-
metastasizing variant of squamous cell carcinoma. First described
by Lauren V. Ackerman in the 1950s, it has thus also been called
"Ackerman's tumor." It occurs in the larynx and oral cavity and grossly
appears as a well-circumscribed, warty, and exophytic, broad-based
white, granular, and friable mass. Microscopically, VC consists of very
thick, dub-shaped papillae with broad pushing bases. These blunt and
downward-pushing projections have sometimes been likened to "ele-
phant's feet." There is usually prominent surface hyperkeratosis, and
the sheets of tumor have bland cells with abundant eosinophilic to clear
cytoplasm, sometimes described as "glassy" in appearance. There is
no cytologic atypia, mitotic activity is very low and basal, and there
is a smooth interface with the stroma. This latter feature is because
the individual tumor cells have not breached the basement mem-
brane. The stroma directly beneath the tumor typically demonstrates
prominent plasma cell rich chronic inflammation (e-Figs. 2.18, 2.19,
and 2.20).
The prognosis for pure VC is excellent. Although it can be large
and locally destructive, complete surgical resection is often curative.
Radiation is also an acceptable treatment, particularly in poor surgical
candidates. However, routine invasive squamous cell carcinoma some-
times arises from VC. When this occurs, the prognosis and behavior
are the same as for typical squamous cell carcinoma.
ii. Spindle cell carcinoma (SpCC} is the term for a poorly dif-
ferentiated carcinoma that adopts a sarcomatoid, spindled, or
mesenchymal-appearing morphology but is, nevertheless, of epithelial
origin/differentiation. It is frequently biphasic with a spindled com-
ponent and intermingled either in situ or invasive squamous cell car-
cinoma. The tumor has the same demographics as routine squamous
cell carcinoma.
Grossly, SpCC is characteristically polypoid with a smooth, ulcer-
ated surface. The glottis is the most common laryngeal site. Microscop-
ically, these tumors can be quite variable, but typically consist of sheets
of spindle cells mimicking a fibrosarcoma or malignant fibrous histi-
ocytoma (e-Fig. 2.21). Sometimes they consist of pleomorphic, hyper-
chromatic cells widely separated in an edematous stroma (e-Fig. 2.22).
There is usually brisk mitotic activity and necrosis. Foci of recognizable
28 I SECTION 1: HEAD AND NECK
or-
lwldootloouoo be!I<Mtllo lai)'I'OC !e.t.. tii>CIIoo, oo1t11oouoo of hOCk ~lnldaoP
Qti!Mic mUICioo of111olon1110. st111> muocloo, 11\YI'OI:I, "'~
TAb Y,.YedwnOid loci! ....... TunwiiW8dot .,.......mbnll•- ..,.,._.,.ltllld
OftOIY,
rnodl&aiNI_.,.
.._ ...l'IIIIPII- (II)
r«
NO
flcPnol ~ph ncdos ""''"*
bo ••"""
No........,., ~til node - •
Nl M-.ol& In a &1/lllle 11JIIfllllnll ~ph nod!,~""' In --dlmonolon
~ M-.ol& In a &1/lllle 11JIIfllllnll ~ph nod!,>$ ern bill <6«n In -wtdlmonolon
~ Molululs In rnLII!plo lplllrtlnolljmp! nodM, non& >6 ern In .,.....W<I-
rt2r:: Molululsln blllllnlorco-lljmp! nodN, ...,..>Gem lntr-dlmonolon
rG M - . l t In 1 ~ph ncdo >fi em In 1 1 -dlrnombl
12 I SECTION I. HEAD AHD NECK
I' 1,1,! I Jill 2010 AIMIIclft Jolltt c.mmm. Oft ca- Stall Ill Cukllllnetllor
' _ Tu- of111e I.MJ!'« (CN~f&Noil
UTUT.:U.SIMIIOO
Ml( - - •. .nnotbee-
MO No d11\111trn<II-
MI
STMUI.IUtll --·
""' ..... , pollool .... .u:c .....
Slqj:IO n. NO MO
SQII T1 NO MO
SQIII '12 NO MO
fllqolll TS NO 1.10
Slqtlll T1 Nl MO
SIQt Ill '12 Nl MO
Slqj:llll T3 Nl MO
SQI~ T4o NO MO
SQI~ T4o Nl MO
Stqom. T1 N2 1.10
SlqtriA '12 N2 MO
SlqtriA T3 N2 MO
SIQ:I PiA T4a N2 MO
SIQ:I Ml Toll> AtoyN MO
SQ>Ml AllYl N3 MO
fllqo IYC Any T AA,<N 1.11
SUGGESTED READINGS
Brandwein-Gensler MS, Mahadevia P, Gnepp DR. Nonsquamous pathologic diseases of the
hypopharynx, larynx, and trachea. In: Gnepp DR, ed. Diagnostic Surgical Pathology of the
Head and Neck. Philadelphia, PA: W.B. Saunders Publishers; 2009.
GaleN. Benign neoplasms of the larynx, hypopharynx, and trachea. In: Thompson LDR, ed. Head
and Neck Pathology. New York, NY: Churchill Livingstone; 2006.
Slootweg PJ, Richardson M. Squamous cell carcinoma of the upper aerodigestive system. In: Gnepp
DR, ed. Diagnostic Surgical Pathology of the Head and Neck. Philadelphia, PA: W.B. Saunders
Publishers; 2009.
Thompson LDR. Malignant neoplasms of the larynx, hypopharynx, and trachea. In: Thompson
LDR, ed. Head and Neck Pathology. New York, NY: Churchill Livingstone; 2006.
Thompson LDR. Non-neoplastic lesions of the larynx, hypopharynx, and trachea. In: Thompson
LDR, ed. Head and Neck Pathology. New York, NY: Churchill Livingstone; 2006.
Zidar N, Boffetta P. Tumours of the hypopharynx, larynx, and trachea. In: Barnes L, Eveson JW,
Reichart P, Sidransky D, eds. Pathology and Genetics Head and Neck Tumours. Lyon, France:
lARC Press; 2005.
Nasal Cavity,
Paranasal Sinuses,
and Nasopharynx
Heather N. Wright and James S. Lewis Jr.
I. NORMAL ANATOMY
A. Nasal cavity. The normal sinonasal region consists of the central nasal cavity,
paired bilateral paranasal sinuses, and the nasopharynx. The nasal cavity con-
sists anteriorly of the nasal vestibule, the small hair-bearing region just inside
the nasal ostia, with the remainder representing the nasal antrum; the nasal
cavity has four walls, a central dividing septum, and paired upper, middle, and
lower turbinates. The nasal vestibule lining is an extension of the surround-
ing facial skin, and as such has a stratified, keratinizing squamous epithelium
with associated dermal appendages and haiL It extends for 1 to 2 em into the
nasal cavity. The nasal antrum is lined by pseudostratified ciliated columnar
(respiratory-type) epithelium of ectodermal origin referred to as the Schneide-
rian membrane (e-Fig. 3.1).* The submucosa consists of minor salivary gland
mucoserous glands embedded in fibrovascular connective tissue with small ducts
that convey their secretions to the surface. The turbinates have a more richly
vascular stroma. The roof of the nasal cavity contains the cribriform plate with
olfactory mucosa, a modified respiratory-type epithelium with olfactory nerve
cells, and supporting cells.
B. Paranasal sinuses. The paranasal sinuses consist of the maxillary (largest),
frontal, sphenoid, and ethmoid sinuses. They drain into the nasal cavity and
are air filled, intraosseous, and open. The ethmoid sinuses are small and com-
plex (referred to as the ethmoid labyrinth or air cells). All paranasal sinuses
are in continuity with the nasal cavity so they have a similar, although thinner,
mucosa. The submucosa is also thinner, looser, and less vascular than in the
nasal cavity, although it does contain prominent seromucinous glands.
C. Nasopharynx. The nasopharynx is the most cephalad portion of the pharynx and
is a cuboidal structure. Its roof is formed by the pharyngeal tonsil. The lateral
walls are the most pathologically important because they contain the openings of
the Eustachian tubes, and a depression posterior to the torus tubarius called the
fossa of Rosenmiiller. The fossa of Rosenmiiller is the most common site of origin
for nasopharyngeal carcinoma (NPC). Since the nasopharynx is surrounded by
bone and vital structures, it is poorly accessible for surgery. The epithelial lining
consists of a mixture of stratified squamous, intermediate (or transitional), and
respiratory-type epithelium.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION
A. Endoscopic biopsies. Most of the specimens from this region consist of endo-
scopic forcep biopsies. The small tissue pieces should be placed immediately
into 10% buffered formalin or other appropriate fixative. If there is a suspicion
of lymphoma, a minimum of three biopsy passes should be submitted in saline
or RPMI medium and send directly for an appropriate hematopathology work-
up. Standard formalin-fixed specimens should undergo gross examination and
description documenting the exact number of pieces present, and then should
*All e-figures are available online via the Solution Site Image Bank.
34
Chapter 3 • Nasal Cavity, Paranasal Sinuses, and Nasopharynx I 35
('!Unp
Cli~ U.uollla, muo
lollon on IIIIO.P1t-
Ull.llltf Ol1l NlUl
~ll'otrllumlnll
oo~-..ortunpJ
hJlii>M; rnlnmol
~
• • - • • tun/il:
8Jpdul.t, ltJIArrJMI#,
~>~r> ..-.tty; Ullllly mUCUS; mlnm.l Oltwlflllt
""-
110110~ lnllornma1!on ~
~
,......
In-
_.....,.iilnwti&;
tevef; l'llliSI!il
Nee"'*; thrombolla;
hetn""""e';
Common: Muool'l!liel:
order-Nwt~~i
tunpi -llfOICOJiu at ~tunpl Riii>Dp<Jt. - .
Vii.ii!Ua nOIII'*'tlc dliflcb hyplioo In Wible ~
~mlnlmol Unoommon:
lnllornma1!on ~
,,....""
Chn~~c
In-
tunpi
Gtluollll
-·-- "....-
Of'llOI; ntUIOki< or
011>11.11 dolll:b; , _
onfnlql"'
Nee"'*; •nP>I•- ldpNgJ/biS
IUrclil!yplioe;
Cl.itWWit, NlsmaiiJJ
38 I SECTION 1: HEAD AND NECK
..-.
1111 .... _ .. 1111_ .. G-~
Squunouacol..,lnana (SinonouWJpo ...........pooleytoml)
~ Bd:rlploiu:ol.lldlltlry flb""'" 111...,..
f'IIDioiYII<UIArnWO col Cllclft...,.
Baooiold II<IU.rnWO col Cllclft...,. ,.,_,.
Belil!ll1tlnoll
Splndlt<d ..rclnoma La!Aii)QM
NJIIfiOiqvlll"'IOUI carc:fnana lioomllrclome
Aclnt!dj11; "'~"""""' c:dl<:~l<tlome Scllwtnnoma
~plooojlllluoilll Cftlnan.t Nouraftl>!oma
Stlot\•1 urwlffef•t'li!lt&d tardntf'nll
Ad ...... ..mma ""'·~
1ltlt1tflboltllll•llllp
,.,.~ ..1-(iii>O....__.
~-~adenoeo....,.mo lllall&nont tum ore
<:horld"""""'""
SI...IY...n~<:~l<tlomu
""""""'11yn\el<llond"""""'me
Adn>ld oystl:: ..- . O.C.••u....,a
Ac:lnlc: col - m o Cllotdoml
Mucooptlormolll ..- . llonfl11tlno111
El>~~olol m)<llptilei~ICirdum•
Gllnt<d-
a.r <Ill""""""""' not tllhei"IOIM
OIIOdllod
GIInt<dturnOf
htl<>ol>llllololoo-•
<:horldroma
~
C.l<tloi!\IIGX pooiT\O(pHc - " " " ' Cl>ondrabluloma
""~ lclw-l>"&llo ~1W11)7dd flbnxna
adeonocltt:lliOmt Otleocllondroma (Joolo!Dols)
Nourcondccmo tumm
Otleold-·
"l'fl>lc:elwel"'**
Alyplc:ol-lnold
Slnol coiloo....,.mo. neu-.doalfto
IY1>0
llllflttllbollalti-
SMn•ll'll>-
--·
Ooleol>-
teooldlcifld~ooencltJrnol 1\om-
llrnlll-aJIIIN-
E:ldr..-1'111111111 klkr (NIQIT....
lymp-
lnvoo!JdflOI>I- llll'vtt 111110 B<ollymphoma
(Sdlnolcloolln popilloml, I.-ted \ill"')
fllll••lldulluy plasmll$ml
Onooeyllopor6>TM
(Sdlnolclnn popllomo. on~ IY1>0)
!lloi>Mc DOI>Iofna
(Sdu\ddoolln popllomo. -llyllc IY!>Ol
~ ..
l!itr.....:!ulluy M)OIIIold .._,.
""""'
...............,
Uli\IIIIIIOno coil h~
SI...IY...nil-typl&llon..,...
Ploom<OpHc adonomo e..tna .tl8f00me.
htf<>ol>lltlolomo Prlml:ht& niiiUmec:tDdermlll bmor
1111--
Onooe:l'>m•
Mall&noftt tull"lln
Fb.-no
Oloclo!Y MII"O-Ml
Ml~roodenou-.tumtlffl
.....-•wlh
lnfaroey
,_
Mu....l moi!Niit molotlorno
Moll&nont flnut hllllo<)4oma
Ldornyo~~""""" IIM"IIIurelo<Rimo
Rhlbdom)'OUrcoma moll,lnonttranll'mnlltion
Anprcoma Shinual)'>l<ac:tuMOf~l
Molpllt r-!flhn,..,., .,_tum..- sl,....luMOf)
Bon!olfnund ""'TM..,.rt po!M111al Sh:lft lllil!ll b!IIVIbX:I!Iii'C!I'ICI.dlreol"'''ll
!UmOIO Md.lr& trera:t.rna
·-""-
Dtomol:kll!O ~brolroo1ooll Dennold C!'lt
lnlllmlnllblly fii)'OIIbro- tumo<
4D I SECTION 1, HEAD AHD NECK
''lhiJJII Schllllk11111111'1$1il-
r.,.
E'loolllt&
-••IDelllfaa
Haloolltlln
....
E'loolllt& flu\d$ of b~ lid
elilleiiJ-UOIII-
...,.wier
LoceJ r~rtence:
-.doitt 110 rW< d
tn!lna!ISor'lllll, M M!lphtory1YJII!I; muM! Olldn"""'
1-plllollll-plllk and
mll:rcqob; llllft tt..mont
mombf.,..
Urlotel ....1Will; Puoll-.1. ••~ , _ ct1 LoceJ reu~rtence:
"""""" I>I/IIIIMIIIIot.u... lliolld eDIIloiiJm-«UIIIItiOUO.
tn!lna!ISor'lllll, M M!lphtory1YJII!I;
~~~!Ilk of
muM! Olldn"""'
lea•or -I\)1Sc COMJlOftM\ al
Umet; trtr..aplthelll
n~~Utro!t!lu nd ml::toqob;
lhl'l lle.o<melrt rnembtaneo
~ Urlotel ....1Will; E'loolllt&•hd ond~IIYtlc LoceJ reu~rtence:
~u..... ~rrw nMta of «lttmner. -s'l!-15'11 ~~s~cot
··~ ..l k - ahl.lldont, muM! Olldn"""'
.,.....,. -pltlc
~ H111011nh•l
n~~Utro!t!lund ml::toqob
Chapter 3 • Nasal Cavity, Paranasal Sinuses, and Nasopharynx I 41
different staging system that reflects this unique behavior (see AJCC stag-
ing below). Finally, despite presenting with advanced disease, nonkera-
tinizing NPC responds well to systemic therapy (5-year survival of approx-
imately 65% in the United States). Keratinizing NPC, although it more
often presents with localized disease, does not respond well to therapy
(5-year survival of approximately 20% to 40% in the United States).
c. Squamous cell carcinoma/nonkeratinizing carcinoma. Although squamous
cell carcinoma is the most common carcinoma in most head and neck
anatomic subsites, it constitutes only approximately 65% of carcinomas
in the nasal cavity and paranasal sinuses. When it occurs in the nasophar-
ynx, it is designated keratinizing squamous cell carcinoma (WHO Type 1;
see section III.C.2.b above and Table 3.4). Keratinizing, or typical, squa-
mous cell carcinoma of the nasal cavity and paranasal sinuses has the
same morphology as that occurring elsewhere in the upper aerodigestive
tract. Squamous cell carcinoma of this subsite, unlike other head and neck
subsites, has only a modest association with smoking. Howevet; tumors
have been related to other exposures including nickel, chlorophenols, and
textile dust.
A distinct variant of nonkeratinizing squamous cell carcinoma occurs
in this region, the so-called cylindrical cell (Schneiderian or transitional)
carcinoma. The recent WHO classification simply terms it nonkeratinizing
carcinoma. It has a papillary configuration with ribbons of invaginating
tumor composed of pleomorphic cells without keratinization. These rib-
bons have a smooth border without clear stromal infiltration, which makes
identification of frank invasion difficult (e-Fig. 3.19). This tumor type has
been associated with HPV and is also reported by several authors to have
a better prognosis, although the latter contention is still controversial.
d. Salivary gland-type tumors constitute a small percentage of sinonasal neo-
plasms. They arise from submucosal seromucinous glands. The most com-
mon are adenoid cystic carcinoma and pleomorphic adenoma, although
almost all other types have been described (e-Figs. 3.20 to 3.22). They are
morphologically identical to their counterparts elsewhere.
e. Adenocarcinoma. Primary non-salivary gland-type adenocarcinomas of
the sinonasal region are uncommon. The WHO classifies them as intesti-
nal and nonintestinal types, the latter of which are subdivided into high-
and low-grade tumors.
i. Intestinal type. Primary intestinal-type adenocarcinomas (ITACs) of the
sinonasal region typically arise from the ethmoid sinuses or high in the
nasal cavity. They have a strong association with long-term occupa-
tional exposures, specifically wood dust (in carpenters), leather dust,
nickel, or chromium compounds. The latency period is several decades.
Microscopically, these tumors recapitulate gastrointestinal adenocar-
cinomas, including very well-differentiated tumors resembling colonic
adenomas, typical "colonic-appearing" tumors with columnar glands
with or without mucinous differentiation (e-Fig. 3.23 ), and high-grade
tumors with a signet ring cell morphology.
By immunohistochemistry, they are positive for cytokeratin 20,
CDX-2, MUC2, and villin, similar to gastrointestinal adenocarcinoma.
Cytokeratin 7 is variably positive and carcinoembryogenic antigen
(CEA) is usually positive. As such, they stain essentially identical to
gastrointestinal adenocarcinomas, so it can be very difficult to separate
them from the rare metastasis to this region from a primary gastroin-
testinal adenocarcinoma.
ITACs should be graded from poorly to well differentiated, and the
type (colonic, mucinous, papillary, signet ring cell, or solid) specified, as
44 I sEcT I 0 N I: HEAD AND NEcK
T4a '*'·-
-.wy.a,_locold-.o. Tumor -•-OII>bl....urtl, 111m
ololloak. p!a:ypd l*los.lnfralompo<al-.•,.,,.,,. pi<1D. ""'""ld ...
1t'ontololnuooo
T4b Ve!Yo.:h••cod locol dhoaoo. 1\Jm,..lnl'ad"'OIIYdlho tlli:NotlJ: Oil>""''"'""
dura. l:ftln, rnkkle c:n~~nW ftMa. <!1"4nl.dll r'IIW'Wil Clhar thlln rrw:ltary dl¥'.alon d
trlptnNI,.... \12, nuorA~~ "'dlwua
-~~U-IIIohlo
1)( Prtnol:yt.mor<Omotllo .... IIOwl
ro tlo <M:IenC$ol 1JiimOJY11lm ...
1k Oln:b>mlln d:u
T1 1Umor rdiStltcbd to .any onl!l.aute.a&. with or~ b:ltu~ ~lon
T2
ra
T4a
Tum... -lo
Tum... -hctwolllbolt.Din&olnilo"'lonor-naloln"""'••l.d)I<M
""""will In tilt ..-rno111o1 "'""Pioot, wtlh orwtlhout bonyI -
- l i l a madill WII 0< flOOf d thoOII>!I, - I Y t l -
i>Oioto. or cll>llfMn llioto
t.lodetat<tJ o<hror.:od locoI dr...e. 1\Jmor IWlde..l\¥ of111o tolooi\11' An1BIIor
ortitlll M'l'lerTI:IJ .dcfn d rt0&6 e~d\flllllllk. m.._....., *ldl!!lftlbft -. ..ft'lretb' <:I'll lieI
r-. iUY.II:*I pldos, spNnold or-,,,..,,..
T4b V«y 111..- d-
teal Tumorlnllld•111Jallhololk!oot\ll: Oltltlllapu,
....,tal"""""_, •-rAtryn<."'
......
du111, br'*'• mlclcllo<:tantol""""'
ra ~tlymJi!MillldMOUin<tben......
NO No "'llfonal ttmpll noclo mo-
Nl - l i s In a olnaloiPIIII!tonollymph ncdo. 3<m ........ In .,...,.d__,
N2a - l i s In a olnaloiPIIII!tonollymph ncdo. >3<m but not >6<m In_,.,.
dmenololl
N2b , _ . , h lnll-.,1& '"*"'"lymph 110dao.110no >6 em 1'1 .,..1ootdmenoion
N21! - + l n b _ . r _ l tlfnpllnodor, nOM >£em lnfl'-
dmonslon
N3 -•lnllymph...,.>6c:mln,-.t<l""""""
. _......... 011
MX Dll1antm""aeio<Ohh<tbee......ed
MO No dlratl!ilrt I'M'M"+
Ml Ollidl!lrrt meteete•
'I'M 3010 ~Joint CommlltM.., CMar WCCl hllllll
Gulolell- fof TU- d llle N.... Cew11J Mil ' - - 1 st111m
(CII..,.,.O
NO MO
NO MO
NO MO
NO MO
Nl MO
Nl MO
Nl MO
NO MO
Nl MO
N2 MO
N2 MO
N2 MO
N2 MO
AtoyN MO
N3 MO
AtoyN Ml
eo...,.,.
'"""' £qpl SB,I!ynl 00, CC, .. al, a:I!I.IIJCC Cilncof . $ ¥ Y - 7th ... -
NV,Sptp: 2010.1M!- ,....,-.
Valle,
'Rllic1l1ar WO.-.ia, .., """"""' 'lfilb l1lllll nw:l<Gli, but ISO o;typia end
mfnlmol mitotic act!.!ty, By bnmu.oob!Jiodlcmlrtty, the liti.'OmOl cd1a
arc poolti~ iw Wmai~D ....t -~~for IIIDOOdt. nmodo UIID. Tbc
c...tcthelW ccn. ut ~ for CD34. 01bc:nNe, the - are
ruptm: la! S-100, Aemin, &D.C! cytt>ktrlllin.
Altbc<lah the t:umJ>ri& hMip, ,..,..._eroc~aa ofdle..tjaa:mhcoe
i& - """""""' lhe <rinu.l1ly eo:c:l..m: ..........,.,. iD ..tolna:o:t
boya, wid! BfOWth arou...t puhe.tt,;, i& """""""' wid! tiLe f&et dill
~l'C<q>lo;IICIUlbod........uttcd!A- .,._
IL lllrali....!IC)WO"(JI·••_,...,... _,.., • .,..,hlllji.,O·
pe~I!:Jill•·ll .. t~~•or). Thit it a -ll.\1 a•~mon tllm<Xr of
~ .,..,;ct·typ< cdls. All .,.. can be aH..ctcd, lntt pouia>ta
iD their .....,a, d.....te p:n:dommate. lh!ilall:nl n.ual a.W:r imd.,.
- .. - .............. ....! die"""'"'" i& 111-'r polypo;d, bcdr·""'-
10 &ra1'1'ialr."""' toft. Mictota>pkd!y, the tumot ........ of .IIUIIIel'
..,. VU~ably o!tcd ........S.r clt•nnclo dill dua!colly b.t.~ • ......,.
:f!J,.noc. '!'be~ terooa ......ut. of • p<olifctacioa of
0Hi1 1'-"'t"'rt ..u. with EOW>d to 01'0id IIJI.clo:i ....d mc<~o:ot ~
of ....;..ophili<: qtaplum. Th.e III.IDar ...US pow iD short 6uMrs,
,.,..;mu ..nh • I<Dri.A>zm paltml. lhere i& ,;..;nul nu.clur pleo·
"""l'hilm, liule mitcnH; doily, &D.C! D.O """"""'"" (o.f"llllo ):IJI). a,.
'-ozaoblotochan!...,. the cello ate IIU'O:!I&Ir ....t d!ffollcly pocitl••
lot *"'>>Cb mutdc end m~ actiof, &D.C! fr>t 'rir"'!•ti" Alld
~or Xllla. The QJ.IDOt cd1a 1..:1< terOD& cli8\a CD34 mining. Tbe
ptCS!losi&is--'lrnt, wilh cmly&modat _ , ,......rut >90%
rnni..J •t s,.....
dtl:r ""''P"Y·
IlL AMol1r lflllldOII)'_,o.,, Grco.Jr, the 1codco b.t.o a tlcohyto lilm.
lllll1II=&Y _.,..,.,.. Mlcrotcoplcal.ly, die IWDOt boa & "bl"" <dl"
·~ b<:iag ~ of ..,.u. 10 ~ ccn. ..nh
... I SECTION I. HEAD AHD NECK
(II)
Rap>nol t,mp/1 ncdB$<:artno\ bo .....""'
No ,...,.llpnJ:tl , _ '"lllllllbo••
Unllii.1Bral rnot""'ooillft conlcol t,mph IIOd.Col, 6 ctn Ofiooo, abo\1&111&
Olllll't<llnleulllr'-, and/or unlotalol t1t bloWal, llll!Oih~IIOOI t,mph
l'll:ldi!IIJ 6 an t:11ea&..
N2 BII!Wal ,......_In ..,_lljmph r'olldo(l), 6""' or ..... • - ttl&
suJ>'liCirolcullr ,_,
....
N3 Mlllllltatlt In aljmp/1 noclots) >fi <m a•lllor to tile ""P"•-~r _ .
N3o GA>IIIor Ilion 6<m
!Ub llr.1onolon 1o tho 01/lftCIIMc:UIIIr fooloo'
, n•oo
M)(
MO
DIIUnt- cannd bo
No<IIUnlm-la
••••sod
..........
Ml
fllq&O
Dlllbo nt ndatt.osls
..., 1.__!11
r« R...nol t,mph min QU1not bo .,..,...,
NO tlo .....,, tl!nllil node~
Nl Re!ilonol t,mph nodorr>""'D&~• p.-rt
IIGrtt..'sl•lfl (1111)
MO tlodllllnl rnolaslnls
Ml llillllnt m - I t
hilt .........
1ho-ll.lliot1oliCI: .... II
Slqa Ill TS NO MO
Slqo IVA T4o NO MO
Slqo IVA T3 Nl MO
5lqo IVA T4o Nl MO
SQIIVII T4b AltJN MO
SQIIVC AnyT AltJN Ml
50 SECTION 1: HEAD AND NECK
I. NORMAL ANATOMY. Of all the bones of the skeleton, the jaws are uniquely distin-
guished by harboring the odontogenic apparatus of the deciduous and permanent
dentitions. The teeth germs are composed of three main components: the enamel
organ, the dental papilla, and the tooth follicle. The enamel organ is composed of
ectodermally derived epithelial cells, the ameloblasts, and is responsible for enamel
formation. The dental papilla is of ectomesenchymal origin and produces the den-
tine. The tooth follicle is also ectomesenchymal; it surrounds the developing tooth
and provides the supporting structures of the formed teeth, the periodontium. The
odontogenic tissues may also be a source of a bewildering array of odontogenic
cysts and tumors. Nonodontogenic cysts and tumors of the jaws will also be dis-
cussed in this chapter.
II. ODONTOGENIC AND NONODONTOGENIC CYSTS. Odontogenic cysts of the mandible
and the maxilla are relatively common and can present over a large age range.
Accurate diagnosis is simplified by location, radiographic correlation, and micro-
scopic examination (e-Fig. 4.1)."' Odontogenic tumors by contrast are uncommon.
They may be epithelial, mesenchymal, or mixed; may be noncalcifying; or they may
contain hard structures that mimic enamel, dentine, cementum, or bone. Although
malignant odontogenic tumors are extremely rare, odontogenic carcinoma, sar-
coma, and carcinosarcoma do occur.
A. Odontogenic cysts. With the exception of a few cysts that may develop along
embryonic lines of fusion (known as "nonodontogenic cysts"), most jaw cysts
are lined with epithelium that is derived from the odontogenic epithelium. Odon-
togenic cysts are classified as either developmental or inflammatory. Various
types of odontogenic cysts are listed in Table 4.1.
1. Developmental odontogenic cysts
a. Dentigerous cyst (follicular cyst) is a unilocular cyst that forms in associa-
tion with the crown of an impacted tooth and is usually associated with
a molar or canine. Patients usually present with an asymptomatic, well-
defined, expansive, radiolucent lesion. Microscopic examination shows
a thin layer of cuboidal to slightly flattened epithelial cells. Focal kera-
tinization, mucous cells, inflammation, and dystrophic calcification are
possible. Enucleation and excision of the associated tooth are the treat-
ment of choice. Eruption cyst is a subclass of dentigerous cysts associated
with the erupting primary or permanent tooth.
b. Keratocystic odontogenic tumor (odontogenic keratocyst) usually presents
as an asymptomatic unilocular cyst in the mandible or the maxilla. While
more frequent in the second to fourth decades of life, they may be seen at
any age. Their incidence is higher in Caucasians, especially in and men, and
roughly 5% of patients with odontogenic keratocysts have nevoid basal
cell carcinoma (Gorlin) syndrome. Gross examination typically yields a
cyst containing keratinous debris. Microscopic examination shows pal-
isading basal cells covered by a few layers of squamous cells under a
"'All e-figures are available online via the Solution Site Image Bank.
51
12 I SECTION I. HEAD AHD NECK
-· ,.,
'' 1."! I m I Oclonltltenlc c,.b
llonllpwo eyst oncl ONplion eyst
l'iortdo<:ywllc -.topnll:lllrna' C"'*"'•'*' ~
lnnll DOtlod0111al <YOt
~~I eyot oltlloadut
<lo""J'n,qotle cdM-Ie1>Jmor i'J:old¥11! ador'otaiiOnle<!lll. Gialfn <!lltl
solid areas (e-Fig. 4.5A and B). Hyaline bodies, ghost cells, and
ciliated cuboidal eosinophilic cells may be present. Care should be
taken to differentiate between a glandular odontogenic cyst and a
central mucoepidermoid carcinoma. Surgical excision is the treatment
of choice.
2. Inflammatory odontogenic cysts. Radicular (periapical) cysts are associated
with a nonviable carious teeth. The usual location is the apical third of the
tooth root, with occasional cases involving the lateral root surface. The cyst
is more common in the mandible. The typical age of patients is the third to
sixth decades of life. The most common presentation is pain and swelling, but
presentation as an incidental finding on routine radiographic examination is
not unusual. Microscopic examination reveals an inflamed, nonkeratinizing,
stratified epithelium. Cholesterol crystals, foamy macrophages, dystrophic
calcifications, and intraepithelial hyaline bodies (Rushton bodies) may be
identified (e-Fig. 4.6A and B). The residual cyst is a variant of radicular cyst
that is seen at the site of an extracted tooth.
B. Nonodontogenic cysts. This class of lesions includes a group of epithelium-lined
cysts as well as non-epithelium-lined bone cysts. The epithelial-lined cysts are
believed to arise from epithelial remnants entrapped along embryonic lines of
fusion and are referred to as fissural cysts (e-Fig. 4.7).
1. Epithelial-lined nonodontogenic cysts {fissural cysts)
a. Nasopalatine duct cyst {incisive canal cyst) is the most common of the
fissural cysts. It is believed to arise from remnants of the nasopalatine
duct. The cyst can develop almost at any age, but it is most common
in the fourth to sixth decades of life. Most studies show a slight male
predilection. The most common presenting symptoms include swelling of
the anterior palate, drainage, and pain.
Radiographs usually demonstrate a well-circumscribed radiolucency,
in or near the midline of the anterior maxilla, between and apical to
the central incisor teeth. The lesion most often is round oval or pear-
shaped. Microscopically, the epithelial lining of the cyst may be strati-
fied squamous, pseudostratified columnat; simple columnar, or cuboidal.
Commonly, more than one epithelial type is present. Because the cyst
arises within the incisive canal, moderate-sized nerves and small muscular
arteries and veins are usually found in the cyst wall (e-Fig. 4.8). Surgical
enucleation is the treatment of choice.
b. Globulomaxillary cyst is believed to develop from epithelium entrapped
during fusion of the globular portion of the medial nasal process with the
maxillary process, although its origin continues to be a subject of debate.
The cyst classically develops between the lateral incisor and cuspid
teeth, although occasionally it has been reported between the central
and lateral incisors. Radiographically, the cyst presents as well-circum-
scribed unilocular radiolucency between and apical to the teeth. The radi-
olucency is often pear-shaped. As the cyst expands, tilting of the adjacent
teeth may occur. Microscopically, many of the cysts are lined with strati-
fied squamous epithelium. Occasionally, howevet; the lining epithelium is
of pseudostratified columnar ciliated type. Enucleation is the treatment of
choice.
c. Median palatal cysts (median palatine cysts) are rare fissural cysts. They
are believed to develop from epithelium entrapped along the embryonic
line of fusion of the lateral palatal shelves of the maxilla. The cyst may be
difficult to distinguish from the nasopalatine duct cysts, and some cases
may actually represent a posteriorly placed nasopalatine duct cyst.
Clinically, the cyst presents as a firm or fluctuant swelling in the midline
of the hard palate, posterior to the incisive papilla. It is more frequent in
54 I SECTION 1: HEAD AND NECK
deposition (e-Fig. 4.28). Two types of mononuclear stromal cells are iden-
tified: spindle-shaped fibroblastic cells and polygonal macrophage-like cells.
Areas of ossification and hemosiderin granules may be present.
A rare aggressive variant characterized by pain, rapid growth, and corti-
cal perforation with a marked tendency for recurrence may be an example of
"true" giant cell tumor of bone. The lesion may show increased mitotic activ-
ity and evenly distributed larger giant cells that have an increased number of
nuclei.
CGCG of the jaws is usually treated by curettage. Recurrent lesions often
respond to further conservative surgery. A number of alternative nonsurgical
approaches have been used in recent years, including intralesional corticos-
teroids injections, subcutaneous calcitonin injections, and interferon alpha
therapy.
2. Brown tumor of hyperparathyroidism is an osseous lesion that develops in bones
affected by primary or secondary hyperparathyroidism. It is currently less
frequently encountered since the diagnosis of hyperparathyroidism is now
often made on the basis of elevated serum calcium levels in asymptomatic
adults.
The lesions may be solitary or multifocal, and the mandible is a common
site of involvement. Radiographically, brown tumors are well-defined lytic
lesions that are microscopically identical to CGCG. Treatment is aimed at
correction of the hyperparathyroid state; complete resolution usually occurs
within 6 months after removal of a parathyroid adenoma.
3. Cherubism is a rare dominant genetic disease with complete male penetrance
and 50% to 70% penetrance in women. It typically presents as painless
bilateral symmetric jaw expansion in children aged 1 to 5 years, which
slowly increases in size until puberty; at puberty, the lesion undergoes vari-
able regression. The lesions may be unilocular or multilocular, with a "soap
bubble" appearance on radiographic examination.
Microscopically, the lesions are essentially similar to CGCG. However,
the giant cells in cherubism tend to be less numerous and placed in a less cellu-
lar stroma. The pathologic process in cherubism is self-limited and treatment
is dictated by cosmetic and functional needs. Curettage and contouring of
bone are the treatments of choice.
SUGGESTED READINGS
Delair D, Bejarano P, Peleg M, et al. Ameloblastic carcinosarcoma of the mandible arising in
ameloblastic fibroma: a case report and review of literature. Oral Surg Oral Med Oral Pathol
Oral Radio/ Endod. 2007;103:516-520.
El-Mofty SK. Bone lesions. In: Gnepp DR, ed. Diagnostic Surgical Pathology of the Head and
Neck. 2nd ed. Philadelphia: WB Saunders Co; 2009:729-784.
El-Mofty SK. Cemento-ossifying fibroma and benign cementoblastoma. In El-Mofty SK Guest
editor. Semin Diag Pathol. 1999;16:302-307.
El-Mofty SK. Psammomatoid and trabecular juvenile ossifying fibroma of the craniofacial skeleton:
two distinct clinicopathologic entities. Oral Surg Oral Med Oral Pathol Oral Radio/ Endod.
2002;93:269-304.
El-Mofty SK, Kyriakos M. Soft tissue and bone lesions. In: Gnepp DR, ed. Diagnostic Surgical
Pathology of the Head and Neck. Philadelphia: WB Saunders Co; 2001:505-604.
Nevill BW, Damm DD, Allen CM, et al, eds. Odontogenic cysts and tumors. Oral and Maxillofacial
Pathology. 3rd ed. Philadelphia: WB Saunders Co; 2009:589-642.
Philipsen HP, Reichart PA, Slootweg PJ, et al. Odontogenic tumors. In: Barnes L, Eveson ]W,
Reichart P, et al, eds. Pathology and Genetics Head and Neck Tumors. Lyon: IARC Press;
2005.
Slootweg PJ, El-Mofty SK. Ossifying fibroma. In: Barnes L, Eveson ]W, Reichart P, et al, eds.
Pathology and Genetics Head and Neck Tumors. Lyon: IARC Press; 2005.
The Eye
George J. Harocopos
*All e-figures are available online via the Solution Site Image Bank.
63
•• I SECTION I. HEAD AHD NECK
T4c Tumorl-odJI<:*rtf>II'INIII'*'-
T4d Turnorl.-linln
...,....,.,.,. ..... Ill)
ra Rogjonol ~r*l nodeo.c:annot ............
NO No • • I I f.lmph node mll11l"'b
Nl R<P>nol ~r*i nodll 11'111-
._lllllllaill 011
MO No dlalant ..........U.
Ml Dlllaniii'III-
JXz'>MI• ..........IIIf•llllllll
No ...... ~""*"' p~ '""'"'mended.
F.,,. ~ SB, e,rd DR,
~. sro~ ....., Com,....
OC:, .. 01,- AJCC Ciw8r511,111y-..: 7111 ed. N'"' - ·
21)10. tJood- pelll'lllolcr\.
hfllllact1'111
Cl- I -•(lllll)
''k
1lC
•F>·•., m Pt1MOJYtum« Cl/lftct llo &Gomod
TO No <!Old..,.. of srt>ti!IYl>lmot
TQ>) M"""""• coni!nod to lila cm)mciMitplhoa.m
n Mdlnllllmo~nanaofllla I>Ubllcm)mciM:
n. """'""""' oqlllll to 1 quodnort
Tib MOfotlllnl IMA~quochnts
TI< M«ohn21MA:$$~
Tid G-rthln SqUS<hnta
T2 Mdlnllll mo~nana of lila nonl>ulblr (palpol>lll~ r..-1, cnncullr)
<OI'junclvl:
II I SECTION I. HEAD AHD NECK
''lhiJ.fll lllllplntllaU...tU.~(ti:II&•IO
T2a No CMVI"tCUiir. S1 qLiiiCII'Irrt
T2b No can:....,r, >1 ~nt
T2<: My CIIUIICUI:r, lOll! ,;1 qUid rani
T2d MyCIIUheUior, lOll! >l qua<f:OIIt
T3 MJIMI!pntoon)ln<IMI,...,n...., ••tllllm I n - :
'l3o Gbbe
Tab eyolld
T3c Orbl
Tad st!UI
T~ 1\unor lwildoo111e Clltlll'al n~t~Wl ,yam
...,........... -Oil
~ R.,..lbmplln-comolbea-
NOe (blopliod) No "''l)onnO tjmp/1 noclo mot-.
~ podonnod
NOb (nol blopllod)
Nl
. . . . . . bill(ll)
R.....l bmpll node-·
No "''l)onnO tjmp/1 noclo mot-.
~nat potfonnod
MO Nodls1anlma-
M1 --·
MIIIIP-(p'IWl
,....,._ Qln
pll( Pnnary tumor a~ni'ICrt bo ••••••
PTO No <Mience <If l>ltno:Ytum«
~TOO ""~"""" of!M ...,Jill- ..mlrood" 1M optiloi~M
~Tl· Me~"""" of!M bulbu ...,jun- not ....,.,INn 0.5 mM l n l l l -
wlll I n -of tho subiUnlla pn>jllla
pTlb Ml~nrm.t of tho bulbu ...,junctM mora than 0.5 mm bill nat mora
Ilion 1.6 mm In 11\ll:linoso will I - aflllo '"botanlla pn>Pfla
Me~"'""' ofllle bulbi! CUI] undilla •l.S 111m ln111~ 101111 l -
of111o """'*'"tlopl<lft,
""~"""" of!M polpobtol, fomblal, 01 """' ....... ...,.....,.. Mt MOll
Ilion OS MM In t h - w l l l l l - ofiM :Wbi!IMtlo ptOp!lo
Ml~nrm.t of tho polpobnll, fomlcolll, "'"'"'""":.....,)In- mora
Ilion 0.6 mm but not >1.5 mm In I l l - - - of tho
..blltanlllprop<la
Me~"""" ofllle polpeblol. fomloool. 01 ..n:"""lor Cl:ti)Jn-
>1S MM In t h - willll-n oftlk,.IJiiotantla ptOpola
~1'$ ""~"""" ln\J&dlngtltu;t•, ..,..:kl, - M a l II)WM>, ''"""'"· ... -
pT4 Ml~nrm.tlnll!ldlnath•....V•I""""""'I)IOIIoln
F.,,. ~ SB, e,rd DR, Clom;lb><l OC:, .. 01,- AJCC Ciw8r511,111y-..: 7111 ed. N'"' - ·
~. srot-: 21)10. tJood-" ' - ·
Chapter 5 • The Eye I 67
...,...,.,.,.-(II)
T4d
KX
••bl<t..,lllapt'eM!
Ill. VASCULAR, INFLAMMATORY, AND INFECTIOUS DISEASES AND TRAUMA. Eyes are often
removed when they become "blind and painful." The reasons for an eye becoming
blind and painful are numerous, but often the final common pathway involves the
development of secondary angle-closure glaucoma and/or chronic retinal detach-
ment, and optic nerve atrophy. The antecedent event can range from previous
accidental trauma or previous surgical trauma to specific disease entities such as
retinal vascular disease (including diabetes, or central retinal artery or vein occlu-
sion), intraocular infection (endophthalmitis), chronic inflammatory disease, or
primary retinal detachment. Surgical methods of removing an eye include enu-
cleation, in which the entire eye is removed intact, or evisceration, in which only
the intraocular contents (lens, retina, uvea) and possibly cornea are removed with
retention of the sclera in the eye socket to house the orbital implant. Exentera-
tion, in which the eye along with surrounding orbital soft tissues are removed, is
sometimes required to achieve complete excision of an eyelid/orbital tumor (see
later).
A. Secondary glaucoma. The key histopathologic finding in most cases of sec-
ondary glaucoma is an anterior chamber angle closed by peripheral anterior
synechiae. The term "closed angle" means that the anatomical angle normally
formed by the cornea and iris, and occupied by the trabecular meshwork (the
main outflow channel for aqueous humor), is occluded by the peripheral iris. If
this occlusion is chronic, then permanent adhesions form between the periph-
eral iris and the trabecular meshwork, that is, peripheral anterior synechiae
(e-Fig. 5.23). These adhesions may be induced by chronic inflammation, such
as in various forms of chronic uveitis, or by the abnormal proliferation of cap-
illaries (neovascular glaucoma), as seen in proliferative diabetic retinopathy or
retinal vascular occlusion (see later). Occasionally, in contrast to the closed-
angle appearance, post-contusion angle recession may be seen in cases of blunt
trauma. The retina in glaucoma exhibits atrophy of the nerve fiber layer and
loss of ganglion cells; the optic nerve shows cupping (e-Fig. 5.24) and atrophy.
B. Chronic retinal detachment. Retinal detachment may be idiopathic, trauma
induced, related to inflammation or infection, or secondary to proliferative
retinopathy of various etiologies (diabetic, post-vascular occlusion, and so on).
Often an attempt is made to repair the detachment surgically, but severe cases
in which surgery fails or the detachment repeatedly recurs may ultimately be
treated by enucleation (e-Fig. 5.25).
C. Retinal vascular diseases. Eyes with severe retinal vascular disease (most com-
monly diabetic retinopathy, or central retinal arterial or venous occlusion)
often exhibit both secondary angle closure due to neovascularization of the
iris (e-Fig. 5.26) and angle (neovascular glaucoma), as well as chronic trac-
tional retinal detachment due to proliferative retinopathy. In cases of diabetic
retinopathy, microscopic examination of the retina reveals the key features of
diabetes: lipoproteinaceous exudates, intraretinal hemorrhages, and neovas-
cularization of the inner surface of the retina (proliferative diabetic retinopa-
thy) (e-Fig. 5.27), often with tractional retinal detachment and hemorrhage
into the vitreous. In cases of old central retinal vein or artery occlusion, the
retina exhibits atrophy and cystoid degeneration of the inner two-thirds of the
retina, lipoproteinaceous exudates, and in the case of venous occlusion, often
a persistence of hemorrhage. There may also be a neovascular epiretinal mem-
brane with tractional retinal detachment and vitreous hemorrhage. As with
neovascular glaucoma of any etiology, eyes with severe diabetic retinopathy or
central retinal vascular occlusion also exhibit atrophy and possibly cupping of
the optic nerve.
D. Intraocular inflammation and infection. Endophthalmitis most commonly occurs
via an exogenous source (e.g., following a surgical procedure, trauma, or per-
forated corneal ulcer), or may arise from an endogenous source (e.g., secondary
Chapter 5 • The Eye I 71
Figure 5.1 The globe is identified as to right eye or left eye using anatomic landmarks (upper
panel}. Two cuts (traditionally refell'ed to as the p.o. sections) are then made in a transverse plane,
passing in relation to the pupil and optic nerve as shown (lower panel}. The superior cap (also
known as the superior calotre) and inferior cap are usually not processed unless abnormalities are
seen or the eye has an intraocular tumor.
>15.0 4 4 4
E' 12.1-15.0 3 3 I 4 4
-
E
9.1-12.0
6.1-9.0 2
3
2
3
2
3
2 I 3
3 3
3
4
4
3.1-6.0 1 1 1 I 2 2 I 3 4
:S;3.0 1 1 1 1 I 2 2 4
:S;3.0 3.1-6.0 6.1-9.0 9.1-12.0 12.1-15.0 15.1-18.0 >18.0
Largest basal diameter (mm)
Figure 5.2 Size categories for ciliary body and choroidal melanoma based on thickness and
diameter.
epithelioid, with the latter having a worse prognosis (e-Figs. 5.36 and 5.37).
Other histopathologic features that predict prognosis are size (e-Fig. 5.38),
extrascleral extension (e-Fig. 5.38), and location (those arising from the iris
have a better prognosis, whereas those involving the ciliary body [e-Fig. 5.39]
or peripapillary region have a poorer prognosis). Other factors associated with
a worse prognosis include necrosis and mitotic figures. The staging scheme for
melanoma of the uvea is described in Table 5.4 and Figure 5.2.
C. Retinoblastoma is the most common ocular neoplasm in children. The usual
age for clinical presentation in nonfamilial cases is 1 year, whereas familial
cases are generally screened within the first few weeks of life and therefore
present earlier. The usual clinical appearance is leukocoria (white pupil). In
nonfamilial cases, the tumor is generally large and unilateral and accordingly
treated with enucleation. In familial cases, tumors are generally seen bilaterally.
The tumor may be much larger in one eye than the other, in which case the
eye with the large tumor is enucleated, whereas the eye with smaller tumor(s)
may be salvaged via laser or cryotherapy applied to the tumor foci, possibly
with antecedent adjuvant systemic chemotherapy. A few centers utilize intra-
arterial chemotherapy (delivered to the ophthalmic artery) to treat unilateral or
bilateral tumors, thereby potentially salvaging eyes that would otherwise need
to be enucleated. Some centers utilize periocular injection of chemotherapy.
Histopathologically, retinoblastoma arises from the retina (e-Fig. 5.40) and
is composed of small blue cells intermingled with anastomosing pools of pink
necrosis giving rise to the traditional description of "islands of blue tumor
in a sea of pink necrosis" (e-Fig. 5.41). Scattered flecks of purple calcium
are often seen. Cytologically, the tumor is composed of cells with hyperchro-
matic, oval-shaped nuclei with scant cytoplasm; the nuclei are densely packed
together and thus show nuclear molding. There are many apoptotic bodies and
numerous mitotic figures. More differentiated retinoblastomas often exhibit
Flexner-Wintersteiner rosettes (e-Fig. 5.42). Extension into the optic nerve,
and especially presence of tumor at the surgical margin of the nerve, predicts
a poor prognosis (e-Fig. 5.43). The other factor predictive of poor progno-
sis is massive invasion of the choroid. If optic nerve invasion past the lamina
cribrosa or massive choroidal invasion is seen on histology, then the patient is
treated with adjuvant systemic chemotherapy. (Some practitioners have a lower
threshold for adjuvant chemotherapy than others; the optimal algorithm for
post-enucleation chemotherapy has not been established). The staging scheme
for retinoblastoma is given in Table 5.5.
V. DISEASES OF THE LACRIMAL GLAND AND LACRIMAL SAC
A. Lacrimal gland lesions are similar to lesions of the parotid gland (see Chap. 6).
Infiltrative lesions include dacryoadenitis, sarcoidosis, lymphoid hyperplasia,
and lymphoma. Intrinsic lesions of the lacrimal gland include pleomorphic
,.. I SECTION I. HEAD AHD NECK
-·-
T1 1\JmoroiZo.....,.f 1''
Tla Tumor slm....,IY 1-ut ciii!Y bo<ly 1-Mnlor-lu
-·-
T~ 1\Jmor oLio ..,.,IY 4',
T4a 1\Jmor 11m c:ft!jlpfY" -utcllary body I"""'"""'"' or -...c.Jlu
T4d
TAo
- · § mm In dla.-
TI.mor sliD ari<FY 4-
n.mortbar'-Y4 -uldlilly l l o d y - l l u t -- · ·
dlluy 11oc1t
-..ion :$5 mm In die_.
.,_on!
and - u l l r
-"·'. .~~-·"'''(II)
MO NodiJI&ntmot_,
Ml Dlot.ont-
Mlo l.a!Bootdlolnder of111o ioiJDot 111-olo ;S$<tn
Mlb l.a!Bootdlolnder of111o ioiJDot 111-olo 3.1-7.9 em
Mle l..tlgeltd- ofllta illlJIOd m-.olt ::;Sem
~IIIIIIU!IIIal•.,.. Tho hiltoprll\'*'llc\YP8""" "'foi-
Spllldlo<oil melon""" (>9011> o~ <dlol
Mllod ..nmolollorre 1>1011> t~~>llhei:id ,.,...lid >lOlC> Olindlo..,lhl
Ejlltl- ool '""'~"""" (>1100:. eptilollotl ..!lo)
ftfllll IUIIII&fel!llft(IJ
GX Gralla Qllnot bo • -
01 Si>Nio ..u,......,.
G2 _.,.., rnollnomo
SQII Tlo NO MO
SQIIIA TliHI NO MO
T2a NO MO
Slqa liB '121> NO MO
l1!o NO MO
5lqo Iilii T2o>-d NO MO
T3b-<: NO MO
To4o NO MO
Slqa I!IB T$d NO MO
T<IIH: NO MO
Slqo I!IC T4<H NO MO
5lqo IV AIVT Nl MO
AIVT AllyN Mla-c:
'So& F\IJ>"' S2.
''""' ~ SB,I!ynl 00, C>m"""'
CC, .. 01, "''· AJCC CilnoorSI¥tlf- 7th ... -
••• Sptwer: 2010. ~- . . . . , - .
Yatk,
'JI I SECTION I. HEAD AHD NECK
-ltiillf'llll
CUIII*I ciHII'II:CII (Cl'lliD
"'-'11-m
1X Primary turntr' ca1not bt MSMiild
TO t w - of """'"" tumo<
T1
Tla rw tumo< 1'1 ekhOf """"'.a
1\.moro 110 more Ilion 2.13 ctelio "'lume ctCii&Ol'&..tli 110 ¥111l>OII. .rwbrelhll....tnt
mm In lo!Bootd..,_ orlo """'*>d ciooer1!1011 1.5 rrvn
tt> ~~~·-- ,.,., ... -
Tlb Atlootonetumllfl!.:..llmmln ~-dmonoloi\Of-elowlhoni.S mmW>11i&
op!Sc...,.. t1t r - . No -lc!*ch...,lor .,bnilfnll fll.6d bo)lond 5 mm ticm tho
bllloctll>otum"'
Tl• Atlootonotum0fb>3mmln ~'l"'ldmonoloi\Oflocolodclolofllllnl.S mm W> 11>o
op!Sc nOf\illor*"">o, Mil RlllhOidelachmentor wblllllhOI!\11:1 ~d 5 mm frOfn
tho buo ct 111& tUI'ICII'
T2 1\.mora no mor& tllan 21!111i&"""mool'11ie ""''""'- · • tlflu.._l...,..ng. Con
"""'relNI-ment'
T2o Focol-.andlotsutnllniiiMdlnJolnno-taofbirnorcolllllptOSOnl, bi.t
no llilp<llmpsor ">n-Il" of tumor ....
T2b ,.,.,.'<l!rooua ....tnuhdlor 011bnlllnol alll(fns lo ,.-.dell ned .. dl'lll..
climDOI"' -bol>' oltumllf.,..
n
Tao
T3b
s-.JntratJaJ~rdl-.e'
TLmOI' flb """"Ilion 2ll ct the "JJ
Ono t1t moMcomplalmure p--', ""'lch ....,.lndu<lo lilmllf.. -od
_,.ulor or on...,..loouro""""""'a. tu.-- Inil> 11>o •nbitlor ""f'11C1CI,
~"'*""· ¥llnlouo hemonlloa or<ftlllelcel~llll4
T4 ~lu dhe&d-lt/ l'nollllnt-.doo:
T4e lnYUion ctoplle...,.
T4b lnYUion lnW> llle trtit
T4c lnlrlc~Wn,.l-• nol pul diillm
T4d lrrlnl<:ranlll- put dilalm
...,....,.,.,. ..... Ill)
ra RoP>no• !lm1lil . -... nno~o.•nNUd
NO tw np.nlltympll nodo - e n t
Nl """""'' tlm1Jil nodi ln,.,_.,.nt (p""'l.flcullr, coM:al, llllrmndllullr)
N2 Dllt..rtlympllnodo-
... ' liM
MO No mfltiMtata.
Ml S:,.Ot.nlc "'lllalloR
Mia Sftp> - to..., oll>otltion Ci'IS
Mlb Mua!plolnlomlotl!aolliOftlliln CNS
Mlc P!'ochlootno.11:: CNS!eolonW
Mid -hlootn<lle CNS - ( I J
Mle l.ept>m&nlnpool Uidi« C S f -
Nil IIIII< cluollo. . Q111111)
Pt'-Jtl- Qlll
~TX ~meryturr.:rolfU'I(rtbe:ttte'!*d
pTO tw - o f pm.IY Ill"""
pTl TLmO<..,.INdlo""'lOfltl noop!Sc """" Ofol•>~otlellnYUion
PT2 TLmO<wlth mlnmalo!*""""•-dlorotlellnYUion,
e~~ . • s. ne. Eyr& I ,,
pl2a Tumor*'porllc~llf lnlllldos optic n.,..- bill does nal8ldlnd pollun!na a1brou
0111.mo< ..tllllb lo<:ll<hO<Oidalln-
pl2b Tumor ..,porllc~llf lnlllldao optic " " " ' - bllldooo nal""*'nd pool lomlna a1brolll
4/ld\lmor Ollllltilo focol cilon*lol.,""""'
pl$ TUmorMI!h ..,11.,...
TUmorl_o_MNOpoot~m-.a<li~
~le """"•nd.l>rdloraklllln-:
pl$1
plab
l!.mor""'lllfl m.,.. -~~-.-
biJinotlb.IIIJtP>oi.-.l!noN
-.....,..
Pl4 TUmor lhlll>do>ollllc 110,..11> ~n horOlllltilo-lor eoc!e~Won flhov~re:
Pl4a TUmor lhlll>do>ollllc ,.,..11> ~· IIIIo butno-lar -•lde11111od
pl<lll Tumorl-ollllt,.,..lo_n hand-.a.~r- klontlled
''k
1lC
•F>·•., m Pltno!Y111n« comet be a......,(!
TO N o - d rointiJ)Iturmr
1lo c.,_•., ....
n TumorSmm orlolslnt7NMIIdl"'"""'"
~ln¥11dlna thalli,... 1>1*« Ofl'dld maQIIn
1\imortnoR>111111 5 mm, bitt 1101 rnore than 10 rnrn. In l)llll1ootdlnenolon
Or, enrtumor111ot l'lw.deohllrool Dlota or 01111\d moQIIn
T'2b Tumor mor& thtn 10 mm, but notm:~r& than 10mm, In "16•--t dlnw\alon
Or,'"""'-1\JI-th- .oyeld
Tumor mo/'6 th111120 mm "p..a1ll!d dimension
Or, any turno< t11at l'lw.deolll)eoont .....,, 0< Oflllliololruduros
Ant T - """""'""tumorl n -
'13b Corn~IAI tumor r-~D~~Uhoenudeo11ort, - * ' · o r bono
~!!edM
TA Tumor it nt:t ~t*: du• tc. 4ldiiW\sJM lrrMbn of O«<:t!!r. Oftl;ltl!ll,
CllnloflciiiiiM:tura, or tnln
...,,..,.,...,...on
r«
eNO
Relllonol trm~h IIOdoo <011110! be"'"""""
No..,.,., tlmril node,...,..,..., bood upon dll'lbll""'bltlon or
lml&lni
pNO
No"""""' tlmril noclo "'"""'"' bood upon"'"'"",_ blot>l't
Nl R""'nol "'"'ph !Oldo m -
IIGrt••s•••t•
MO
(1111)
Nodllmrrt metutuk
Ml OIIUnl m.WW!a
~~~.,,...,....._.,...,.
$QIO 11o NO MO
Slqa lA Tl NO 1.10
Slqa 18 T2a NO 1.10
Slqo tC T211 NO 1.10
5lqo II 1"311 NO 1.10
5lqo IIIII. 1"31> NO 1.10
$QII!I~ A"!T Nl MO
Slqa I!IC T4 Any N 1.10
Slqa IV AAtT AllyN 1.11
r"'"'' £die sa. e..:t DR. eom.,.., <:e." •'· ""·A./IX C#oeu/llti(Jii;f--7111 011. Hew 'll>ri<,
NY:Sp'""· 2010. t.Joed...., pernilolon.
SUGGESTED READINGS
Eagle RC. Eye Pathology, an Atlas and Text. 2nd ed. Philadelphia, PA: Lippincott Williams &
Wilkins; 2011.
Font RL, Croxatto JO, Rao NA. In: AFIP Atlas of Tumor Pathology, Series 4. Vol5. Washington,
DC: ARP Press; 2006.
Smith ME, Kincaid MC, West CE. In: Krachmer JH, ed. The Requisites in Ophthalmology.
St. Louis, MO: Mosby; 2002; chapters 3-9,23.
Yanoff M, Sassani JW. Ocular Pathology. 6th ed. Philadelphia, PA: Elsevier/Mosby; 2009.
Salivary Glands
Joshua I. Warrick, Elise L. Krejci,
and James S. Lewis Jr.
I. NORMAL ANATOMY
A. Macroscopic/gross. The salivary glands are exocrine organs that secrete compo-
nents of saliva to both break down carbohydrates and lubricate the passage of
food. There are three major paired salivary glands: the parotid, the submandibu-
lar, and the sublingual. There are also numerous minor salivary glands located
in the submucosa of the entire upper aerodigestive tract (UADT), from the lips
and nasal cavity to the major bronchi.
The parotid glands are the largest major salivary glands and are located
between the ramus of the mandible and the mastoid process. Each gland is
composed of a superficial lobe and a smaller deep lobe, and the facial nerve is
intimately associated with both lobes. Each gland normally contains approx-
imately 20 intraglandular lymph nodes. The secretions of the parotid gland
empty via Stensen's duct into the buccal mucosa of the oral cavity near the
second maxillary molar.
The submandibular glands (also referred to as the submaxillary glands) are
located just medial to the body of the mandible, and are smaller than the parotid
glands. Their secretions drain through Wharton's duct into the floor of the
mouth.
The sublingual glands are the smallest of the major salivary glands and
are located in the floor of the mouth between the genioglossus muscle and the
mandible. Their secretions drain into the floor of the mouth directly via small
ducts or through the larger (Bartholin's) duct which drains into Wharton's duct.
B. Microscopic. The major salivary glands are enclosed by a connective tissue cap-
sule and divided into lobules composed of ducts and acini, whereas the minor
salivary glands are unencapsulated. The acini are composed of mucinous and/or
serous epithelial cells surrounded by a layer of myoepithelial cells which con-
tract to aid in the movement of glandular secretions. Myoepithelial cells are
inconspicuous in normal salivary gland sections.
A single acinus may be composed of a mixture of serous and mucous cells.
Serous cells are pyramidal in shape and contain basophilic periodic acid-Schiff
(PAS)-positive granules within their cytoplasm. Mucous cells are more rounded,
have basally oriented nuclei, and contain abundant clear mucin. The parotid
gland is almost exclusively serous, the sublingual gland almost exclusively muci-
nous, and the submandibular gland is mixed serous and mucinous (e-Figs. 6.1
to 6.3 ). * The acini secrete fluid into the intercalated ducts, which are lined by a
low simple cuboidal epithelium. Intercalated ducts are also the source of reserve
cells that can repopulate the acinar system. The intercalated ducts join to form
striated ducts, which merge to form interlobular ducts, which ultimately empty
into the large named ducts.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION
A. Biopsies of many salivary gland lesions are taken prior to surgery to characterize
the lesion and direct management.
1. Fine needle aspiration (FNA) is the procedure of choice for initial character-
ization of salivary gland lesions for many reasons (see cytology section that
follows).
*All e-figures are available online via the Solution Site Image Bank.
81
82 I SECTION 1: HEAD AND NECK
2. Core needle and incisional biopsies of the parotid gland can damage the facial
nerve branches so are rarely performed.
B. Resections. Some salivary gland masses are removed in their entirety without a
previous tissue diagnosis. Superficial parotidectomy remains the initial proce-
dure of choice for benign parotid gland tumors. Submandibular gland resections
are usually performed without taking any significant periglandular soft tissue,
although more tissue may be included with the specimen for submandibular or
parotid tumors that are known or suspected to be malignant. The rare sublingual
gland tumors necessitate a resection of the floor of mouth that is qualitatively
similar to those performed for mucosal-based squamous carcinomas of the same
area.
In general, salivary gland specimens are small enough to prosect the day
of receipt. The gland should be described, and its dimensions recorded. The
deep (covered by muscle and fascia) and superficial (covered by subcutaneous
fat) surfaces of the parotid gland can sometimes be discerned, and should be
differentially inked. The specimen should be serially sectioned, and any mass
or focal lesion described including dimensions, color, texture, and distance to
the margins. Four to five sections of the tumor, including representative areas of
the closest inked margins, should be taken. At least one section of the normal
surrounding gland should also be taken. For the parotid gland, the surrounding
gland and any periglandular soft tissue should be thoroughly searched for lymph
nodes, which should be separately submitted.
Ill. DIAGNOSTIC FEATURES OF COMMON DISEASES
A. Inflammation and infection. Sialadenitis, or inflammation of the salivary glands,
can be divided into bacterial causes, viral causes, and autoimmune disease.
1. Bacterial sialadenitis is rare and is generally a result of obstruction by stones
(sialolithiasis). The causative organism is usually Staphylococcus aureus. fol-
lowed by Streptococcus viridans and gram-negative rods. Surgical specimens
from bacterial sialadenitis are almost never seen. However, sialadenitis may
lead to an abscess that requires surgical drainage. Gross specimens may show
a purulent exudate or a relatively intact gland. Microscopic sections can show
abscesses, cystic degeneration, and/or bacterial colonies.
2. Viral sialadenitis is most commonly caused by mumps (paramyxovirus), but
can also be associated with Epstein-Barr, coxsackie, influenza A, and parain-
fluenza viruses. Surgical specimens are rarely encountered. Grossly infected
glands are boggy and edematous, and chronic inflammation may be seen
microscopically.
3. Autoimmune sialadenitis is relatively common, and usually presents as
Sjogren's syndrome, characterized by facial swelling, dry mouth, and dry eyes
(termed Sicca syndrome) with inflammatory arthritis and elevated autoan-
tibodies. On physical examination, bilateral, symmetric enlargement of the
salivary and lacrimal glands is seen. Diagnosis may be facilitated by a minor
salivary gland biopsy, typically taken from the inner lip.
Grossly, the major glands usually have a discrete, tan nodularity. Micro-
scopically, in early disease, there is a lymphoplasmacytic septal inflamma-
tory infiltrate with little to no abnormality of the parenchyma. The so-called
"focus score," a nodular collection of >50 lymphocytes, is considered diag-
nostic of autoimmune sialadenitis in the appropriate clinical context. In larger
glands or in more severe disease, there is typically an extensive lymphoid
infiltrate with germinal centers (e-Fig. 6.4). The acini are often atrophic,
and interstitial fibrosis may be present. In late disease, acini may be com-
pletely absent leaving only isolated residual ducts, termed "epimyoepithelial
islands," with an associated dense intraepitheliallymphocytosis. In Sjogren's
syndrome, the lymphocytic infiltrate is polyclonal, in contrast to the mon-
oclonal lymphoid proliferation seen in mucosa-associated lymphoid tissue
(MALT) lymphoma.
Chapter 6 • Salivary Glands I 83
wall has dense lymphoid tissue with germinal centers (e-Fig. 6.8). Excision
is curative, and the lesion does not recur.
3. Mucoceles are the most common non-neoplastic lesion of salivary gland tis-
sue and are defined as pooling of mucin in a cystic cavity. Two types are
recognized. In the retention type, the mucin is within a dilated duct. In the
extravasation type, the mucin accumulates in the soft tissue. The lower lip
is the most common site for mucoceles, followed by the tongue, the floor of
the mouth (where the lesion is termed a "ranula"), and the buccal mucosa.
The peak incidence of mucoceles is in the third decade. Grossly, a mucocele
presents as a cystic cavity in the connective tissue that is filled with glistening
fluid. Microscopically, the cyst wall may or may not have an epithelial lining,
depending on the type. Those of the retention type have a lining and usu-
ally no surrounding inflammation, whereas those of the extravasation type
consist of mucin with surrounding inflammatory cells and no lining. Some
late lesions consist only of a collection of foamy histiocytes containing mucin
(e-Fig. 6.9), which can be confirmed by PAS or mucicarmine stains. Surgical
excision is usually curative.
IV. NEOPLASMS. Neoplasms of the salivary gland can be roughly classified based on
the cell type(s) of the normal salivary gland toward which they differentiate: aci-
nar, myoepithelial, or ductal. However, most neoplasms have dual differentiation
because almost all show of those with ductal differentiation also have some myoep-
ithelial differentiation. Most benign neoplasms have a malignant counterpart. The
World Health Organization (WHO) classification of tumors of the salivary gland
is listed in Table 6.1.
A. Benign neoplasms
1. Pleomorphic adenoma (PA) is the most common neoplasm of the salivary
glands. Ninety percent of PAs occur in the parotid gland and PAs represent
60% of parotid gland neoplasms; the majority of the remaining cases occur
in the hard palate or submandibular gland. The tumor occurs over a wide age
range, but the peak incidence is in the fourth and fifth decades. The tumor
presents as a slow-growing, painless mass that usually is between 2 and 5 em
in diameter. On gross examination, these tumors appear well-circumscribed
but are not usually encapsulated. They are never encapsulated when occur-
ring in minor salivary glands. Sectioning reveals a rubbery, myxoid, tan-
white mass (e-Fig. 6.10). The microscopic appearance, as the name suggests,
is highly variable, but always shows an intimate admixture of epithelial and
mesenchymal elements. The epithelial component consists of ductal struc-
tures with an associated myoepithelial layer but also may contain collections
of myoepithelial cells that can range from spindled to clear, plasmacytoid,
or basaloid. The mesenchymal, or stromal, component is typically myxoid,
hyaline, or chondroid (e-Fig. 6.11). Although defined subcategories have no
clinical significance, pleomorphic adenomas have been divided into a myxoid
type (>80% mesenchymal-type tissue), cellular type (>80% epithelial-type
tissue), and mixed or classic type (generally an equal mix of components).
Treatment requires complete excision because PAs are likely to recur if
tumor is left behind or transected. Multinodular growth is uncommon in pri-
mary tumors but is frequent in recurrent disease, yielding a so-called "buck-
shot" pattern (e-Fig. 6.12). Therapy for recurrence is consequently more
difficult.
2. Warthin tumor (papillary cystadenoma lymphomatosum) is the second most
common benign salivary gland tumor and is found exclusively in the parotid
gland. It is the most common bilateral or multifocal salivary gland tumor,
usually presents in the sixth or seventh decade, and is associated with smok-
ing. Grossly, it presents as a soft, brown or yellow mass that is composed of
cysts that are classically filled with viscous brown fluid. Microscopically, the
CN~6· 5alftey81•11el:a I ••
,,(,I ]IJJH WHO Hlltolollc Clulllcatlofto!Tumotuf111e llltwuyGflnta
1111 .... _ .. 1111_ ..
Adnlc ... ar<:mme
M._tlarmold ..,..,..,.
AdOIIOid<'>'*<On:t>omo
~~*'--""'
Eplt\alel IYT)'Oflpthe!l!ll cud\tf'nll
Cloorcolardn<mt, nd
Buol Cllll """"""'-'
-·-"'>' dffed
~~~!rana
Se!Jo<eo11o t.'rn~l'lol!lll
C'.i*donOCII<IIIOitlO
~·«1- qo!Od""""n:h>""'
Mudnow: .tdlnoc:llrcttoma
Onoct:r1t:... - .
Sl""ty duct Qlldnoma
Ad...,..n:h>me, not-- "'"''f'fod
~~~~~•I<On:h>mo
a..mma a pleomorpllle edon<ma
~
Moe•* tzicploon... pltk:l<llnom•
Squunoua ... Cli'Cinana
Smoll Cllll<:~n:Nma
~CIIII<On:t>omo
l.)m~oloiCIII\:1"'""'
Slmbluome
..........,ti_
Pioomcopllle ooonome
a~~~~~·""'
..!Cllll odonorno
llfel!ll"'ll to.mor
~·
Clnlbllr-••
~·"""""""
L,.,.plledonoma
Se!Jo<eo~IO
Non. - a
Duouolpopl-
lnvooted duc:lal Pll>lbna
lni-IIIOI'Ibnl
- ...,.llfnm
C'.i*don<mt
1111--
Koma ....m•
""' .......... -
._..,_
Hoc!J,!I*1'•1Jmphoma
Dt!IIM ltu.. lkool t.'rnPhM>O
Bmnodol malfljnll !til& 8-GIIIIJmpltM~e
II I SECTION I. HEAD AHD NECK
mitotic activity is low. The epithelial cells express cytokeratin and S-1 00, but
are negative for calponin.
The differential diagnosis for canalicular adenoma includes pleomorphic
adenoma and basal cell adenoma. However, the pattern of growth of canalic-
ular adenoma is virtually always distinctive enough to make the diagnosis.
Simple local excision is performed with an attempt to obtain dear margins.
Recurrence is rare.
5. Myoepitheliomas are benign tumors composed of myoepithelial cells. These
tumors occur with approximately equal frequency in the parotid gland and
minor salivary glands, particularly the hard and the soft palate, and present
as slowly growing, painless masses in adults. Men and women are affected
equally.
Grossly, these neoplasms are well-circumscribed and encapsulated with a
tan to white cut surface. Histologically, they are composed almost exclusively
of sheets and cords of myoepithelial cells, though ductal cells may comprise
a small percentage ( <10%) of individual tumors. In this regard, myoepithe-
liomas are sometimes considered to lie at one end of a biologic spectrum,
with pleomorphic adenoma in the middle, and basal cell adenoma at the
opposite end. The tumors can be classified into four major subtypes, based
on myoepithelial cell morphology: spindle cell, hyaline, plasmacytoid, and
clear cell. All subtypes commonly have a collagenous or myxoid stroma. The
spindle cell type shows an interlacing, fascicular pattern of growth (e-Fig.
6.19). Loosely cohesive myoepithelial cells with eosinophilic cytoplasm and
eccentric round nuclei are present in the plasmacytoid variant (e-Fig. 6.20).
The clear cell type is composed of sheets and aggregates of dear cells (e-Fig.
6.21). The tumor cells express typical myoepithelial markers such as S-100,
pan-cytokeratin, smooth muscle actin, and p63.
Simple excision is generally the treatment of choice unless malignant fea-
tures, such as infiltrative growth or perineural invasion, are encountered,
in which the diagnosis is myoepithelial carcinoma. Myoepitheliomas have a
slightly lower recurrence rate than pleomorphic adenomas.
B. Malignant neoplasms
1. Mucoepidermoid carcinoma is the most common salivary gland malignancy.
The major salivary glands account for more than half of all cases, with most
arising in the parotid gland. These tumors also arise from minor salivary
glands in the oral cavity, particularly in the hard palate, buccal mucosa,
lip, and retromolar trigone. They may also develop intraosseously in the
mandible and maxilla, but such tumors are considered odontogenic in ori-
gin and have a different clinical behavior. Mucoepidermoid carcinomas are
slightly more common in women, and the mean age of affected patients is
approximately 45 years. The tumor also occurs in children and is the most
common pediatric salivary gland carcinoma. Patients usually present with a
painless, slowly growing mass, but may also present with a blue-red superfi-
cial nodule along the oral mucosa mimicking a mucocele or vascular lesion.
Grossly, both solid and cystic components may be present, often with
mucinous material within the cysts. Microscopically, the hallmark of these
tumors is the presence of the three different cell types: mucous, squamoid
(epidermoid), and intermediate, although frequently squamoid cell are incon-
spicuous. These three cell types are present in variable proportions in individ-
ual tumors and form sheets, nests, duct-like structures, or cysts. Intermediate
cells frequently predominate, and range from small basal cells with minimal
basophilic cytoplasm to larger oval cells with pale eosinophilic cytoplasm.
The mucous cells occur singly or in clusters and have pale, foamy cytoplasm,
distinct cell membranes, and eccentric small nuclei (e-Fig. 6.22). They fre-
quently line cystic spaces and are positive with mucicarmine and PAS stains.
Squamoid cells have abundant eosinophilic cytoplasm and vesicular nuclei
Qjt!lc c:ompononl45lr. +2
1\nno<fiUrtlnvadooln ..,.II ,_.hd r.iando +2
Pronoui!OIId nucleoroiYI>Io +2
-Ia
"fmpht!fe •ndlor...,ulorl........., +$
-.,11- +3
+3
4 Of """"m~O hW! _ , llalcb i3
Bony-ion i3
I 'IIIII Polit-
~ o
~rmedll1D w
HW! 4 or mont
Chapter 6 • Salivary Glands I 89
5. 11Malignant mixed tumor" is a broad term that is used to encompass true malig-
nant mixed tumors, carcinoma ex pleomorphic adenoma, and metastasizing
mixed tumor.
a. True malignant mixed tumor (or carcinosarcoma) is a malignant neoplasm
that is composed of both carcinomatous and sarcomatous components,
and is exceedingly rare. The mean age is 58 years, and one-third of patients
have evidence of a preexisting pleomorphic adenoma. Approximately two-
thirds of cases arise in the parotid gland, 15% in the submandibular gland,
and 15% in the minor salivary glands of the palate.
Grossly, there is typically a firm, tan-white mass with hemorrhage,
necrosis, and, on occasion, grittiness or calcification. Microscopically,
there is an intimate admixture of the two components. The carcinoma-
tous component typically takes the form of high-grade salivary duct carci-
noma or undifferentiated carcinoma (e-Fig. 6.31). The sarcomatous com-
ponent is usually chondrosarcoma or osteosarcoma, but fibrosarcoma,
leiomyosarcoma, and even liposarcoma occur rarely. Treatment consists
of wide local excision combined with radiotherapy. The tumors are very
aggressive, with up to two-thirds of patients dying of disease, usually
within 30 months.
b. Carcinoma ex pleomorphic adenoma is defined as a mixed tumor in which
carcinoma is present. This tumor accounts for >95% of malignant mixed
tumors, and is most common in the parotid gland, followed by the minor
salivary glands, submandibular gland, and sublingual gland. Most patients
are in their sixth or seventh decade, which is approximately 1 decade older
than the age of most patients who have pleomorphic adenomas. The classic
history is a patient with a longstanding mass that undergoes rapid growth
over a period of several months.
Grossly, these tumors can reach up to 25 em in diameter, and the aver-
age size is more than twice that of pleomorphic adenomas. The carcinoma-
tous component is usually an infiltrative, hard, white to tan-gray mass with
hemorrhage and necrosis. Microscopically, the proportions of the carci-
noma and pleomorphic adenoma are quite variable, and the pleomorphic
adenoma component may be replaced by scarring or may be overgrown
by the malignant component. The malignant component most often is a
poorly differentiated adenocarcinoma not otherwise specified (e-Figs 6.32
and 6.33), salivary duct carcinoma, or undifferentiated carcinoma. Low-
grade carcinomas such as myoepithelial carcinoma occasionally occur.
Prognosis and management are highly dependent on the type of
carcinoma and extent of invasion. The malignant component should be
classified as noninvasive (intracapsular), minimally invasive (~1.5 mm
in greatest extent), or widely invasive (>1.5 mm). Wide resection is the
treatment of choice, with lymph node dissection and radiation therapy
reserved for widely invasive tumors or tumors with obvious cervical
lymph node metastases.
c. Metastasizing mixed tumor is the least common form of malignant mixed
tumor. These tumors have the same bland morphology of a pleomorphic
adenoma, but metastasize either to local lymph nodes or distantly, usually
to bone and lung. Often, there is a protracted clinical course with many
recurrences at the primary site before nodal or distant metastases develop.
Overall mortality due to the tumor is 40%.
6. Salivary duct carcinoma is one of the most aggressive primary salivary gland
tumors and accounts for <10% of salivary gland tumors. Men are more
commonly affected in a ratio of 4:1. Patients generally present in the sixth
decade with a rapidly growing parotid mass with facial nerve involvement.
Grossly, the tumors are solid and white with hemorrhage, necrosis,
and cystic areas. Infiltration of the surrounding tissue is usually apparent.
92 I SECTION 1: HEAD AND NECK
Toll>
Vf!l'/--d-
Turl'IQ' 1 -sit!, mondiiM.-<anal, •ndlor 11<:~1 norw
Cytopathology of the
Salivary Glands
Brian Collins
The parotid and minor salivary glands are superficially located and palpable, and so
salivary gland lesions are readily amenable to percutaneous FNA. A pattern approach
is typically utilized for the evaluation of FNA specimens for diagnosis, classification,
and clinical management of patients.
I. NON-NEOPLASTIC CONDITIONS
A. Chronic sialadenitis. Chronic inflammation can cause diffuse or focal enlarge-
ment which by palpation can be indistinguishable from a neoplasm. However,
chronic sialadenitis is very tender on FNA, which can be a helpful due to diag-
nosis. Aspiration smears are hypocellular with benign ductal epithelial groups
which have a minimal degree of nuclear atypia. Acinar elements can be scant
or absent due to the underlying destructive nature of the process. A variable
mixture of large and small lymphocytes is present. Acinic cell carcinoma is
an important differential diagnostic consideration when presented with bland
acinar-type cells and lymphocytes on aspirate smears.
B. Lymph nodes. The parotid is a common site of intra-parenchymal lymph nodes
which can be clinically difficult to distinguish from a neoplasm. Aspirate smears
will show a mixture of lymphocytes in the pattern typically seen with reactive
lymphoid hyperplasia.
II. BENIGN NEOPLASMS
A. Warthin tumor. This tumor commonly arises in the parotid gland and by aspi-
ration shows a mixture of lymphocytes and cohesive groups of oncocytic-type
epithelial fragments (e-Figs. 6.39 and 6.40). The lymphoid elements are inti-
mately admixed with the oncocytic cells, and the background usually shows
cystic change and with macrophages and granular debris.
B. Pleomorphic adenoma. Aspiration smears can show a wide variety of patterns
and cellular elements. Commonly, there is a mixture of bland epithelial, spin-
dled, and myoepithelial-like cells which occur as single cells and are intimately
admixed with a variable amount of fibromyxoid stroma (e-Figs. 6.41 and 6.42).
The relative proportions of these elements can vary significantly between cases.
Diff-Quik staining of smears helps to highlight the metachromatic fibromyxoid
stroma. When present in the customary pattern, a definitive diagnosis can be
rendered.
96 I SECTION 1: HEAD AND NECK
C. Basal cell adenoma. Aspiration smears are cellular and contain small uniform
basaloid cells with scant cytoplasm and round to oval nuclei with indistinct
nucleoli. The dense extracellular material present, usually less than that seen in
pleomorphic adenoma, tends to be arranged in globules and trabecular-tubular
configurations (e-Fig. 6.43). This pattern typically is classified as a basaloid neo-
plasm and the differential diagnosis includes adenoid cystic carcinoma, since the
cytomorphologic pattern on aspiration does not permit definitive classification.
Ill. MALIGNANT NEOPLASMS
A. Adenoid cystic carcinoma. Aspiration shows small basaloid cells with globules
of dense matrix material. The cells are monomorphic with round to oval nuclei,
even chromatin, indistinct nucleoli, and scant cytoplasm. Aspirates are usually
cellular with cell groups arranged in syncytial and three-dimensional fragments.
The extracellular matrix consists of dense amorphous material with tubular
and/or globular shapes (e-Figs. 6.44 to 6.46). Classically, the cells appear to
surround the round globules. Because of the cytomorphologic overlap with other
basaloid tumors, definitive classification is not possible.
B. Mucoepidermoid carcinoma. These neoplasms vary in their cellular composition,
and this difference is reflected in the patterns and cellular elements encountered
by FNA. For low-grade neoplasms, aspiration smears are hypocellular and con-
tain thick mucoid-like material with granular debris. The three cell types that
characterize the tumor (squamoid or epidermoid), mucous, and intermediate
are all present, and they can be arranged separately or be admixed. The nuclear
features of all three cell types are bland (e-Figs. 6.47 and 6.48). For high-grade
neoplasms, aspirate smears are cellular with groups and sheets of cells with a
squamoid appearance. Nuclei are large with coarse chromatin and prominent
nucleoli; the cytoplasm is dense, and moderate in amount with well-defined cell
borders. Mucin cells with intracytoplasmic vacuoles are usually few and can be
difficult to identify (e-Figs. 6.49 and 6.50).
C. Acinic cell carcinoma. Aspirate smears are cellular with a mixture of single cells,
small groups of cells, and three-dimensional clusters. The cells are intermediate
in size with monomorphic round bland nuclei with nucleoli. The cytoplasm is
critical in the diagnosis and will demonstrate features of acinar differentiation
with delicate vacuoles and granules (e-Figs. 6.51 and 6.52). While usually dean,
the background can show the features characteristic of cystic lesions and show
significant lymphoid elements.
SUGGESTED READINGS
Eveson ]w. Malignant neoplasms of the salivary glands. In: Thompson LDR, ed. Head and Neck
Pathology. New York, NY: Churchill Livingstone; 2006.
Eveson JW, Auclair P, Gnepp DR, et al. Tumors of the salivary glands. In: Barnes L, Eveson JW,
Reichart P, Sidransky D, eds. Pathology and Genetics Head and Neck Tumors. Lyon, France:
IARC Press; 2005.
Faquin WC, Powers CN. Salivary Gland Cytopathology: Essentials in Cytopathology. New York,
NY: Springer; 2008.
Gnepp DR, Henley JD, Simpson RHW, et al. Salivary and lacrimal glands. In: Gnepp DR, ed. Diag-
nostic Surgical Pathology of the Head and Neck. Philadelphia, PA: W.B. Saunders Publishers;
2009.
Layfield L. Cytopathology of the Head and Neck. Chicago: ASCP Press; 1997.
Powers CN, Frable WJ. Fine Needle Aspiration Biopsy of the Head and Neck. Boston: Butterworth
Heinemann; 1996.
Richardson MS. Non-neoplastic lesions of the salivary glands. In: Thompson WR, ed. Head and
Neck Pathology. New York, NY: Churchill Livingstone; 2006.
Torske K. Benign neoplasms of the salivary glands. In: Thompson LDR, ed. Head and Neck Pathol-
ogy. New York, NY: Churchill Livingstone; 2006.
Wenig BM. Major and minor salivary glands. In: Wenig BM, ed. Atlas of Head and Neck Pathology.
Philadelphia, PA: Saunders Elsevier; 2008.
The Ear
Peter A. Humphrey and Rebecca D. Chernock
I. NORMAL ANATOMY. The ear is composed of the external ear, the middle ear, and the
inner ear. The external ear is made up of the auricle, which leads to the external
auditory canal. The auricle has a supporting plate of elastic cartilage, which also
helps form the outer two-thirds of the external auditory canal. Skin covers both
the auricle and the canal; the main distinctive histologic features of this skin are
that the squamous lining of the inner half of the canal is thinned, and that modified
apocrine glands called ceruminal glands are present in the outer third of the canal.
The clustered ceruminal glands are lined by cuboidal epithelial cells that have
an eosinophilic cytoplasm that often harbors a granular golden-yellow pigment
(e-Fig. 7.1).*
The middle ear, or tympanic cavity, lies within the temporal bone. It is separated
from the external auditory canal by the tympanic membrane, a thin fibrous sheet
that has an external keratinizing squamous epithelial lining and an inner cuboidal
cell lining. The middle ear contains the three auditory ossicles (malleus, incus,
and stapes), ossicle ligaments, tendons of the ossicular muscles, the auditory tube,
the tympanic cavity itself, and the epitympanic recess, the mastoid cavity, and the
chorda tympani of the facial nerve (cranial nerve VII). The auditory or eustachian
tube connects the tympanic cavity with the nasopharynx. The tympanic cavity is
lined by a single layer of flattened to cuboidal respiratory epithelium, whereas most
of the auditory tube is lined by low ciliated epithelium.
The inner ear is located within the petrous portion of the temporal bone and
is composed of a membranous labyrinth surrounded by an osseous labyrinth. The
membranous labyrinth houses the cochlea and the vestibular apparatus, both of
which are supplied by cranial nerve VIII. There are several parts to the cochlea:
the cochlear duct with the organ of Corti (the end organ of hearing), and the
scala vestibuli and scala tympani, which hold the perilymph. The organ of Corti
has thousands of neurotransmitting hair cells. The vestibular apparatus, which
functions in motion and position sensing, consists of three semicircular canals and
the utricle and the saccule. The ampullae of the canals have a sensory end organ,
the crista ampullaris, with neurosensory hair cells. The utricle and the saccule
also possess a sensory end organ, the macula, which has neurosensory hair cells
and otoliths. There is also a blind sac in the membranous labyrinth known as the
endolymphatic sac, which is lined by tall columnar epithelium arranged on papillae.
II. GROSS EXAMINATION AND TISSUE SAMPLING
A. External ear. Biopsy and excision specimens should be handled as skin specimens
from other anatomic sites (see Chaps. 38-40).
B. Middle and inner ear. Samples from the middle ear are often obtained in cases
of suspected cholesteatoma. For these cases, it should be noted whether bone
fragments are present. Standard hematoxylin and eosin (H&E) slide preparation
is sufficient. Ossicles from the middle ear can be processed by gross examination
only unless microscopic examination is requested by the surgeon. For middle ear
and inner ear neoplasms, which are uncommon, use of ink to mark the peripheral
margins is not usually necessary because these specimens are typically received
as small fragments. In those rare cases in which the patient has a history of
*All e-figures are available online via the Solution Site Image Bank.
97
98 I SECTION 1: HEAD AND NECK
Ad-lei .,.a...- .
Mucool>i:lo!Tilold oon:t>omo
$qUI/I'oOII4 col Clll'eliiOitlO
Elnbl)oonal ltlabd..,_ms
~and-
An&l<tlmpldd hyJ>o!Jliall lO!Ih eoolnophlll
n ...tl'tltlllilllll..,
AdOIIOINI of1ho mlddloeor
Popllory bJII\Oil
~po1JI~rytumtlf
Selin- Pll>lloma
111'111111dPII>lloma
Squtll'IOUIIcol """!""""
Menln&tomo
Tl_..tl'tl•nw-
v...tllullr tdwwtl"'a"'W
u- oflho . , _ audt.alycol'lll
Homa...ma
E'lldotlmpho11:: ec111nor
He~M'>t!mllhotltumoro
9-aoll eh""le lympkoe)lle l a u - d t,omphoeylfe ljmpi\Grna
·-""-
LM1.Jt1N na coiiiiiiiSoqiDsls
100 I SECTION 1: HEAD AND NECK
overlie the granulation tissue causing diagnostic confusion with reactive pro-
cesses. Cystic spaces with a colloid-like material simulating thyroid follicles
can also be present. Immunostains show positivity for keratin, with variable
S-100, glial fibrillary acid protein (GFAP), and synaptophysin immunoreac-
tivity; thyroglobulin immunostaining is negative. Despite the bland histologic
appearance, the tumor is a slowly growing, locally aggressive, but nonmetas-
tasizing neoplasm. Temporal bone invasion is common, and the tumor may
extend into the cerebellum. Outcome is related to size of the tumor and ade-
quacy of excision; radical surgical excision affords the best chance for cure.
Recurrence is seen in about 20% of cases, with tumor-specific death in about
13% of patients.
4. Meningioma in the middle ear is a rare neoplasm that is more likely to rep-
resent secondary extension from an intracranial meningioma than from a
primary middle ear meningioma. Patients with primary middle ear menin-
giomas present at a mean age of 50 years with hearing changes and sometimes
otitis and pain (Mod Pathol. 2003;16:236). The histopathologic features
are similar to intracranial meningiomas, with meningothelial meningioma
predominating (e-Fig. 7.17). Vimentin, progesterone receptot; and epithelial
membrane antigen immunostains are positive, and cytokeratin immunostains
are negative. Meningiomas are slowly growing neoplasms and can recur fol-
lowing incomplete surgical excision. The 5-year survival is 83%.
5. Squamous cell carcinoma in the middle ear is uncommon and is typically
advanced at presentation. Its development is not clearly related to chronic
otitis media and is not related to cholesteatoma. Microscopically, the carci-
noma is keratinizing and displays a variable degree of differentiation. Out-
come is related to tumor extent and margin status at surgery, but not his-
tologic grade. The 5-year survival is roughly 50% (Int j Radiat Oncol Biol
Phys. 2007;68:1326).
6. Other rare neoplasms of the middle ear include embryonal rhabdomyosar-
coma (e-Fig. 7.18), lipoma, hemangioma, osteoma, ossifying fibroma, and
teratoma.
7. Metastasis to the ear is uncommon, accounting for only 2% to 6% of all
neoplasms of the ear. Most patients have known, widely disseminated can-
cer. The middle ear is the most common site for metastatic spread. Breast,
lung, and prostate are, in ordet; the common primary sites of origin for the
metastatic deposits.
V. NEOPLASMS OF THE INNER EAR. Vestibular schwannoma, lipoma, and hemangioma
are the most common neoplasms of the inner ear. Endolymphatic sac tumors also
arise in the inner ear (the aforementioned aggressive papillary tumor of the middle
ear is thought to represent an endolymphatic sac tumor with extension into the
middle ear).
A. Vestibular schwannoma (acoustic neuroma) is relatively common. Unilateral
vestibular schwannoma accounts for 5% to 10% of all intracranial tumors,
and has been found in about 1% of autopsies. Bilateral vestibular schwan-
noma, found in 5% of all vestibular schwannoma cases, is characteristic of
neurofibromatosis type 2 (an autosomal dominant disease due to mutations in
the NF2 gene on 22q12). Patients, usually in their 40s or 50s, present with pro-
gressive hearing loss and tinnitus. Grossly, the size range of the tumor is a few
millimeters up to 6 em in maximal dimension. The smaller tumors are round
to oval, whereas large tumors can assume a mushroom shape. The cut surfaces
are yellow, and can exhibit hemorrhage and cystic change. Microscopically, the
attributes are the same as in soft tissue schwannomas (e-Fig. 7.19), with Antoni
A regions with Verocay bodies, and Antoni B areas. Mitotic figures should be
rare. Degenerative nuclear atypia should not be taken as a sign of malignancy.
S-100 immunoreactivity is strong.
Chapter 7 • The Ear I 103
B. Lipomas of the internal auditory canal resemble lipomas at other anatomic sites,
except that cranial nerve VII or VITI or their branches may be present in the
lipoma.
SUGGESTED READINGS
Barnes L. The ear. In: Silverberg SG, ed in chief, Silverberg·s Principles and Practices of Surgical
Pathology and Cytopathology. Philadelphia, PA: Churchill Livingstone Elsevier; 2006:2268-
2287.
Barnes L, Eveson JW, Reichart P, Sidransky D, eds. Tumors of the Ear, Chapter 7. In: Pathology
and Genetics Head and Neck Tumours. Lyon, France: IARC Press; 2005.
Wenig BM. Diseases of the External Ear, Middle Ear and Temporal Bone. Chapter 9.ln: Barnes L,
ed. Surgical Pathology of the Head and Neck. New York, NY: Informa Healthcare; 2009:423-
474.
SECTION II
Thorax
Lung
Jon H. Ritter and Hannah R. Krigman
I. NORMAL ANATOMY. The lung is defined by airway branching, first into right and
left lobes, then segments, and finally, the functional unit of the lung, the lob-
ule. Arteries follow the airways, while veins and lymphatics flow toward lobular
septa and finally to the hilum and main pulmonary veins. Bronchi are lined by
a pseudostratified respiratory epithelium that includes goblet cells, which is sep-
arated by basement membrane from a delicate submucosa. Larger airways have
a smooth muscle wall that contains minor salivary glands and a cartilaginous
skeleton; bronchioles have lost the latter components. Each lobule has a cen-
tral bronchovascular bundle; the interstitial space between the pulmonary artery
branches and the bronchioles is eventually continuous with the alveolar intersti-
tium. Lymph nodes occur within the lung, and also are discontinuous along the
bronchi.
Progressive branching of the airways leads to the alveolar ducts, from which
alveoli spring. Normal alveolar walls are very delicate and have a fine elastic tissue
matrix. The barrier between the blood in capillaries and air in the alveolar space
consists of the endothelial cell, the basement membrane, the wispy alveolar inter-
stitium, epithelial cell basement membrane, and the flattened type 1 pneumocyte.
Type 2 pneumocytes can proliferate to replace injured type 1 cells.
The visceral pleura consists of an inner vascular layer that abuts the alveolar
tissue, a connective tissue layet; and an outer layer of mesothelium. The visceral
pleura is reflected back at the hilum and becomes continuous with the parietal
pleural layer that lines the chest cavity.
II. SPECIMEN HANDLING AND REPORTING
A. Samples. Biopsies of lung tissue for diagnosis are obtained endoscopically
(transbronchial or endobronchial biopsy), via radiologically guided procedures
(usually needle core biopsies), or for peripheral disease, via wedge biopsies.
Resections can involve a single lobe (lobectomy), two lobes (bilobectomy), or
an entire lung (pneumonectomy.)
B. Gross examination and sampling
1. Endoscopic biopsies are usually submitted in a single cassette. The fragments
are small; use of a nylon mesh bag or a filter paper wrap is preferable to
submission on biopsy sponges since tissue can be compressed or lost in
the pores of sponges. The number of fragments, range of size, and color
should be recorded. Three hematoxylin and eosin stained levels should be
examined. For smaller biopsies, additional unstained levels may be cut at
the time of initial sectioning in case special stains are needed.
104
Chapter 8 • Lung I 105
*Aile-figures are available online via the Solution Site Image Bank.
Cl'l••a• urc I 1a7
~if.t:]lj:l'l
.................
Yhi ...
~IMino
Olt.er lftfed:lor'l$ ... fMMUftoetlfnpra"nlill!td
l"!!llnl>
Olnlln
Smoke
ChGII'lOihe,.,.,IIII:OIIIIltO
Amlode"""'
NllroiiJro-
Sllcc:k Tra~mtl.lk:
C.....nit
Ollw
Al\'f-nlltn
Rodla1!orl
au....
OonloptJIMOMIJ by-
Pin-
Lllpul
1aa I SECTION 11. THORAX
LAM is centered in the lung, it also involves lymph nodes in the pulmonary
hilum, mediastinum, and abdomen in most cases. The smooth muscle-like
cells stain with vimentin, desmin, and smooth muscle actin, and also may
express estrogen and progesterone receptor. LAM also shows cross-lineage
staining with melanoma markers including HMB-45 and melan-A. In this
regard, the cells of LAM share the staining attributes of the members of the
PEComa family (including the sugar tumor of lung), renal angiomyolipo-
mas, and some soft tissue tumors (see Chap. 46). LAM also shows overlap
with tuberous sclerosis (TS) in that some TS patients develop an identi-
cal cystic lung disease, and both LAM and TS share an association with
angiomyolipomas in the kidney and elsewhere. The course of the disease is
unpredictable; transplantation has been the only long term option for those
with severe disease.
7. Alveolar proteinosis refers to a peculiar accumulation of intra-alveolar
eosinophilic, granular PAS-positive protein and phospholipid. It was ini-
tially reported as an idiopathic process, but a relation to immune deficiency,
hematologic malignancies, infections, and various exposures is now recog-
nized. Classic exposure-related cases are associated with massive acute silica
exposure, which is believed to poison the alveolar macrophages and thus
inhibit their ability to dear alveolar debris.
Clinically, patients present with slowly progressive alveolar infiltrates
and complain of dyspnea, cough, or sputum production with fever; CT scans
show "crazy-paving" with alveolar infiltrates and septal line thickening. The
diagnosis is often apparent from the milky appearance of lavage fluid; biop-
sies show complete filling of the alveolar spaces by granular eosinophilic
debris that is PAS-positive and diastase-resistant (e-Fig. 8.12). Findings
often include cholesterol clefts (acicular clefts), globular eosinophilic debris,
and macrophages. In most cases the underlying alveolar structure appears
normal, although some chronic cases may eventually show fibrosis. Sec-
ondary infection of the fluid by Nocardia, mycobacteria, and fungi has
been reported. Pneumocystis infection can microscopically mimic alveolar
proteinosis, although the material in alveolar proteinosis lacks the frothy
appearance characteristic of Pneumocystis infection.
D. 11AIIergic" diseases
1. Eosinophilic pneumonia can be divided into acute and chronic forms. The
acute forms include the "simple form" also known as Loeffler syndrome;
this is an acute, self-limited process with fleeting infiltrates and peripheral
blood eosinophilia and is rarely biopsied. The tropical form is usually linked
to filaria infection, and also presents as an acute illness. The chronic form
is more likely to require biopsy for diagnosis.
Chronic eosinophilic pneumonia (CEP) has a variable presentation from
acute illness with fever, dyspnea, and weight loss, to vague respiratory com-
plaints. Many patients have a history of asthma, and laboratory tests reveal
elevated blood IgE and peripheral blood eosinophilia. Chest radiographs
show patchy non-segmental infiltrates, often peripheral, that may cross fis-
sures, a pattern that is sometimes described as the "photographic negative of
pulmonary edema." There are myriad underlying causes; major categories
include drugs (antibiotics such as nitrofurantoin, sulfonamides, penicillins,
anti-inflammatory agents, and chemotherapeutics), fungus (Aspergillus and
Candida), parasites, nickel vapor, and idiopathic cases. Histologic sec-
tions of CEP (e-Fig. 8.13) show an alveolar-filling process consisting of
a mixture of eosinophils and macrophages. Necrosis of eosinophils may be
present, forming an eosinophilic abscess. Charcot-Leyden crystals will also
be present, as well as interstitial and perivascular eosinophils, lymphocytes,
plasma cells, and areas of BOOP.
.,._,,.• C..IIbulla III•IGIIG-
Goodpuluro'l +1- +, UnMJitl!n.,a
ldlojolllll:: - I t
w..>non +
Ml:rooooi>lc """"""!til
~lqon """"'~'d..... (SLE)
+
+
ldlojollllle rapi<IO,q:ro.,.- GN +
Tadnt
11& I SECTION lit THORAX
is basophilic (e-Fig. 8.20). Epithelial cells, vascular cells, and even stromal
cells can exhibit inclusions. Immunohistochemistry for CMV will decorate
the enlarged cells; electron microscopy demonstrates virions in the inclu-
sions, while a PAS stain with diastase highlights the cytoplasmic deposits.
2. Herpesvirus (HSV). Both HSV types I and ll can induce pneumonitis, and
immunocompromised hosts are more prone to Herpes viral pneumonia.
Clinically, HSV pneumonia may result from extension of upperway disease
with primarily bronchiolar inflammation, or from systemic infection which
presents as multiple small perivascular inflammatory nodules. Three find-
ings are characteristic of HSV: necrosis, individual cells with inclusions (e-
Fig. 8.21) (featuring eosinophilic nuclear inclusions with perinuclear clear-
ing; amphophilic nucleoplasm in single cells may represent early inclusions
or giant cell formation), and herpes viral giant cells (which contain two or
more nuclei with ground glass central nuclear clearing and coarsely gran-
ular, sharply defined nuclear borders; the nuclei are often molded against
one another). Immunohistochemistry using antibodies to HSV decorates
the giant cells and the individual cells with inclusions.
3. Varicella zoster (VZV) is the causative agent for chicken pox and shingles.
Primary infection in healthy adults and immunocompromised children can
result in pneumonia. The underlying lung injury pattern can be either DAD
with hyaline membranes and proteinaceous exudates, or nodular inflamma-
tion with central necrosis that calcifies and persists on chest radiographs.
Biopsies of VZV infections show giant cells similar to those of HSV.
4. Adenovirus infection generally presents with symptoms typical of an upper
respiratory infection; pneumonia develops in a small percentage of healthy
and immunocompromised children and adults. Two patterns of infection
evolve. Some patients develop necrotizing bronchiolitis and pneumonia,
while others respond with DAD with hyaline membranes and exudates.
In adenoviral infections both alveolar lining cells and bronchial epithelial
cells may exhibit blurred and hyperchromatic nuclear chromatin (so-called
smudge cells) (e-Fig. 8.22). The bronchial damage of adenovirus may result
in fibrosis or constricting bronchiolitis.
5. Respiratory syncytial virus (RSV) affects primarily small children and infants;
premature infants and immunocompromised children are particularly prone
to infection. RSV infection exhibits some seasonality, and is more common
in fall and winter. RSV can induce bronchiolitis with symptoms of cough,
wheezing, and respiratory distress. Biopsies show necrotizing bronchiolitis
and/or interstitial pneumonia.
6. Measles virus. With the advent of vaccination, infection by measles virus is
rare, and evolution to pneumonia rarer. Most patients are immunocompro-
mised, and the characteristic skin rash is present. The underlying pathology
is DAD, with associated individual and giant cells with viral inclusions;
the alveolar spaces may contain exudates, and necrosis may be present.
Both eosinophilic intranuclear and cytoplasmic inclusions develop in both
alveolar and vascular lining cells. Measles pneumonia features a distinc-
tive multinucleate giant cell thought to derive from coalescence of type n
pneumocytes, the Warthin-Finkeldey giant cell, which contains up to 60
nuclei.
7. Parainfluenza and influenza generate non-specific patterns, consisting of
varying degrees of DAD, bronchial necrosis, and peribronchial inflamma-
tion.
B. Bacterial infections
1. Mycoplasma induces acute and chronic bronchiolitis, with necrosis and
denudment of bronchial epithelium. The bronchial lumina may contain
acute inflammatory cells admixed with denuded epithelium. Acute and
Chapter 8 • Lung I 1 19
chronic inflammatory cells often traverse the bronchial wall. Alveolar spaces
may exhibit bronchopneumonia, with BOOP or DAD.
2. Mycobacterial infections are typically grouped into tuberculosis and other
(atypical) mycobacterial infections. The most common stain used to demon-
strate organisms in tissue sections is the Ziehl-Neelsen stain; immunoflu-
orescent (auramine-rhodamine) and immunohistochemical stains can also
be used to identify mycobacteria. PCR based genetic methods can also be
used to detect (and speciate) the organism in tissue sections.
a. Tuberculosis (TB). Multidrug resistance has emerged in this organism and
consequently the pathologic spectrum of tubercular infection is expand-
ing. The causative organism is Mycobacterium tuberculosis. which is
transmitted via inhalation of organisms. Histologically, the tubercular
granuloma is a classic palisaded necrotizing granuloma. Granulomas
may caseate and coalesce, creating nodules with central necrosis, or even
cavitary masses. Organisms may be found in multinucleate giant cells or
at the periphery of necrosis (e-Fig. 8.23). Lymph nodal involvement may
be present. Miliary tuberculosis is unlikely to be sampled in biopsy or
resection specimens.
b. Atypical mycobacterial infection. Mycobacterium avium intracellulare
(MAl) is the most common of the atypical mycobacterial infections, and
is more common among immunocompromised patients where it presents
with non-specific fever and malaise. Radiographs show diffuse or patchy
infiltrates. Histologic findings vary from more typical non-necrotizing
punctuate granulomas, to necrotizing granulomatous pneumonia, to dif-
fuse pneumonitis (in which abundant pneumocytes or interstitial cells
contain abundant organisms by acid fast stains).
C. Fungal Infections
1. Candida infection arises in several contexts. Mucocutaneous candidiasis can
arise in immunocompetent adults, but arises more frequently in immuno-
compromised patients; the trachea or bronchial tree can have plaques of
fungus admixed with desquamated cells and neutrophils. Impaired host
defenses potentiate invasive disease; in the lungs, vascular invasion results in
hemorrhagic necrosis. Direct inoculation into the bloodstream from iatro-
genic sources (catheters, surgery) or other inoculation (drug abuse) results in
disseminated disease. Transplant patients can develop infection at the sites
of anastomosis. C. albicans is the most prevalent species; other species more
often infect compromised hosts. All species exhibit the same basic morphol-
ogy: non-branching, aseptate pseudohyphae forming "box-car" like chains
of cells; yeast forms bud from pseudohyphae (e-Fig. 8.24). Yeast forms are
visible on H&E, PAS, or silver stains.
2. Mucormycosis. Several members of the Phycomycetes result in clinically and
morphologically identical disease, including Mucor, Absidia. and Rhizopus.
among others. Almost all cases occur in the setting of diabetic ketoacidosis
(sinonasal or rhinocerebral disease) or immunosuppression from hemato-
logic malignancies. Lung involvement may be the primary focus, or develop
secondary to head and neck disease. Lung lesions are typified by hemor-
rhagic pneumonia; fungal thrombi with distal infarction are often present.
The dual circulation of the lung (bronchial and pulmonary arterial systems)
results in perfusion of infarcted areas with resulting hemorrhagic necrosis,
associated with varying amounts of inflammation. A key to recognition of
the infection is identification of wide, ribbon-like non-septate hyphae with
irregular wide angle branching. The pseudohyphae are often described as
"empty" and stain poorly with most special stains.
3. Aspergillus species cause a wide spectrum of disease, dependent on both
host immune status and site of growth. Colonization with Aspergillus can
120 I SECTION II: THORAX
""'-
c. Clll• otc alrRJ 11111:11•
oo,.boent
II ..:tllol•tull..._
Cl,_.
~ -·..-•nJ-
124 I SECTION lit THORAX
I'1,1,! J!U1
I :=~e~~llktlra.~~nt '" tht Gnd~ 4f .Nqratt c:.~r.- •
.,.,...... ClrcJIIIIIIMIIII C]IIIIIIP; IMIIIM
IIIII• (llllla1aiu lll~allll all tll~ata
NJ None
A1
.,.,,.ft.-.
RaA>, hen! to ... at
... _!&_
~.~
celllllek """"'
1-2
J4::n*~"
lnftlbft,MJIIfNOn lymph..,. IIIIo
ot ...mlnt-r. IIOIIM/lllhlh
2-o...uthll:tt cufl'>
Often hiOI\Y-Ie, More ocd\la1Dd.
Inn-Into nw1Y
....lit_, eai11~h!II:J<!:I!In
"llt.eli111. btnelbopllb
Mq bo <~>nfluon~ S!mllrloA3
-
.......... .......,_
B0
BIR
8211
NMO
Mid
...,.1111od .,"•'"~"
Moda1111D to - •
(jlltlllll<la-
N/A
No
Yao, lncHdual ..!la(IIIC>ols.to u - .
11*1 d.., lldo11>h o!<llilllelllrn
Chapter 8 • Lung I 125
history suggests nodules or a mass. Cases usually fall into three general
categories: plasma cell hyperplasia, consisting of cases that show low-grade
findings; polymorphous PTLD, which is comprised of a mixed population of
atypical lymphoid cells; and monomorphous PTLD, consisting of cases that
consist of a monotonous population of high grade, malignant-appearing
cells (e-Fig. 8.37). Monomorphous cases are much less likely to regress
following diminution of immunosuppression as compared with the lower
grade lesions; thus, higher grade cases are likely to require cytotoxic therapy.
In allograft biopsies, if there is a question of PTLD versus a rejection-related
infiltrate, a simple panel of CD3, CD20/CD79a, and EBV immunostains will
provide dichotomous results: the infiltrate in severe rejection or infections
is almost always a CD3 predominant infiltrate, while the infiltrate in PTLD
is positive for CD20 and CD79a, as well as EBV, in the great majority
of cases. Adjunctive studies to indicate clonality may also be of value in
terms of diagnosis, classification, and prognosis. It is important to note
that rare examples of PTLD may have the morphology of Hodgkin disease,
and that late cases of EBV negative lymphoid neoplasms (4 or 5 years after
transplant) which resemble non-Hodgkins lymphomas can also develop.
7. Recurrent disease. With the exception of a few cases of sarcoid, recurrence
of the native lung disease in allografts is not a significant problem. There
are reports of occasional cases of carcinoma arising in transplanted lungs,
as well as in the native lung in the setting of unilateral transplant; these
patients fare poorly.
VII. NEOPLASMS. The WHO classification of lung neoplasms is presented in Table 8.12.
Malignancies of the lung are the most common cause of cancer-related mortality
in the United States. Despite decades of warnings, cigarette smoking remains the
predominant risk factor for the development of pulmonary carcinoma. There has
been speculation that a shift in location and cell type for pulmonary carcinomas
is related to changes in cigarette usage. Some data suggest that filtered cigarettes,
which remove larger tar particles, have allowed carcinogens to penetrate to more
distal parts of the lung and produce peripheral adenocarcinomas instead of cen-
tral squamous carcinomas or small cell carcinomas caused by larger particles.
Changes in tobacco formulation over the years may also have played some role
in these changes. Other risk factors for lung carcinoma, either proven or specula-
tive, include asbestos exposure, radiation, various chemicals, heavy metals, viral
infection, fibrosing lung diseases, immunosuppression, and genetic syndromes.
Outside of these causes, there remain some cases of lung carcinomas for which
there is no clear etiologic factor.
Up to 10% of patients with head and neck carcinomas may harbor a concur-
rent lung primary carcinoma; therefore, the finding of a lung nodule in a head and
neck cancer patient should not automatically be assumed to represent metastatic
disease. Similarly, 2%-5% of lung carcinoma patients present with apparent syn-
chronous lung primary tumors (which may be due to the greatly improved resolu-
tion of CT techniques which can now detect additional small nodules in patients
with a dominant lung masses); histologic sampling of these lesions often shows
alveolar proliferations with varying degrees of atypia, which may represent pre-
cursor lesions in the peripheral lung similar to squamous dysplasia in the more
central airways.
A. Pathologic reporting. The important data to be included in a diagnostic report
of a primary pulmonary neoplasm are summarized in Table 8.13. Generally,
tumor size is assessed by gross examination, and this measurement suffices
unless there is confounding fibrosis or peritumoral pneumonia. The extension
of tumor through the pleura covering the lung (visceral pleura) or into the
pleura lining the chest wall (parietal pleura) should be noted; an elastin stain
can delineate these layers in some cases. Lymphatic and venous invasion should
12& I SECTION lit THORAX
-
Slnallt:flll-..
Combllll!ld .,.,.., c:o!lcorcmm•
--
Uoll!l<eii<Orcr.orno~ 11\obd<*l llilen<Cli>G
~ CdJ1:Iir<Jmi>
Pioomorplilc ..,,...,.m,
Splndll col carclnana
Giant oall <::e~a
Cl_,..ft\11
Pu"-Y lilolomo
CiHtlflolt1 """"'
'Pfl>bll-lndd
Alypbl a,...,.ld
SIJfwqfl{itnd liNnIn
MUCIIO~ ..tli:ln"""
--
hllf\OI:I oY*<Ordnotr~~~
F~>iiii•H'n)ololihollol Clldnorna
(~
Cl'l••a• urc I 127
~ .......... poplkma
Eaoplljl!o
Inwo11d
O~lllr>oJ:ilbrno
--...
Mlad tquarnoua col ond -..ndulor 1JOI>IIo1N
A-...
Popllory allan"""
Ad.,.maa of 11>o ~pond IYJlO
MUCO<Il.atond adenM>O
f'loomorglje odei'Jolno
Ollw&
Mudnow:QJ~:tld«<cn,..
~ltllfiiiiiii!IM-ra
MolltlollMO lkollll!nDhomo of Che MALT"""
L,..pi\Gnl_ .,...,_Ia
DI!I'IM la,...lkool "'"PhM>O
LlnJIIIIN,. coil hiiiSi>qiDSII
... ~ ....... tl. .
Hlrmltoma
~homiiii&IM>o
O.r <:611:l.l"n0r
Gttm 011111 tum.:R
~~mllure
Inmltne
Oll>or..., .... tl.mot>
lrrtn~pul'noiiiJIIdl)momo
---MolafiOitlll
-d-l'tifhtJ/J:cy111111---cc.•
dthe"""'-.
r . , , - WD, Bnmi>!IIa E. MOIIer.....,dink HK.
lp: W\C Preoo; 2004. U&od Mfi pe-.
IIIMtt-. - .
'lll)m<nlllld-
'''
1X
,n..-m Prtnlr.ttiiiTIOf.. mot bo 110101011. ..-!Lmor pnMII1 i>Jt!IO prooon01t<ll
m...,..nt .... ,., "'l'//lln or tnodllal WMI!Inp bit not "'""llzod 1>J
1m,..« l:toh<iloooo!11'
TO N o - at roimii)IM>•>r
1k C.!d>oruln d:u
TI Tumor a em or ms1n arut.t dlmllnslonJ aurroundMI "t tunc a
-.u piMn, - u t Iron""""""'I : -tllnvulon """'
'"""*""11Nn t110 lol8' b<"onl:llua O.e., not In IIH> meln bfml:tlul)'
n. 111m..- 2.tm or lao>lh ·-dmonolorl
nb 11im«mor&thon2<m but$em0flouln _ _ d _
T2
fdk>oll\ll IMtllr• (T2 tllmriiS""'-
Turnor mor&thtn $<:rn but7 an Of lieu OftuMOrwttk .arrtdthe
flllu,..o1'6cllsslfled T21 H
oem ..-loll); 1 - . moln l••nctuA. 2.<m Of moro dii\OII to tho
<~rinl; '"""""'-.' plouro;A:ooo<;ll!lodwtlhn-or
T.lo
T21>
l3
IIM>IIoo""' de knw
11im«mor&thon$<m butSemOflouln _ _
Tum« mol'61hlll Scm but 7 em Of lou In fii'IOI1olld-
Tum«mol'61hll17<m0font1Nftd~l-•nyoftho~
d_
obol.rlical'& lltiOUrnoNI!o lllot-.:14'> lllo Nlar ret1o1r blrtdoeo
-· -1
Nl
mlolllld lntnopumo- -ln<ludqln"'llanonlt>ydnct
WilD
Tlllllroalt,o
Clrdnotl
.. ...
~i(.1:!1j:l(il l'<liltura of Put_,.,- .... IOII*>cdllllleoplu!M
,,
AI tJii'RIIII:IN
GraoolNEC
Colli Slit
Moclum
toler•
_,.
llllrr:IMr
Non It
....
<2/hpf N..,.
lollallplll
llacttollt hlaltlal
'-
~I Gred&2NEC Mtdum Mlil 2-lMIDI Focol tntorrnodlllo
Co- to flllll'
Lilli" Col Nou· G-3NEC, Llrp MI!Ud >!MIDI Exllns!\lre Hfall
IQM'Idoc:tfne lMIIt Coil
c.rmoma tJpe
Small Col Nou· Graoo3NEC, Smllll Hl!;l Eans!'te Htpl
"""rlald
I'Ofllldocdno sm•eon
Cormoma typo
Chapter 8 • Lung I 135
and stains TfF-1 are also positive in the majority of cases of pulmonary
small cell carcinoma. The cells of small cell carcinoma may also express
CD99, bcl2, p53, and CEA, although these stains generally are not obtained
in a diagnostic context.
Since small cell carcinoma has spread extensively at the time of diagnosis
in most cases, treatment is generally non-surgical. Survival remains poor.
Modem chemotherapy and radiation regimens produce significant disease
remissions in the majority of patients, but relapse within a few months is
common. OverallS year survival is in the range of 5% to 10%.
Neuroendocrine carcinoma may also be mixed with non-small cell car-
cinoma. This combination is rarely if ever seen with low grade lesions, but
is seen in a small minority of cases of large cell NEC and small cell car-
cinoma. Admixed non-small cell components are rarely seen in bronchial
biopsy specimens, but are detected in up to 10% of resected small cell can-
cer cases; this discrepancy is probably related to sampling volume. When
recurrences occur after treatment of small cell carcinoma, a dominant non-
small cell component may be present; it is likely that this shift in phe-
notype represents selective survival of a previously minor admixed non-
small cell component not targeted by treatment directed against small cell
carcmoma.
D. Sarcomatoid carcinoma and related neoplasms. A subset of primary pulmonary
carcinomas have sarcoma-like features. These tumors have been given a vari-
ety of names, including carcinosarcoma and spindle cell carcinoma; sarcoma-
toid carcinoma is the currently preferred designation for all carcinomas with
sarcoma-like features. Patients with sarcomatoid carcinoma have a similar age
and smoking history as those with other forms of pulmonary carcinoma. Sar-
comatoid carcinoma, however, presents in two distinct patterns. Some cases
present as a polypoid intraluminal mass within a large central airway; these
tumors tend to be small, most likely because patients present with airway-
related symptoms early in the course of the tumor's growth. In contrast, other
patients present with a bulky peripheral mass, often with pleural and chest
wall invasion. Microscopic sections show malignant spindled tumor cells (e-
Fig. 8.54) which may transition from areas of non-small cell carcinoma. Var-
ious heterologous elements such as malignant cartilage or osteoid may also
be seen.
The major differential is with true pulmonary sarcomas and sarcomatoid
mesotheliomas. In order to diagnose sarcomatoid carcinoma, the sarcoma-like
tumor must be proven to show some evidence of an epithelial lineage. This
can be accomplished by immunostaining; reactivity for cytokeratins, EMA,
p63, or other generic carcinoma markers such as carcinoembryonic antigen
will be present in the majority of (although not all) examples of sarcomatoid
carcinoma. Electron microscopy, with identification of epithelial features such
as cell junctions, may be quite useful in this context. In many cases, exten-
sive sampling is enough to demonstrate an epithelial component by showing
transition from sarcoma-like areas to classic areas of squamous carcinoma
or adenocarcinoma; the finding of such a transition provides definitive evi-
dence as to the nature of the neoplasm. Differentiation from mesothelioma
may be difficult in that immunophentoypes may overlap; Wf1 and calre-
tinin expression would favor mesothelioma. Clinical presentation as a single
intrapulmonary mass may be a critical feature that favors sarcomatoid carci-
noma, since mesothelioma tends to present as a diffuse pleural neoplasm.
The behavior of sarcomatoid carcinomas varies with the clinical presenta-
tion. Cases presenting as a small intraluminal polypoid airway lesion have a fair
prognosis. In contrast, those cases occurring as large bulky peripheral tumors
have a poor outcome since high stage disease at presentation is common.
Cl'l••a• urc I ,,.
~if.t:]lj:IDI ll. . . . t-lllce Lu~t~ lieopiMIIu
.....
,...,.,.
Monopllosl::
DIJIMOIIIII)'
blu1lmt
dill.....
tlllnl-fcl/1111
decode
Sllall!q;
a~~~orr
,...
,...
Still
omoll
,,....... --
Jllllplll M - -
no
.,.....
--
Blptalo $~-4"1
iMlll>.
puiMonll:y deoade ~
these metastases tend to be slow growing and many patients survive with
metastatic disease for an extended period of time.
I. Sarcomas. Primary sarcomas of the lung are distinctly uncommon. Before con-
cluding that a tumor is a primary pulmonary sarcoma, primary pulmonary sar-
comatoid carcinoma and a sarcoma metastatic to the lung must be excluded. It
is, therefore, important to know details of the clinical history and radiographic
findings before labeling a lung tumor as a primary lung sarcoma. Fibrohistio-
cytic, fibroblastic, smooth muscle, and vascular sarcomas have been reported
as primary lung sarcomas, as have primary neurogenic, osteogenic, and car-
tilaginous sarcomas. Small blue cell tumors of the lung include members of
the Ewing sarcoma/primitive neuroectodermal tumor (EWS/PNET) family and
rhabdomyosarcoma. Lung sarcomas are essentially identical in appearance to
their soft tissue counterparts.
1. Synovial sarcoma is a tumor that has recently been recognized to occur in
both the lung and pleural spaces. It occurs in relatively younger adults than
do primary lung carcinomas. The tumor may show calcifications on radio-
graphs, but otherwise has few defining clinical or radiographic features
(e-Fig. 8.60). Histologic sections show features identical to synovial sarco-
mas of the soft tissue (see Chap. 46). Immunostains are tremendously help-
ful for averting a misdiagnosis; synovial sarcomas of the lung demonstrate
expression of cytokeratin and EMA staining in both the glandular and spin-
dle cell areas, and the tumor cells may also be positive for CD34 and CD99.
The differential diagnosis includes other biphasic pulmonary tumors such
as sarcomatoid carcinoma and biphasic mesothelioma. Molecular demon-
stration of a t(X;18) translocation can be a helpful aid in diagnosis. As with
synovial sarcoma arising in other locations, recurrence or metastasis may
take many years to develop, but the ultimate prognosis is poor, with the
majority of patients eventually dying of the disease.
2. Epithelioid hemangioendothelioma {EH} was originally termed intravascular
bronchioloalveolar tumor. EH presents most commonly in women of young
to middle age. Patients may be asymptomatic, or present with cough or
shortness of breath. Radiology typically shows multiple small pulmonary
nodules. Histologic sections show nodules containing pale-staining, hyaline
to myxoid stroma in which the preexisting alveolar structure is often still
apparent. The neoplastic cells are present within this stoma; they are small
and epithelioid and often contain an intracytoplasmic lumen which repre-
sents a primitive attempt at vessel formation (e-Fig. 8.61). Ultrastructural
studies show endothelial cell features such as Weibel-Palade bodies; the
cells also react with antibodies for vascular markers, including CD31 and
CD34. Although the tumor grows slowly, most patients eventually develop
progressive disease with respiratory failure.
3. Kaposi sarcoma (KS} is another vascular tumor that may involve the lung.
Pulmonary involvement occurs almost exclusively in the setting of immuno-
compromise, predominantly HIV/AIDS, and rarely in solid organ trans-
plant recipients. Pulmonary KS most often co-exists with cutaneous disease.
Radiographic studies show nodular infiltrates and a pleural effusion may
be present; symptoms include cough, fever, and hemoptysis. KS spreads via
lymphovascular routes and thus is seen along the bronchial tree, along pul-
monary vessels, and along the pleural and lobular septa. Histology shows
typical lesions of KS, with spindled cells arranged to form slit-like vascular
spaces, extravasated erythrocytes, and hemosiderin (e-Fig. 8.62). Patients
who have pulmonary KS have a poor prognosis, determined not only by the
response of KS to chemotherapy but also by the course of the underlying
HIV infection.
142 I SECTION II: THORAX
4. Pulmonary artery sarcoma is another rare but deadly lung sarcoma. Patients
tend to be middle aged or older, and typically present with shortness of
breath or signs of right-sided heart failure. Imaging studies often show
intravascular filling of the pulmonary artery trunk which may be inter-
preted as thromboembolic disease. The tumor may be situated in the main
pulmonary artery trunk or in one or both of the main right and left
artery branches; the sarcoma may extend distally into progressively smaller
branches within the lung. The histologic features of pulmonary artery
sarcoma are variable, including smooth muscle, fibrohistiocytic, endothe-
lial, and even chondroid or osteoid differentiation. The prognosis of pul-
monary artery sarcoma is poor; even in cases with complete resection,
distal recurrences within the ipsilateral lung are the rule. To date, radia-
tion and chemotherapy have not been particularly effective in treating this
sarcoma.
5. Thoracopulmonary small cell tumor, also known as Askin tumor, is now
known to be a member of the EWS/PNET family of neoplasms. This highly
malignant tumor most often occurs in children and young adults and is
typically very large at presentation; it may literally fill an entire hemitho-
rax (e-Fig. 8.63). The exact site of origin is often unclear, although most
tumors probably originate from the chest wall with secondary invasion into
the lung. The tumor consists of a classic small blue cell proliferation, grow-
ing in sheets or rosettes. Necrosis may be prominent. The tumor shows
membrane staining for CD99 as well as various neuroendocrine markers.
Most cases demonstrate a characteristic t(11;22) translocation {see Chap.
46).
J. MISCELLANEOUS NEOPLASMS
1. Pulmonary hamartoma (chondroid hamartoma) is a benign proliferation usu-
ally seen in adults. The tumor usually occurs as a solitary peripheral mass
with a radiographic appearance of a so-called "coin lesion." A minority
of cases involve the more central airways, or even the trachea. Chondroid
hamartomas may be part of a heritable syndrome in some cases; Carney
triad consists of pulmonary hamartomas, gastric stromal tumors, and extra-
adrenal pheochromocytomas. Grossly, the tumor is a well circumscribed,
nodular lesion (e-Fig. 8.64). Histologic sections disclose a mixture of benign
mesenchymal components, including cartilage, mature adipose tissue, and
smooth muscle. Bronchial type epithelium is usually present within the
lesion, although this is thought to represent entrapped tissue rather than a
true component of the proliferation. Radiographic diagnosis of this lesion
can be confidently made based on the presence of adipose tissue and calci-
fications within the cartilaginous component; for this reason, hamartomas
are often not resected.
2. Pulmonary clear cell tumor, or sugar tumor, is a unique pulmonary tumor
composed of cells with clear cytoplasm. The tumor generally arises in adults
and presents as a well circumscribed, nodular mass; most are found inci-
dentally in asymptomatic patients. The tumor usually measures <5 mm in
greatest dimension. Histologic sections show epithelioid cells with bland
nuclei and abundant clear cytoplasm (e-Fig. 8.65). The cells grow either in
nests separated by fine fibrovascular stroma or in a more sheet-like pattern.
PAS stains are positive due to abundant intracellular glycogen, as confirmed
by ultrastructural studies which may also show pre-melanosomes within the
tumor cells. Immunostains show a unique pattern of vimentin positivity, as
well as positivity with melanocyte markers such as HMB-45 and melan-A,
with expression of actin and CD117 as well.
The histogenesis of this tumor has long been debated. Most recently it
has been suggested that the tumor is a member of the family of tumors
Chapter 8 • Lung I 14 3
and plasmacytoid cells (e-Fig. 8.67) that may replace or disrupt the appear-
ance of pre-existing germinal centers. The cells also infiltrate the bronchial
mucosa with production of lymphoepitheliallesions as are seen with MAL-
Toma in other sites. The infiltrating cells are positive for CD19, CD20, and
bcl-2, but negative for CDS, CD10, CD23, and cyclin Dl.
2. Large Cell Lymphoma. Pulmonary large cell lymphoma generally presents
as a solitary mass lesion involving a single lobe of the lung. These masses
may develop central necrosis. Pulmonary large cell lymphomas are usually
either of diffuse large cell type or immunoblastic, and are of B-celllineage
(e-Fig. 8.68).
3. Lymphomatoid granulomatosis (LYG} is the name for a lung lesion now rec-
ognized to be a malignant lymphoma, now also known as angiocentric
immunoproliferative lesion (AIL). It is an Epstein-Barr virus (EBV)-related
B-cell proliferation analogous to post-transplant lymphoproliferative dis-
order or AIDS related lymphoma. In fact, patients with LYG often have
some underlying immunodeficiency. LYG usually is seen in middle aged
and older adults, and shows multiple nodules that may suggest metastatic
disease; concomitant skin and CNS involvement is quite common. Patients
have respiratory symptoms such as cough or dyspnea, or may have consti-
tutional symptoms. Microscopically, vasculocentric and angio-destructive
lymphoid infiltrates that involve all layers of the vessel are the hallmark
of LYG (e-Fig. 8.69); this vascular infiltration is hypothesized to result in
infarct-like necrosis of the lung seen in the higher grade cases. Grade 1
lesions have infiltrates composed mainly of small T-cells, plasma cells, and
histiocytes; CD20 and EBV stains show only rare atypical B-cells (<5 per
hpf). Grade 2 cases show a greater number of atypical B-cells (5-20 per
hpf). Grade 3 cases have abundant large, atypical B-cells.
4. Leukemic infiltrates. The lung may be the site of acute leukemic infiltrates
in cases of acute myelogenous leukemia (AML), representing a granulo-
cytic sarcoma or extra-medullary myeloid tumor. Infiltration of airways,
interstitial infiltrates, or nodular parenchymal masses are possible manifes-
tations. Histologic clues to a diagnosis of AML include blast-like cells and
the presence of immature eosinophilic precursors. Many cases are initially
thought to represent a large cell lymphoma; one of the first clues to the
correct diagnosis is the absence of expression of both B- or T-cell markers
in the presumed lymphomatous cells.
The lung may also show extensive involvement by chronic lympho-
cytic leukemia/small lymphocytic lymphoma. In this setting, the infiltrate
presents as nodular expansions along lymphatic routes in the lung.
5. Other hematolymphoid diseases. The lung has been the reported primary
site of a variety of hematolymphoid lesions, including Hodgkin disease,
intravascular lymphomatosis, plasmacytomas (e-Fig. 8.70), and Castle-
man disease. However, it should be reiterated that secondary involvement
of the lung in patients with advanced hematolymphoid disease is very
common, can occur with essentially all entities, and so must always be
excluded.
6. Pseudolymphoma. Small nodular lymphoid deposits in the lung have tradi-
tionally been called pseudolymphoma. However, with the advent of more
specific diagnostic techniques, many of these lesions have been shown to be
lymphomas, such as MALTomas. For lesions that can be demonstrated to
be polyclonal, the term lymphoid hyperplasia is more appropriate.
L. Metastatic tumors. It is prudent to include metastatic disease in the differential
diagnosis of every lung tumor. Metastases should be suspected when a tumor
type is encountered that would be unusual as a lung primary tumor. Also,
presentation as multiple nodules, the finding of a tumor that is predominantly
Chapter 8 • Lung I 14 5
within lymphatics or vessels, and a tumor that appears very well circumscribed
without a host stromal and inflammatory reaction should also raise suspicion
for a secondary lung tumor. Clinical history, liberal use of special stains, and
radiologic consultation can all be used to evaluate the possibility of metastatic
disease.
C. Suspicious for malignancy. This diagnosis is rendered when rare malignant cells
are present, but the quantity is insufficient for a definitive diagnosis of malig-
nancy. This diagnosis usually prompts a repeat diagnostic procedure before
definitive surgical treatment.
D. Positive for malignancy. This diagnosis is rendered when both the quality and
quantity of malignant cells are sufficient for an unequivocal diagnosis of malig-
nancy. Diagnoses of malignancy are best considered a function of both quality
and quantity of atypical cells; either rare markedly atypical cells or an abundance
of only minimally atypical cells can suggest malignancy.
IV. NON-NEOPLASTIC CONDITIONS
A. Infections
1. Viral. The radiologic correlates to viral infection typically include diffuse
pulmonary infiltrates. Cytopathologically, viral changes are best appreciated
on bronchial washing and bronchioloalveolar lavage specimens. The viral
cytopathic changes seen in Herpes virus infections include multinucleate and
single infected cells that have large nuclei with clear to faintly basophilic cen-
ters and peripheralized chromatin; the nuclei are molded with one another.
CMV infected cells have both nuclear and cytoplasmic inclusions that are
PAS positive. Adenovirus, which yields a typical "smudged" appearance on
histologic section, shows a polygonal nuclear inclusion and multinucleate
cells. Measles shows a characteristic multinucleate giant cell.
2. Fungal. The chest X-ray of patients with fungal infection can exhibit either a
mass effect, multiple nodules, or, on occasion, diffuse infiltrates.
a. The cytologic features of Pneumocystis jiroveci are well described. Alve-
olar casts containing fibrin and the organisms have a bubbly or foamy
look on Papanicolaou-stained materials. Romanowsky stained prepara-
tions show a central organism and a dear coat. Methenamine silver stains
can highlight the organism, and the stain can be applied to formalin-
fixed paraffin embedded cell blocks, or to cytospins. Patients taking
prophylactic antibiotics for Pneumocystis may not have alveolar casts,
and their samples may have rare, cup shaped organisms located within
macrophages.
b. Fragments of Rhizopus or Aspergillus species can be seen in washings,
either from cavitary masses or in ABPA. In the latter, concretions of allergic
type mucin or Charcot Leyden crystals rna y be noted.
c. Histoplasma infection rarely shows detectable organisms; however, a
granulomatous reaction may be present in aspirations of pulmonary
masses or of involved lymph nodes.
d. Cryptococcus, Coccidiomycosis, and Blastomycosis can all induce solitary
masses, which can be sampled by aspiration. The yeast forms are best
appreciated on Romanowsky stains; often the organisms are clear. Again,
cell blocks, direct smears, or cytospins can be stained with traditional
techniques to better highlight the organisms.
3. Bacterial
a. The prototypical bacterial infection for which cytologic sampling is pur-
sued is tuberculosis. Infection with M. tuberculosis can yield both diffuse
infiltrates, as in military tuberculosis, or mass-like lesions, occasionally
with cavitation.
Washings or lavage may be pursued primarily for obtaining material
for culture, molecular laboratory test to detect the organism, or antibody-
mediated studies. Cytopathologically, washings or lavage fluid can show
no significant changes or may exhibit necrosis and acute inflammation.
Cavitary lesions can have secondary squamous metaplasia, which may
be atypical; for this reason, the possibility of a false positive cytology
should always be considered in the setting of a cavitary lung mass. Patients
with M. tuberculum infection can also have extensive hilar adenopathy,
Chapter 8 • Lung I 14 7
HEART
I. NORMAL ANATOMY. The normal weight of the adult heart is 300 to 350 g (male) and
250 to 300 g (female). Cardiomegaly above a critical weight of 500 g is associated
with ischemic changes (see later) and is termed cor bovinum. The normal ventricular
thickness is 0.3 to 0.5 em on the right and 1.2 to 1.5 em on the left (e-Fig. 9.1),*
measured at the base of the papillary muscles (Fig. 9.1). The heart is composed
of three layers: the epicardium (including the serous or visceral pericardium, and
the main branches of the coronary arteries), the muscular myocardium, and the
endocardium (with an ill-defined subendocardial layer that contains many Purkinje
fibers).
Microscopically, the normal myocardium is a functional syncytium of myocar-
dial fibers (cardiac myocytes) that have centrally located nuclei (e-Fig. 9.1 ). Cardiac
myocytes are a specialized form of striated muscle; faint dark eosinophilic inter-
calated discs between the myocytes form the mechanical and electrical couplings.
Numerous capillaries with sparse interstitial tissue are found between the myocar-
dial fibers (e-Fig. 9.1).
The atrioventricular valves (mitral and tricuspid) are composed of an annulus,
leaflets, chordae tendineae, and papillary muscles. The semilunar valves (aortic
and pulmonic) are composed of three cusps (each with a sinus), which meet at
the three commissures (corpora arantii, e-Fig. 9.2). Valves are relatively avascular,
and are lined by endothelial cells on a thin layer of collagen and elastic tissue on
the atriaVarterial side, a thicker layer of dense collagen on the ventricular side,
and loose myxoid connective tissue (zona spongiosa) in between. The fibrous and
spongiotic regions are normally of equal thickness (e-Fig. 9.2).
The conduction system is composed of specialized myocytes, with fewer interca-
lated discs and higher glycogen content. Masson trichrome, Verhoeff-van Gieson,
and Alcian Blue stains can be used to demonstrate the conduction system (e-Fig.
9.3 ). Exact knowledge of the topographic anatomy and correct sampling techniques
are paramount (e-Fig. 9.4).
II. GROSS EXAMINATION AND TISSUE HANDLING
A. Endomyocardial biopsies are usually taken via a right-sided cardiac catheter;
the most common indications are monitoring of heart transplant rejection, and
grading of Adriamycin toxicity. To avoid sampling errors, a minimum of three,
preferably four, samples of myocardium are recommended (e-Fig. 9.5). The tis-
sue fragments should be counted and measured during gross examination; their
color and consistency should be noted. The tissue should be placed between
foam pads or wrapped in filter paper for routine processing. Examination of
at least three levels is recommended; some laboratories keep the intervening
sections for additional stains if required to assess myocyte damage and fibrosis.
Histologically, an adequate biopsy contains at least 50% myocardium, exclud-
ing previous biopsy sites (e-Fig. 9.5). Occasional cases require fresh frozen tissue
or glutaraldehyde fixation for special techniques such as molecular diagnostics
*Aile-figures are available online via the Solution Site Image Bank.
149
Coronary Arteries - Bypasses - Sectioning
B
LAD iii anterior
D RCA LVT
Figure 9.1 Coronary arteries, bypasses, sectioning of the ventricles. (A) The left coro-
nary artery branches into LCX (left circumtlex) and LAD (left anrerior descending),
the latter supplies the anterior septum via SP (septal perforators). The RCA (right
coronary arrery) supplies the AV node (not shown), branches into the AMB (acute
marginal branch) and the PD (posterior descending artery). The origin of the PD
derermines the distribution type (right vs. left). Examples of ACVB (aorta-coronary
venous bypass) and UMA (left internal mammary artery) grafts are displayed in gray.
(B) Slice from section illustrated in A. The myocardium displays the supplying arteries
for mapping of myocardial infarctions. The left ventricular thiclcness (LVT) is mea-
sured on the level of the anterior papillary muscle. The large septal square illustrates a
section to determine myocyte disarray. The small rectangle close to the right ventricle
(RV) illustrates a myomectomy specimen, which are sectioned perpendicular to the
endocardial surface; preferred is horizontal (a) or vertical (b).
150
Chapter 9 • Cardiovascular System I 15 1
Short-Axis Method
Setting: Virtually any heart disease, infarction (territories,
see Fig. 9.1)
Method: Place diaphragmatic side of the heart on paper
towel and perform firm cuts parallel to the atrio-
ventricular groove (Fig. 9.1). Continue "slicing"
until the left anterior papillary muscle is visible.
Then continue with Inflow-Outflow Method (A).
c
Window Method
Setting: Previous heart surgery. display of known
malformation
Method: Dissect coronary vessels
Display ventricles. outflow tract(s)
atria, foramen ovale or valves according to
underlying anatomy/findings.
·'l'
lJ '- E Four-Chamber Cut
Setting: Cardiomyopathy
~ethod: Place diaphragmatic side of the
hgart on paper tow€11. P€1rform one
firm cut in plane displayed, opening
both atria and ventricles.
Primary malignancies of the heart are very rare; involvement of the heart by a
malignancy is far more likely to represent metastasis by lung carcinoma, breast
carcinoma, melanoma, lymphoma, leukemia, renal cell carcinoma, and choriocar-
cinoma. In cases of metastatic spread to the heart, the pericardium is often involved.
A. Cardiac myxoma is the most common primary tumor of the heart. In the spo-
radic form, the tumor typically occurs in middle-aged women, and is grossly
a spherical, soft gray-white, gelatinous, lobulated tumor 1 to 10 em in maxi-
mal dimension, typically attached by a stalk to the left atrium near the fossa
ovalis (e-Fig. 9.22). In familial cases (e.g., Carney complex, NAME syndrome
[Nevi, Atrial myxoma, Myxoid neurofibroma, and Ephelides], or LAMB syn-
drome [Lentigines, Atrial myxomas, Mucocutaneous myxomas, and Blue nevi]),
the mean age of patients is mid-20s, and the tumor is more often attached to
the right atrium or is multicentric. Microscopically, myxomas consist of plump
spindled or stellate cells in abundant loose myxoid stroma (e-Fig. 9.22). Het-
erologous elements including cartilage, foci of ossification (petrified myxoma),
or gland formation (glandular myxoma) can be seen, but have no prognostic
significance (e-Fig. 9.22).
B. Papillary fibroelastoma occurs typically on the ventricular surface of the semilu-
nar valves or the atrial surface of the AV valves. The tumor accounts for 75% of
all valvular tumors, and can be up to 7 em in greatest dimension. In children, the
right side is predominantly affected. The branching avascular papillae are com-
posed of fibroelastic myxoid stroma and are lined by hyperplastic endothelium
(e-Fig 9.22).
C. Lipomas typically have a subendocardial location in the left ventricle.
D. Rhabdomyoma presents as a single (10% of cases) or multiple (90% of cases)
well-circumscribed gray-white firm myocardial nodule up to 6 em in size that
often protrudes into the ventricle. The tumor is often discovered in the first year
oflife, and is the most common cardiac tumor in the pediatric age group. Patients
usually present with heart failure or arrhythmias. Microscopically, the tumor is
composed of mixtures of round and polygonal cells with glycogen-rich vacuoles
(e-Fig. 9.22) that are separated by strands of cytoplasm radiating from the center
of the cell (the so-called "spider-cells"). Rhabdomyoma is mitotically inactive,
noninvasive, and nonmetastasizing; some tumors even regress spontaneously
after the first year of life. The tumor is thought to be hamartomatous and is
associated with tuberous sclerosis. Rhabdomyoma cells are immunopositive for
vimentin, desmin, actin, and myoglobin; focal HMB45 positive cells can be
present.
E. Intramural cardiac fibroma usually occurs as a single, white, rubbery lesion
(e-Fig. 9.22). The tumor cells are typically immunopositive for vimentin and
smooth muscle actin, indicating myofibroblastic origin; immunoreactivity for
the muscle-specific markers desmin and myoD1 is absent.
F. Other benign tumors include mesotheliaVmonocytic incidental cardiac excres-
cences (also known as cardiac MICE; e-Fig. 9.21), calcified amorphous tumor
of the heart (also known as cardiac CAT), lipomatous hypertrophy of the atrial
septum, mesothelioma of the atrioventricular node, adenomatoid tumor, epithe-
lioid or histiocytoid hemangioma, paraganglioma (extra-adrenal pheochromo-
cytoma), schwannoma, and granular cell tumor.
G. Angiosarcoma is the most common primary malignant tumor of the heart. It
typically involves the right atrium as a large mass with intracavitary extension,
and may also infiltrate the myocardium. Primary angiosarcoma of the heart is
typically more poorly differentiated than elsewhere (e-Fig. 9.22). Other rare pri-
mary cardiac sarcomas include Kaposi sarcoma, leiomyosarcoma, liposarcoma,
and rhabdomyosarcoma.
H. Carcinoid heart disease, seen in "'50% of patients with carcinoid syndrome, typ-
ically affects the heart's right side, particularly the ventricular outflow tract and
Chapter 9 • Cardiovascular System I 1 59
processing that simulate contraction band necrosis (e-Fig. 9.19) must not be
confused with true ischemic injury.
E. Infection and lymphoproliferative disorders are the two other nonrejection find-
ings in biopsies, characterized by diffuse infiltration by small to medium-
sized lymphocytes in a pattern resembling rejection(] Heart Lung Transplant.
2005;24:1710).
F. The so-called transplant arteriopathy or cardiac allograft vasculopathy is char-
acteristic of chronic rejection. It features concentric luminal narrowing of small
vessels by intimal thickening and medial proliferation with relative preservation
of the internal elastic lamina, a pattern thought to represent an accelerated fonn
of atherosclerosis. In cases with complete vascular obstruction ischemic damage
can be found, although ischemic events are clinically silent due to the lack of
cardiac reinnervation after transplantation.
VESSELS
I. NORMAL ANATOMY. The luminal endothelial cell layer defines vessels and arteries
have three layers (e-Fig. 9.20): the intima (composed of the endothelium, internal
elastic lamella, and subendothelial connective tissue), media (smooth muscle), and
adventitia (connective tissue). Venous vessels have the same three layers, but a
thinner media and a thicker adventitia.
Endothelial cells are characterized by immunoreactivity for CD34, CD31,
vimentin, endothelin, and von Willebrand factor. Endothelium also stains for Fac-
tor VIII-related antigen and Ulex europaeus I lectin, both stronger in blood vessels
in comparison to lymphatic vessels. The smooth muscle cells of the media express
desmin. Depending on the anatomic site, pericytes and smooth muscle or glomus
cells are located along the outside of the vessel; these cells show immunoreactivity
for actin, vimentin, and myosin.
The size of arterial vessels is typically defined in relation to vessels in the kidney.
The aorta is categorized as a large artery, the renal and lobar arteries as medium-
sized arteries, and the arcuate and interlobular arteries as small arteries (Fig. 9.4).
The next smallest arterial vessels, arterioles, are defined by a media that has two
to five layers of smooth muscle cells, or as having a luminal radius that equals the
wall thickness.
II. GROSS EXAMINATION AND TISSUE HANDLING. Temporal artery biopsies are typically
about 2 to 3 em long. Because arteritis can have a patchy distribution with the so-
called skip areas (see later), proper tissue handling is essential to ensure a maximum
diagnostic yield from the biopsy. The external aspect of the vessel should be inked
(which is used to ensure that the microscopic sections include the entire wall);
the vessel should then be serially sectioned at 3-mm intervals. Mter processing,
embedding of the vessel segments should result in tissue sections with complete
ring-like profiles that have an inked external surface. At least three levels should
be examined. Orienting the vessel segments in agar before processing (Ann Diagn
Pathol. 2001;5:107) is a simple way to ensure that proper orientation is achieved
during embedding.
Although embolectomy specimens are easy to gross, the submission of all tissues
can have tremendous clinical impact, because the pathologist may ascertain the
exact source of an embolus (e.g., endocarditis, atrial myxoma) in a minute piece of
tissue.
Ill. DIAGNOSTIC FEATURES OF COMMON DISEASES. Vascular diseases affect all organs
and contribute to the histopathologic presentation of a variety of diseases.
A. Vasculitides. Vasculitis is a noninfectious inflammatory disease of the vessel
wall and surrounding tissue. The Chapel Hill classification system for vasculi-
tis based on the size of the vessels (Fig. 9.4) is widely used (Arthritis Rheum.
1994;3 7:187).
182 I SECTION II: THORAX
@ 0 D D
Large
Medium
Small
910-40y TAKAYASU'S
>50y&mm/h GCA ="temporal arteritis"
PAN; KD
WG; MPA; CSS
CG; CLV; HSP
Figure 9.4 Overview of the vascular tree and typical vasculitides. TA = Takayasu•s arteritis;
GCA = giant cell arteritis = remporal arteritis; PAN = Panarteritis nodose; KD = Kawasaki
disease; WG = Wegener's granulomatosis; MPA = microscopic polyangii.tis; CSS = Chw:g-Strauss
syndrome; CG = cryoglobulemia; CLV = cutaneous leucocytodastic vasculitis; HSP = Henoch-
Schonlein Purpura. Boldface type indicates ANCA-positi.ve. See also e-Fig. 9.20 and e-Fig. 9.29.
I. GROSS ANATOMY. The mediastinum is located in the thoracic cavity and is gen-
erally divided into superior, anteriot; middle, and posterior compartments and is
bounded by the pleura laterally. Generally, the first rib defines its superior limit
and the diaphragm its inferior border. The sternum, ribs, and thoracic vertebrae
(T1 through T11-12) constitute the skeletal confines of the mediastinum. The thy-
mus, heart and great vessels, lungs, and esophagus are among the most obvious
organs which occupy the anterior (thymus), middle (heart), and posterior (esopha-
gus and aorta) mediastinum. The aortic arch and the proximal segment of the aorta
(ascending and proximal aorta) are located in the superior mediastinum, which is
bounded by the manubrium sterni anteriorly and thoracic vertebrae 1 through 4.
The embryological aspects of the mediastinum are basically those of the organs
and structures which occupy this compartment.
The definition of the mediastinum relates to the structures and organs with
observable pathology on imaging studies and the associated differential diagnosis.
For instance, the anterior mediastinum is the site of the thymus with its varied asso-
ciated pathology from Hodgkin and non-Hodgkin lymphoma (NHL), to thymoma,
to germ cell neoplasms. The pathology of the posterior mediastinum is dominated
by a variety of neurogenic neoplasms and bronchoenteric developmental cysts.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION
A. Fine needle aspiration {FNA) biopsy. FNA is generally performed as an image-
guided or endoscopically directed procedure on suspected pathology in the ante-
rior or middle mediastinum. These specimens are processed in the same manner
as other FNA specimens, and are commonly examined for adequacy so that the
results can be transmitted contemporaneously with the procedure. Given the
broad range of pathologic processes in the anterior and middle mediastinum,
an advanced level of experience is recommended for FNA interpretation of spec-
imens from these sites.
One of the more common specimens is a lymph node with the differential
diagnosis of an infectious and/or granulomatous process, metastasis, or lym-
phoma. Metastasis, usually a carcinoma of the lung or elsewhere (most often
squamous cell carcinoma of the head and neck, papillary thyroid carcinoma,
or renal cell carcinoma), accounts for over 50% of the diagnoses. NHL (lym-
phoblastic lymphoma and mediastinal large B-celllymphoma) and Hodgkin
lymphoma (HL) are the most common primary malignant neoplasms of the
mediastinum; howevet; while strong suspicion about an NHL can be voiced in
the case of a lymphoblastic lymphoma, both the large B-celllymphoma and HL
of the nodular sclerosis subtype have a considerable fibrous component, which
may complicate the ability to obtain a sufficiently cellular FNA specimen for
diagnosis.
Biopsy. Tissue samplings from the anterior mediastinum are generally small
(<1 em) and often consist of multiple fragments which have been obtained
via mediastinoscopy. These specimens, commonly intrathoracic lymph nodes,
are submitted for intraoperative frozen section consultation to ascertain their
metastatic status for purposes of operability of a nonsmall cell carcinoma of the
lung. Often biopsies that show no evidence of malignancy contain granulomas
in varying stages of activity and type (including the so-called naked granulomas
of sarcoidosis), or simply a carpet of pigmented macrophages.
165
166 I SECTION II: THORAX
*All e-figures are available online via the Solution Site Image Bank.
,•• I SECTION lit THORAX
...... ...,,..•
M~ill1lt/lnoma
rndg)laney
Motal>lldc d\ll'nomo lllolloolhiii.IIIIIIOioa.,.
M-plelllyiMna ~
Scli!n>Uiil dl)morna lk>illftnphomo
Upollbrao611noma T-eollftnfilome
Th)1nlc ....:!noma (lndudhiJ-• Hodt<Jn lftnpk..,.. oflho modlldnurn
epllldlal tumo1> ol 11>o tlljmusl "'G"'f"""'• - · Hodpln and
$qUIII'lOII4 col CIII'Ci1101110 non.fol~ t~mpllornaa
8t.llllold eare!l"'l:r'''':::l
Mu...ptlomtold c.a-• Hlllloq1li: •nd-..ntumo,.
Mll>lol:l ..rcomo end -rnd!IIIY
~~corcln~m~ llCIIa ln'!lliol:lleukolrio
s.-tllld ..- . (catl....rcoma)
car 0D1 (;:lfdnOI'l'lia - ....1111-tftla.,_n
AdllllOCI.....,mo Mlllolfl•
PIIDiotYodo,_l1101110 Thymol_.,rn•
Oo.....,rna \Oiih l!IS.I!Il tran..,,.,.,. Ll!>omo of111o mod~~a~m
...,_,..,... ollho modlldnurn
Wtii-<!III!Willl8d nM~roendoalne
..,.,..,.... (CRindd lllmo!O)
Sdllluy tbi'OUI; 111,....
~W'l:OMI
-r,plei)OIIdndd
VUcullir n...,~a~ms
A~CIRttold
Rhe.bclorn)IOWII'OOme
Pl>otit dl'lllll1111otod neu-.doalno
....,,....,.. ~oloU>llimoto
~ Clll! nllllLiroendoalne a.rdn~ 1\UI'rOI'O ot Clio DOIII>herd 110rvoo
Smd ail cwc!l"'a''\f, MC.rmendocm.&~ &ne-nrlltla . , _
Und!l!'--me [doplo tum,.. al lire U.,..uo
Cllrn~nod lll)mlc eplllloltolll.mo~>, [doplo llljl1>ll tumors
lncll<ln1 110u100Moeltto «<cl""""" Edot>lo DOlo11!l!'old111nore
1om ealt" - (11:00 aftla••ltnH
GCTa ol one llilli>ll!ll<ai!JPe (purt CCTI)
..........to..,_. ...-.
Mlllall••
s.m-.ome
E'lnbr!l>nll Clldnomo
1'10 I SECTION lit THORAX
''/.I,! I J (.!)1 WltO Clullll:atloft of~ (A. Bl, Ill. Bs. All lid C .,m.)
r.,. 'li.Tallll 'JI.IIwlllilla llla•~:etlc ......
"
Ill
a
I&
10
45
htlln~ma ..ko -I'll,
Si>lndlo" IM>Id e*">llol .... \01111 <lffllso«
110 tr'rnl>hooYioo
R....mllleo norrnll~ua..tii-W111r.. or
lrnrnollr"' tlryrnr>qW and ...,!Wid .piiM!III aolls,
wl!lar-..H....,.oorpusdos
82 28 70 Lobirlol<>lll'll' potf&ONIOIIfii\Oiol cdlueperalodllf
lmmotuR> T.Jtm~
83 II 8S Lobirlol "' 1811!> potf&ol\01 eollllelol <Olio ... ~ ""' •
mlrlmll tr'rnP~ ..,.,_-of mild
oplllol~lftlplrl rn11 ,.ba tile poalbll~ Ill' lll)ml:
Cll't!noma
AB 31 Lobirlol- mbo!d ptdlamsof~poA 01mJII>oclta pmr)
01\d IY1>e B O!m~ rtoill p01tomo..tllornol
Glindledto oodd oiiiPOd 6lill\oli!l coli>; OWIOI(
c 5
---·-""'""""·
ond B oq~ '''"'"""'tid or OM
...
~i(J:]I Jt.t.JI P&*lotk l)pelofNuoblutfc lumen
l.lndll!'ellllldotod N8 IUFl
~~~~~~- ... ,...
lt!h I1IJd& moi!NM ""hd ¢Ill ,.,...,101111
hlltl MIO 01\d need f>or l'nmii\ONot-
odlonllll!ytod-fl'llmotller
molpnt round colllumon
Pcorl)'diiii!Willl8d NB (UF or FH on- MllpntcollumdoriNn uhdllll,.._
of qDtnd MKU NB, with nsutonb,...JY p10c:o:naa.. vartetM
r-loWo<hW. MIO.IIId ........,of
""""l!lllti>U>orsa-ntan oaomo
Dlfll.o6or I!Dimbd p!Wionauro-.. lndlotlullMm orlobu~rfoel ofnouro-
(FH) will promlnonl-bfllilly-
Pl',ljbn coli dlffMMrtlo!Son, and
1101.10mlllouutn•ne.
Nodular ON ruHJ 0111fnct nodLieo<>l I>OO!t;dl!lllr-
....,_101111 hWI « l:>w MIQ In a
~IM.
-~ngp!Wtonouroma !FHJ M.,_plefod<>l-ln •• Cllfl.....,
mowropl\lllonouromo
"""''" Pftli~urorne (fHJ A-..oof1101.10-
1'J& I SECTION lit THORAX
--<)01
PonerM11c: ~
MLIIIIIIn (H-o qol
CyodcadenomalOidtnnor
PlelraP<J-I'f bluumo (t,'pO I or 10
C\OIIt-
I. INTRODUCTION. The serosal membranes are derived from the mesoderm, and form
the visceral and parietal surfaces of the pleural cavity, peritoneal cavity, peri-
cardium, and tunica vaginalis testis. Histologically, the serosal membranes consist
of a single layer of flat mesothelial cells that rest on a basement membrane, below
which is a poorly delimited connective tissue layer. The parietal surfaces of the
serosal membranes are perforated by numerous narrow stomas, the so-called lym-
phatic lacuna, that connect with the extensive lymphatic plexus which drains the
enclosed cavities. Under an electron microscope, mesothelial cells show character-
istic long, slender surface microvilli; their demonstration can be used to support a
mesothelial origin for a neoplasm that is indeterminate by other histopathologic
methods.
II. SPECIMEN PROCESSING
A. Biopsy samples, from procedures performed for diagnosis or in the context
of staging procedures, are usually small tissue fragments in the range of 1
to 5 mm in maximal dimension. Detailed gross descriptions are unnecessary,
although documentation of the number and size of the fragments is important
to ensure that they are adequately represented on the slides. The tissue should
be submitted in its entirety, and three hematoxylin and eosin (H&E) stained
levels should be examined microscopically.
B. Excision specimens, from procedures performed for benign or malignant dis-
eases, include tissue from pleural decortication procedures (stripping proce-
dures to remove thick visceral pleural peels that encase the lung and decrease
ventilatory function), debulking procedures, and resections. The aggregate
size of the tissue should be described, as well as its color and texture. The pres-
ence of gross lesions should also be documented. Gross abnormalities should
be thoroughly sampled. When no gross lesions are identified, as a general rule,
at least one section per centimeter of aggregate tissue should be submitted for
microscopic examination.
Ill. NONNEOPLASTIC LESIONS OF THE SEROSAL MEMBRANES
A. Acute serositis
1. Acute pleuritis is usually infectious in origin and is most commonly associ-
ated with pneumonia. Gram-positive bacteria are most commonly isolated,
although a wide variety of pathogens can be responsible. Spontaneous bac-
terial pleuritis occurs occasionally in patients who have cirrhosis. Autoim-
mune pleuritis, although sterile, can produce clinical and pathologic findings
that resemble infectious pleuritis.
2. Acute peritonitis is usually associated with a perforated viscus. If it is due to
gastric, biliary, or pancreatic rupture, it has a chemical etiology; on the other
hand, if it is due to intestinal rupture, it has a bacterial etiology. Spontaneous
bacterial peritonitis also occurs, usually in children, immunocompromised
patients, or patients with cirrhosis. Localized acute peritonitis is a feature
of pelvic inflammatory disease.
3. Acute pericarditis can have an infectious etiology or can be a manifestation
of autoimmune disease.
B. Granulomatous serositis can present in a number of different patterns; stud-
ding of the serosa by innumerable small nodules can be especially worrisome
clinically for disseminated tumor.
177
178 I SECTION II: THORAX
*All e-figures are available online via the Solution Site Image Bank.
Chapter 11 • Serosal Membranes I 17 9
...........
'' l.t! I Jf!'l WltO Hld*lfc CIIMIItlcl1fouf r - of 1M"'-
_..,..._""
~- tlfnplloml
""...."""""
Ei>llll ...k:l hern~ ..loi!\11
$fllowlallll......
Momphoolc
Blphoole
Solllly flbrouoiiJmOf
ColcfJ!nltnnor of1ho Dlou10
Doom01>la* ..,.II rouhd "'!ltnnor
~ 11 • Set'lsal ~braNa I ,.a
,,(,I ]IJffll l'NIIhltfi!IIClaultlcatloeforPIIMnl IIIMOv.r.-
''k
1X
•F>·•.,PB••rlllmor<~nnotllo-IOCI
m
ro No-""'"' prfmery tumOf
TI Tumor-lpoliiWII~Iplturnlllor-utmed-.lplourunrl
n.
wlllor-ut<lrrplrrqmiillc
No ln ...Mmrli!l ofllle _ , pleura
J>ilu'"' """"""'"'t
nb 1\rrnor IIIIo ln,..Mng111a- pleura
'12 1\Jmor i'MM'll eocil of file IDOIIDnil J>ilu!OIOUrfacao (llOJ1oW, rnodlool:lnol,
dlol>hi'III)M1Ic. ofld ..w:.n1 lliourallwttlllrtloaotonoolelle fr:iklollnr.:
-1.-ont <il dlirplrrqmiilli: muoclo
- o f tumor from -•lplturalmo file undllfl)tla
p,~l'lnCtl)'m&
"""""'"Y
l.ocliitt ..t.lriiO&d lrlrt ~tt .-tAo tumOf. T11110r l'r\lolllrw d of111o
IDO!Ia1Bral plelel ou- (llOJ1oW. rnodlaafnol. d~r:llrwri011e. anrl"'-ol
ploura)- at lout ono <illllo-..,
-lrrvot.orrrnont <iltha-......C-
- IriD tilt medluUnol Ill
-&aaJy, oompiMit, r-bla loa.itoltumor -..ng lmo file toft- <II
file-wolf
T4
-Non1r"""'urolln,..Miment of file I>OIIcordlum
lpsi-..J plrro.,..ltu- (pcrlotal, modlirllnol, d~plr-illllc,
ploura) -~~- ono <illllrr-..,
ilnrl-
l.ocliif1 &IMnood lor:hnll:tly unr....:talib timor. Tumor ln"'Mn,t el al 11\o
-OI'IICI
-n
-lltaoooelated rlbdootruc11on
-OI'oet 1rallod ~ phr0111nallc -..!on of 11./mor to file l>flltl:nelm
-OI'oct - · <11 tum"' 1o ttro ..mnr~rr~a..r plollll
-OI'oct <II tum Of lo mod-al 0f111N
of brm,..lnto thrr •liN
-Tumor 6ibndlna throu&!l ID the lntolrnlllturfeoo of the prut:ilnl~m wtt11 or
-~~~· pe!bRiol.-n ortlmlr lnlrolf!rcllle myoaudllm
flltllonrl .... -011
r« R.....,.l !lmltr nodol..nnotl"' no1111id
NO No r"'l)oniiillympll noclo Oll!tiOIIOIIb
Nl Mo-lntllo lpoli111nolllronch0j)Umorwyor lil~r IJmph naiM
N2 MlltM:b : aIn the subarhtl or the mediutfnllll)mph ncdl!llllncMtna the
11JOiflllnlintrlmal mammooy •nd perldhph-lc nodot
~ Moll,..,..,. 11\o OOIIII'alrila!OI rnodloodnll, OOII!I'illrila!OII'rtemll mammOIY.
IJ>OIIa1Bral « -ldolraiOUP!OcilwbJ~r IJmfil-
Mllln•n•••••
MX Olllant - ·<1nnol llo ..,.,.id
MO Nodll1ani-
ftql_,.
MI fllllant m-llil PI-'
Slqir 1 n NO MO
$Qir lA na NO MO
$QIIS Tib NO MO
5lqo II T2 NO MO
51qo 111 n. T2 Nl MO
nf 12 N2 MO
l3 NO, Nl,N2 MO
Slqir ti T4 AltyN MO
AeyT NS MO
AeyT AnyN Ml
184 I SECTION II: THORAX
cytoplasm, although in some cases the cells have more anaplastic fea-
tures. Architecturally, sheet-like, microglandular (adenomatoid), and
tubulopapillary patterns are common (e-Figs. 11.6 and 11.7). Psam-
moma bodies are occasionally encountered.
b. Sarcomatoid mesothelioma is composed of spindle cells that have a
haphazard distribution (e-Fig. 11.8). Some cases resemble fibrosar-
coma, others have a pattern that resembles undifferentiated pleomor-
phic sarcoma. Immunohistochemically, sarcomatoid mesothelioma is
less likely to express cytokeratin 5/6; areas of chondrosarcomatous
or osteosarcomatous differentiation may show positive staining for
actin, desmin, vimentin, and/or 5100. Many cases retain expression
of calretinin. The potential overlap of histologic and immunohisto-
logic features of sarcomatoid mesothelioma with sarcomatoid carci-
noma and various soft tissue sarcomas highlights the necessity for
correlation with clinical and radiographic findings; a solitary mass
should raise concern for another nonmesothelial sarcomatoid pro-
cess. Sarcomatoid mesothelioma is generally thought to have a more
aggressive course than the epithelioid varieties, and so patients with
this subtype are generally excluded from consideration for surgical
therapy.
c. Desmoplastic mesothelioma. By definition, this type of sarcomatoid
mesothelioma consists of scattered atypical cells in a storiform or non-
specific pattern in >50% of the tumor, set in a dense collagenous back-
ground. This subtype is the most likely to be misdiagnosed as organizing
pleuritis in small biopsy specimens.
d. Biphasic mesothelioma. This subtype contains a combination of the other
patterns, in most cases a combination of the epithelioid and sarcomatous
patterns (e-Fig. 11.9). By definition, each component should comprise at
least 10% of the tumor.
2. Well-differentiated papillary mesothelioma is a rare type of mesothelioma
that occurs in a wide range of patients, although most patients are elderly.
An association with asbestos exposure has not been established. Patients
usually present with dyspnea or a recurrent pleural effusion, but rarely with
chest pain. At presentation, the tumor may be either solitary and localized,
multifocal, or widespread.
Microscopically, the tumor features fibrovascular cores (that often have
a myxoid stroma) covered by a single layer of bland, cuboidal-to-flattened
mesothelial cells. Focal areas of limited stromal invasion may be present. In
cases with widespread invasion, DMM with papillary architecture must be
excluded. The distinction is important, since when strictly defined, well-
differentiated papillary mesothelioma has an indolent course with pro-
longed patient survival.
3. Localized malignant mesothelioma is a circumscribed nodular lesion attached
to the parietal or visceral pleura that is usually < 10 em in the greatest dimen-
sion. It is usually discovered incidentally on imaging studies. Microscopi-
cally, the tumor has architectural patterns that are identical to DMM. Some
cases are cured by surgical excision. It is interesting to note that recurrent
tumors often metastasize in a pattern more typical of sarcomas, without
spread along the pleura surfaces.
B. Mesenchymal
1. Epithelioid hemangioendothelioma (termed intravascular bronchioloalveolar
tumor when it arises in the lung) is a low-grade malignant neoplasm of
endothelial cells that can develop at virtually any anatomic site. Primary
cases arising from the serosal surfaces occur, albeit rarely (lnt] Surg Pathol.
2006;14:257).
Chapter 11 • Serosal Membranes I 185
...........
'' 1.1'! I J f!ll WltO Hld*lfc CIIMIItlcl1fon •f r - of 1M~
--·-
AIJ.nGMmkf bJMOf
the same histologic types as tumors arising from the pleura. The development
of pericardia! mesothelioma is also associated with asbestos exposure.
B. Germ cell tumors. Intrapericardial germ cell tumors are rare, but occur over
a wide age range, from neonates to the elderly. Intrauterine presentations are
increasingly being recognized in second and third trimester gestations due to the
widespread use of prenatal ultrasound examination. Germ cell tumors of the
pericardium, as with extragonadal germ cell tumors at other sites, are thought
to arise from germ cells that lodge in midline structures early in embryogenesis
along the normal route of migration from the yolk sack to the gonad.
Teratomas account for the vast majority of pericardia! germ cell tumors.
Over 75% of the cases occur in children under the age of 15 years. Teratomas
can achieve remarkable sizes, up to 15 em in the greatest dimension. Grossly,
they usually have a lobulated, smooth surface. Histologically, the vast majority
are mature teratomas that resemble their counterparts arising in the gonads or
mediastinum, in which case the differential diagnosis includes a bronchogenic
cyst. While teratomas are benign, tumors that contain other germ cell elements
(e.g., embryonal carcinoma, choriocarcinoma, endodermal sinus tumor) are
malignant. They are exceedingly rare, and most cases arise in adults.
C. Secondary neoplasms. Metastases are the most common tumor of the peri-
cardium. In a significant percentage of cases, a biopsy (often performed to
establish the cause of pericarditis or life-threatening tamponade), provides the
first evidence that the patient has a malignancy. The most common primary
tumors that metastasize to the pericardium, in decreasing order of frequency,
are carcinoma and adenocarcinoma of the lung, breast, and thyroid; lym-
phoma; and sarcoma. Although lymphatic or hematogenous spread is the most
common route of involvement, direct extension (e.g., by pleural mesothelioma
or malignant thymoma) also occurs.
VIII. MALIGNANT TUNICA VAGINALIS TESTIS NEOPLASMS
A. DMM occasionally arises from the tunica, and rarely, even from hernia sacs.
Grossly, the tumor forms nodules or papillary excrescences. Microscopically,
the most common pattern consists of a prominent papillary architecture with
associated tubular and solid areas.
B. Epithelial tumors of Mullerian type have been reported.
C. Secondary neoplasms. The tunica, as well as the lining of hernia sacs, can be
involved by metastatic carcinoma. In some cases, the involvement of the tunica
or hernia sac is the first manifestation of metastatic disease.
SUGGESTED READINGS
Battifora H, McCaughey WTE. Tumors of the Serosal Membranes. Atlas of Tumor Pathology, 3rd
Series, Fascicle 15. Washington, DC: Armed Forces Institute of Pathology; 1995.
Clement PB. Diseases of the peritoneum. In: Kurman RJ, ed. Blaustein's Pathology of the Female
Genital Tract, 5th ed. New York: Springer; 2002.
Tavassoli FA, Devilee P, eds. Pathology and Genetics of Tumors of the Breast and Female Genital
Organs. World Health Organization Classification of Tumors. Lyon: IARC Press; 2003.
Travis WD, Brambilla E, Miiller-Hermelink HK, et al. eds. Pathology and Genetics of Tumors of
the Lung, Pleura, Thymus and Heart. World Health Organization Classification of Tumors.
Lyon: IARC Press; 2004.
SECTION Ill
Gl Tract
The Esophagus
Dan ielle H. Carpenter and Elizabeth M. Brunt
I. NORMAL ANATOMY. The esophagus, a tubular structure that connects the pharynx to
the stomach, is composed of cervical, thoracic, and abdominal segments. It begins
at the level of the cricoid cartilage and ends at the gastroesophageal junction (GEJ).
The GEJ is the junction of the tubular esophagus and the saccular stomach, and
is distinct from the squamocolumnar junction.
By endoscopy, the esophagus is measured beginning 16 em distal to the incisors
and extends 35 to 40 em to the GEJ. Precise anatomic location within the esoph-
agus is a significant parameter in the differential diagnoses of various pathologic
processes, as well as for staging squamous carcinoma.
The esophageal mucosa is composed of stratified squamous epithelium that
extends distally to the squamocolumnar junction; this overlies paucicellular lam-
ina propria and is delimited by thin muscularis mucosae that have a rich network
of lymphatics (the latter allows for early metastases of relatively superficial malig-
nancies). The deeper submucosa also has a rich lymphovascular network as well as
submucosal glands connected to the lumen by ducts. The deep muscularis propria
is composed of an inner circular layer and outer longitudinal layer; the proximal
third of the muscularis is striated, the distal third is smooth muscle, and the mid-
dle third is a mixture. There is no serosal surface on the esophagus, but rather an
adventitia.
II. GROSS EXAMINATION AND TISSUE HANDLING
A. Endoscopic biopsy. The standard endoscopic biopsy consists of several small
(1 to 5 mm) unoriented pieces of mucosa with varying amounts of attached
muscularis mucosae. In some cases, the endoscopist may use "jumbo forceps"
to obtain larger fragments (4 to 8 mm); submucosa may be present in these
biopsies. All fragments are submitted and three hematoxylin and eosin (H&E)-
stained slides are examined.
B. Endoscopic mucosal resection {EMR}. EMR is a more conservative approach
than esophagectomy for resection of superficial malignant and premalignant
lesions. These en bloc resections of 1 to 2 em lesions are obtained by elevation
of the mucosa with submucosal saline injection, followed by removal of the
mucosa with variable amounts of attached submucosa. The specimens range
from 1 to 4 em in the longest dimension and up to 1 em in thickness.
EMR specimens are carefully pinned flat for fixation, all deep and radial
margins inked, and the specimen serially sectioned and submitted entirely to
assess not only the mucosal lesion, but also the deep and radial margins (Fig.
12.1). Initially, at least three H&E levels are evaluated.
190
lllli~Il111111~1jiltf ti.!li 1 1!
~
•
~
-
J~
_~~a.~~!~1ll ai~! il~ilil ;~11~1 ~-S
l!i
..i• ~ ~'S.!Iiii'·• ~-2<. l! •:ij S· Bi!'l:l
- _iiJf<l:l 0
"""'~!It! !l
K I
-~ :t;;j~ ~ !i~'f ~1-tf d.JIIIlll-..·!l lJfR~ . ll "'·
~
·J 8Jtu:;:~j] -~1iiPn~~i~ f~~~-g"' il
l IJ1::1~~1i It~i!~] Dil~i s.Jl~ ~~ ~1
-r il. ·t f~ 1h] 5=!i11t d t jlu"i ~-
\1 j cp
... ::!
I!
1f e] ;11 1..., .. -g ll,.'h 1 1141 -a
11
e l
d iii.!
IIi
192 I SECTION Ill: Gl TRACT
Proxrdimal
sh:margin
Uninvolved
stomach
Stomach
*All ewfigures are available online via the Solution Site Image Bank.
,.,. l!o.lll~llllc aao,..&tl•
More QOII1MMin c111il11111
b'la...,.Jik
M01o comnonlh adullo
SG:c More QOII1MMin mol& l<lllllitt lh ~ ooxoo
Malnovm- D)o~ HM111>urn
End._plc: nnc~~~p Sllldllrt, ~na Vwblo
lrMlfwment Entlrusopllqla DIIUI-hlaua
M""""'plcflndlna >20~hlslhpf; -plr!lc <Ooosln01>hlslllpl
rrlcllllblcoaaa
"""*"
-··
Tol>lcol
194 I SECTION Ill: Gl TRACT
b. Herpes simplex virus (HSV). The classic lesion of HSV esophagitis is shal-
low ulceration, ranging from millimeters to centimeters in diameter. Viral
cytopathic change, characterized by multinucleation, molding of nuclei,
and margination of chromatin, are seen most frequently at the edges of
the ulcer (e-Figs. 12.16 to 12.18). Immunohistochemistry (IHC) is con-
firmatory in this regard.
c. Cytomegalovirus (CMV). Focal, discrete ulceration in the distal esophagus
occurs in CMV esophagitis. Granulation tissue from the ulcer base (as
opposed to the edge in HSV) is optimal for identifying viral cytopathic
change in endothelial and stromal cells. Large eosinophilic nuclear and/or
cytoplasmic inclusions can be seen on routine H&E stained sections,
although immunostains for CMV can highlight or confirm the virus.
IV. BENIGN POLYPS
A. Fibrovascular polyps are elongate, often large ("giant fibrovascular polyp"),
pedunculated intraluminal growths that can fill the esophagus and present as
an intraoral mass if regurgitated. Histologically, these polyps are composed of
an edematous, loose stroma with a rich vascular network covered by benign
squamous mucosa.
B. Inflammatory polyps. Often related to GERD, inflammatory polyps are present
near the squamocolumnar junction and may endoscopically resemble adeno-
carcinoma. Howevet; histologically they are benign and composed of inflamed
squamous and/or foveolar mucosa.
C. Squamous papilloma. Also often related to GERD, squamous papillomas can
have a filiform appearance microscopically, if not grossly. Delicate fibrovascular
cores are covered by benign reactive squamous epithelium with varying degrees
of inflammation (e-Fig. 12.19). Although rare cases of virus-related papillomas
of the esophagus have been reported in immunocompromised patients, squa-
mous papillomas are most often seen in patients with GERD.
V. EPITHELIAL NEOPLASMS AND PRENEOPLASMS. The current World Health Orga-
nization (WHO) histologic classification of esophageal tumors is given in Table
12.2. The AJCC Staging guidelines for esophagus and esophagogastric junction
are applied to primary esophageal and esophagogastric junction mucosal tumors,
including the proximal 5 em of stomach (Table 12.3 ).
A. Squamous dysplasia. Endoscopic evidence of squamous dysplasia may be subtle
and most likely identified in association with plaque-like, mass-forming lesions.
Dysplastic squamous mucosa has cells with enlarged, hyperchromatic nuclei
and increased nuclear to cytoplasmic ratios, as well as overall dysmaturity of the
epithelium. Unlike squamous carcinoma, dysplasia is limited by the basement
membrane; however, unlike benign reactive changes, dysplasia lacks cytologic
uniformity and orderly epithelial maturation.
B. Squamous cell carcinoma (SCC) has a range of gross appearances, from ery-
thematous eroded mucosa, to plaque-like growths, to mass-forming exophytic
lesions. It is most commonly found in the middle third of the esophagus, fol-
lowed by the lower then upper thirds. By definition, neoplastic cells extend
through the basement membrane into the lamina propria ("carcinoma in situ"
is a term no longer used; it has been replaced by "high grade dysplasia")
(e-Figs.12.20 and 12.21). Depending on tumor grade, there are varying degrees
of keratin formation in SCC of the esophagus (e-Fig. 12.22), although the
various subtypes of SCC (basaloid, spindle cell, and verrucous) often seen in
other body sites can also occur in the esophagus. In addition to tumor, lymph
node, metastasis, and grading characterizations, tumor location also factors
into tumor staging (Table 12.3).
C. Barrett esophagus. A diagnosis of Barrett esophagus entails endoscopic find-
ings of salmon-colored mucosa of any length above the GEJ with correlat-
ing histologic findings of a specialized columnar epithelium with goblet cells.
a..~ u. ne. ~· I ,••
~i f.t:) I j FIJI WHO C&Mik&tJon of TurMtt of ttt.e EMtt spa
ljllhlllll ~~
Pnotnal(rnant-
Squtll'IOUII
Ihi~ l'flOI:Wo, lciH pdo
lhl~ l'flOI:Wo, ,.., ll'od&
GIIMJiu
~·J lh~ bw.,..a
~·J lh~ hWI &rid&
c-Jnom/o
$qUI/I'oOII4 col Clll'eiiiOitlO
Ad ................
Ad ...ld .,.aaeCireNn\1
NJ~ut.n'dl& oue!l"'l:::''''::l
Bm!o!d tqu.mow:oel CIIJ'dn~
M._tll11mold ..,..,..,.
Splndlo<:d ('"""mow)....,,,...,.
VeiTUCIOIIO Ct<~uo....W wcln....,.
Undiii'•IMIDd .....,-.....
-f~MJ~l{Mmt;
Nourcondccmo tumor (NET)
NET Gl (con:Nid)
NETG2
NoUI'*'dccltiO .....-..... (N[C)
LQo-coiNEC
Stnol-ooiNEC
...............
Mkld adlf\OniUI'OIII"'<b:rr,. a.rdnana
""""""""'
Rhobdo.,.,.,.....,.,.,.
Synooolol-
·-""-
" I'-
11& I SECTION llh <II lAACT
_,.
It mraalardt ...
stqoO
T
111/H~D
II
NO
II .... 1H.I1Ioft
MO I,X Nrt
............
AfiAUiftft ...
r
llllltO 111/HQD NO
II ..
Ill)
....
I.X
ltqoll
~~qoa
Tl
Tl
NO
NO
MO I,X Nrt
MO ~ Nrt ...... Tl
Tl
NG
NG
Ill) I~X
Ill)$
....
T2-a NO MO l,X ~.x T2 NO Ill) I~
llqtM ~ NO MO !,X UP110r. mid hilt lA T2 NO 111)3
~ NO MO 2-3 ~.x
T2-3 NO MO ~ UPIIOr.rnld SllpU T.:l NO III)Artl
TH Nl MO Arr! Nrt TH. Nl III)Artl
ltqoal Tl~ N2 MO Aey Nrt .....lA Tl~ N2 III)Artl
l3 Nl MO Aey Nrt T3 Nl III)Artl
T4o NO MO Aey Nrt T4a NO III)Artl
. . . . . l3 N1 MO Aey Nrt 111101a T3 N2 III)Artl
stqoa T4o Nl-2 MO Arr! Nrt 111101~ T4a Nl-2 Ill) Alti
T4b Nr1 MO Arr! Nrt T4b AAJ III)Artl
ltqol't
MPj
MPj
NS
Arr1
MO
Ml
Aey
Aey
Nrt
Nrt ....., Arr1
Arr1
NS III)Artl
Arr1 1.11 Alti
C.fllttfi IIMIIJIIIS
Nopllll'tlorcl)oi>IUI • ~if blond with in111ctnudoor polul\';allunllent
!Htp. l:I.Z. .nd IO!>Iat ....
II .at! • Nonnal alendulor ard\IIDdllre ...,...
1 -.. l'ord)opnla
(HtJ. 11.m1
-
• M/ mtllmol O)'llio9:or ardl-.relllly!Jbl-...""'
I~ 11> Ill& d•por pm!Gr\ of til• ala ndo, """",_ ot111•
...
• o,toqqc..,d . .hbotu"'l•...a11ompn>IOIIIIMHUU-nt
for unoqu"""'l d...,...a!.,.,....
• F«O)!Omple. lhoomo....,.dll1fndloll-"'
lnftomm-...ln!d , . , . , _ " " ' • and ct,olplula I'M'1
be d!lllout
• In .sane Cl!lit:l, the reilltklnshlpdl:ht twf'ICt t.nd dMper
pndoQIInotboes-duoll>lln...,U.I-"a
• o,toqqc a~Jpia: olrolllod, ponot.oliopod, and
hl'l*'Chrr.mlllc ""'*'v.tll m.;ular llUCieu rnembran""' ~
IMeet ldalt1 01\d lode ctlllll<lool: looo ol muefn
• Mid ardl-hll•llo- minimal pndulu erowdll1!
mqbo-t
• o,toq1c aiJpla, boo a! mudn, and mtllmol ~lu
c.-t>alo<olyodond ll>tlle ourfoatauo!lllllt t11o ~
Phdo elld wr.... ap-etnlior at low-· R&rnlnllooilt
oftullular -ollll&..,lon
H~ dJOI:io*' • ~ otyt>lt: ltlolll<.edlt•niiiB!<I, lt>Und-ld
(Hip.lutand h_.:hrernlllc """""""" """"""""t171111allon and lou t1
12.14) pdalty; alinOfmol m - fl.llu'• miJ b6 ..-rt
• Ardlllocturalallo- pnd'*'r c,_ln,t 101111 mtllmol
IMeMitli~Wiaml'lo r>ror>!lo; -llli'Lial\tohor>!ld. budd~
branclllrw, Ull.fflllm~ «t.'/1tll;tltJ dla1od &Iondo. >illft.nn
alrfsa&OOMrrd\
• Onlfeyll'*Ofard\_1_,,.._1,.10111&
tu-1"' ,_IIIYID d . _ HGD, tliouJitlboth are
oftan-t
• o,toq1c a~Jpia: u 111><n t1 hW!po<l)olplolll
.,_--..ttl-··""
• Arcllllocturelallo-ln......,111~ tile llolld
beoemeM memb10h0 1111o111& lomn PI'Or>l1a « mUI<Ualll
a1bl!fmn, --11>-taek,
or...,plel ll(lftq1IIIIP'O"lh!>l1twn, and lnd-aldylplullc
••II• ar srnoll ciiDllra of d)lll)!.ullc: .... In tilt limn r>roPIII;
dosm/llllalla not""""""
11a SECTION llh <II lAACT
*All e-figures areavailable online via the Solution Site Image Bank.
201
202 I SECTION Ill: Gl TRACT
Gulrlc 1->enl
pemll:lcua ........
Bocty•ncl fund111on11
"Pill' Gl bleeclna
Mainly al'lbum, or mull:tfocll
M""""'plcflndlnp Qumll:pobllb. P"'''- Qrronlo .....,11>.1.-.1
oo•b'"'~'" of fundII: pnot, rn&<plriMI. pjloflo ........
, , _ , m<CaPloolo.ll'.l4olb Hbo1t1 l>lrr.rolted
"""'ploola• ECLoel
hyperpluls
Solum p.llrln IMI EIINtad Norm.tlor""'
Tumo< ""'"""mont Qm:tlold, oclan...,..lnoma MALT ltmphoma
204 I SECTION Ill: Gl TRACT
thickness or deep) (e-Figs. 13.27 and 13.28). In iron pill gastritis, there may
be mild inflammation or erosions (e-Fig. 13.29).
8. xanthelasma or xanthoma is characterized by collections of lipid-laden foamy
macrophages in the lamina propria and is significant for its potential con-
fusion with poorly differentiated signet ring cell adenocarcinoma on biopsy
(e-Fig. 13.30). In problematic cases, the diagnosis can be clarified with lliC
stains; positivity for pan-cytokeratins and CDX2 is seen in most adenocar-
cinomas, whereas positivity for CD68 and CD163 would favor xanthoma.
IV. DIAGNOSTIC FEATURES OF COMMON GASTRIC POLYPS
A. Hyperplastic Polyps (HPs) are composed of elongated, dilated, branching foveola
in association with inflamed, edematous lamina propria. These polyps can show
surface erosions, regenerative changes, and intestinal metaplasia (e-Fig. 13.31).
In addition, foci of dysplasia or invasive carcinoma are present in ""'2% of HPs.
The likelihood of these findings increases with polyp size, especially in those > 2
em. HPs may occur in a subset of patients with Osler-Weber-Rendu syndrome.
B. Fundic gland polyps are composed of cystically dilated spaces partially lined
by parietal cells which may be hyperplastic or attenuated (e-Fig. 13.32). They
can be syndromic, where they represent the most common gastric lesion of
familial adenomatous polyposis (FAP), or sporadic. A significant proportion of
syndromic fundic gland polyps are associated with low grade or indefinite dys-
plasia. However, high-grade dysplasia and invasive carcinoma are exceedingly
rare. Sporadic fundic gland polyps are sometimes associated with PPI treatment
for gastroesophageal reflux.
C. Inflammatory fibroid polyp (IFP} is most commonly found in the gastric antrum
and consists of a submucosal collection of bland spindle cells, which are charac-
teristically CD34 positive and c-kit negative, in a background of dilated vascular
channels and mixed inflammation, which often contains abundant eosinophils
(e-Figs. 13.33 to 13.35).
D. Hamartomatous polyps can be seen in patients with Peutz-Jeghers syndrome,
juvenile polyposis, and Cowden's disease. The histologic features of many of
these polyps may be reminiscent of hyperplastic gastric polyps.
V. DIAGNOSTIC FEATURES OF COMMON NEOPLASMS OF THE STOMACH. The World
Health Organization (WHO) histologic classification of gastric tumors is given
in Table 13.2. The American Joint Committee on Cancer (AJCC) staging schema
is given in Table 13.3. Molecular tests used in the work-up of stomach specimens
are listed in Table 13.4.
A. Adenoma and dysplasia. Unlike colonic adenomas, most gastric adenomas are
not sporadic but rather arise in the setting of chronic gastric injury. Adenoma
and dysplasia are distinguished by architecture: polypoid = adenoma, flat =
dysplasia. Both can contain either low-grade (e-Fig. 13.36) or high-grade dys-
plasia (e-Fig. 13.37) defined by cytologic features and architectural complexity.
There are three types of stomach adenomas: intestinal type (e-Fig. 13.38),
resembling those in the colon and containing goblet cells and/or Paneth cells,
and two gastric types, foveolar- and pyloric-type adenomas. Foveolar-type ade-
nomas are composed of foveolar epithelial cells containing neutral mucin with
apical mucin caps, stain positively for MUC5AC, and are sometimes associated
with FAP. Pyloric gland adenomas are composed of tightly packed pyloric gland-
type tubules with a single layer of cuboidal to low columnar epithelium that
shows round nuclei and pale to eosinophilic ground glass cytoplasm, and that
stains for MUC5AC and MUC6 (e-Fig. 13.39). Pyloric gland adenomas have
been shown to occur more commonly in women and are more frequently asso-
ciated with autoimmune atrophic gastritis (Am] Surg Pathol. 2009;33:186;
Virchows Arch. 2003;442:317). Pyloric gland adenomas, as is the case with
intestinal-type adenomas, are also associated with a higher rate of dysplasia
and malignant transformation than gastric foveolar-type adenomas (Am] Surg
Pathol. 2002;26:1276).
2D& I SECTION llh <II lAACT
""'--·
~
Papll.tiJ - . . . ........
Tubullr lllonoc:an:l""""
Mucl•ouolldo•"""""omo
Poo!\'cdleotJo<Or'*>orno GftW<Iftl.,ot lt\.l <Ill COrciftOinO end OC!Ierlillllolrtol
Mlood.odonoolrm"""'
AIJeno~q.-noua ~!1"1£1'1\1
C.-•Willi !Jmplloi<lllroma (modullly corclnotl'll)
~CI~me.
Squamou• cd! Cl~me
Ulldl'l&1111111r1ed ..,,...,.m•
-ll&lljllolm<
rwr..ntloe!lrl& 111m or (N[T)
NETGI (<Jrclnold)
NETG2
rwrc.ntloe!lrl& .,.,...,.m• (NEC)
Uoii!IQIIII NEC
smoiJQOII NEC
Mlood-...u--1""""'
EC col, . .ton-.i>rodUctiJ NET
......_.........
~ocq NET (psiJ!noma)
Glom~lUh\Of
Gftftultr eel tumor
LA...,.,ma
Ploldform ftbromyoo>mo
-lelllnols!A>molt.mo<
u..._....,.
IC:Ipool-
$fllowlallll-
&rwr~awa
IK151FT.,_
''I.I IIJl !II 1'IIIMI. NOCS.. Mttlltula (1M II) S111flll kllemt fwCMII!c Clld-
''' , . . . . (!)
1X Prtr.:Yt.mor.. motboo-
r-Io-""'
111
11>
T1
ol polme I)' 111m or
eo,...,.... 1'1 stu: ~llUmOf ...,Itt!""""""' oteho laml'lo1>10DIIo
lUMOf ~ Wn!l'lll P~*~ MLUsa.lflll1t fni.IC!($!I.b0f .SUbrnl.lel:lM
Tl• 1Urn0f ~ Wn!M p~ltt Of muac:uii.N: n'liUC(),IIM
TI b TumOf ~ submUCOIII
T2 TumOf tt\lad6l rnuscuiula proprta•
l3 TumOf pon- ..- con.-llnua\OIIIIout . , _ ol -~~
DOI1iolleum or~ ocruct.n"""'
TA 1\JmOf-.o ..,...(,....ralpo-m)arad)lant_..,....
TAe 1\JMOf-..,... (......ral ...- M )
T4b TumOf-.. a.d)lconl oiNdu!'M
llillnll,..... IIOdlltOU
r« Retlollollimli!IIOdeo CO/lnot be ooomed
NO
Nl
r-Io .....I tlmlil node,._..,
!W<'I'+In 1-2 r...nlltJ'Mph node&
1t2 -olillna-6,...nlltjmph-
IG - t i l l In......, ar,.,. "'lonll tjmpll ncdos
roo -.r1s1n 7-lo t~mph · -
~ Melaeeaoioln l60frnoR>IIllbtollimPh IIOdeo
IIGrt••s•••t• (1111)
M)( Dtdlrrt met•st•• e~mot 1M a-....d
MO
'lodlsllrnt - ·
Ml llllllnt moCIIItoril
I!W-111
SlqoO 'Ill NO MO S1apiiiA T.. Nl MO
Slqo lA Tl NO MO Ta ~ MO
51qr:!IB T2 NO MO T2 IG MO
Tl Nl MO Stop 1118 T41> NO MO
$QII!A 1'3 NO MO T41> Nl MO
T2 Nl MO TAo N2 MO
Tl ~ MO 1'3 IG MO
Slqo JIB T.. NO MO S1apiiiC T41> ~ MO
l3 Nl MO T41> IG MO
T2 lt2 MO T4a IG MO
Tl ro MO S.l\l AII)'T MIN Ml
2aa I SECTION 111. 01 lAACT
10asmlltii1Sorlwlloh
""'"*
lrrnlua.mlb In eddllon lo-
lrlerll!y pctienlnllll HNPCC anclar.1<ICII!ood .,.,_ - ·
In 11111J1c wraa..iOINIID p<Odlclr--ID
dlomaiiiOtiPJ
11"""""nt llionnl'wln IIOCielrto\0!111 mllaolldc d ' -
kldhomm11111:1n
be .. nod-
EYal-~r HD&C In IIOdont'e mooltwo111Blto-..ilo rntt1
for 11\lpl\jll<llt fiiii'I'OdOM)'.
Clllptwl:t •llt-tl!i'tMele" I 2a•
,,, , . . . . (!)
1X Pltnl,tumot' annct be Blllutllllid
111
11>
r-Io- ol plmuy tnnor
C."*>omolh ~ (tumo< ol.!o ..0.5 mml. con11•od to 1n110000
n 1\lm<rln""'• lamina prcptlo <rwbnv.loolla and :!;I «n In aloe
T2 1\rmtrlnlll!ki'l!ll mu~<euillria ptCJJ)ltl Of> 1 an'" .m
T3
T4
-
..., ,.,....,_{1,
Tum£1' P6l'lllrt'tat tubwosa
Tum.,..lnllldos _ , '*""""'m (Nroal) «alii Of - · • . , .O)Ialnl
....
For OilY T, odd (mlf<>r mull! pi& tuh'lOfO
Nit
NO
Nl
r-Io......,., bmpllnodosnoclo.,.,.,.tl,.
~nlll,lmph
R""'nlll,lmph ncdo m -
IN!Io!l•*
•liM!
Mllln•n•••••
MQ r-iodlllol!t m-.ol&
ftql--
Ml
51qo0
SQII T1
OIIUrimol&ll&sla
lis NO
NO
MO
MO
MO
SQIII~ T2. NO
Slqa liB TS NO MO
Slqo II~ T4 NO MO
Slqo 1118 AnyT Nl MO
5lqo IV AnyT A111N Ml
210 I SECTION llh <II lAACT
,...,.. _m
1X Pttr.lfytl.l"n()r CUt not b&Ma.e!!Md
ro No oMienoo"' 1JIIrna.ytumor
T1 Tumor2anoriMI
T2 TUtnOf >2 Q1l blt I"Mrt >5 em
l3 TUrr>Of>5cmi>IAnot>l0<:m
T4 1\lmor >10 an 1'1 .,..,..,. dl""""""
...,..., ....... -Oil
~ Rl!PNIIJmJilmc!es C1111nol b o -
NO Noreafonll tlfnpllnoclo-
Nl RIPNIIJmJil,_ -.Ia
. _..., ..... 011
MQ
Ml
III.P ....... IIIIIII!UIItl
Slqj:IIA TlcwT2 NO 1110 Low
SQIIB T3 NO 1110 Low
SQIII Tl NO 1110 Hl!h
T2 NO 1110 Hi&ll
T4 NO 1110 Low
Slqti!IA l3 NO 1110 HIIJII
SIQ:II!IB T4 NO 1110 Htpl
SQIIV AIIYT Nl 1110 Any""'
AIIYT NO Ml Any""'
C111ptw n • T~t-t l!itMe~e~t I 211
<-~tit mlll&tion Jl<llllift. Palia1Zi<: G!Sfr, lhov,sll Gftm ohow pooicift for c-IO:t
_.,..;on by IHC, olao laid CD be <-~tit mutaJiDo noptm:.
Muoy p.oU!c GISI'a bchm: full o-.uaD.lll.dobt ..,•• ...., ....t thctc <2.""
ue~ll!li~~H~GISTadoluvc«~CD-·
cuize, whlch caD C)("«t m:n 10 CD 20 J'Wf d"!cr iaiiW fU111ClV• A number of
"""""""'h.&.. hem clm:lopal CD rille malify GIST, and proposal pidrtinrs
o.....t oo o.ltlldy of > 1SOO 1!1111D< GISTa wilhi""B"U:zm C..U.,.,...lll' ..., lirted
ill Toble 13.6 tAm J Smg Pli/JxJ. 200S;l~..S2). The <\I'CC lll4l&illll tw ~
GISTalllD. Table 13.7.
!. ,,..,....,_. kolll ~JIIIa- at••--'-"IJ'IIIIIolllllluue{IW.f)~·
il«~~c:rtr;~•odallJmllh=oompottdof~yhta:t~-n
Jl.celiJ (..l'ijJ. US). U,pto CII.Oo<bird ofpdli<: MAmmay ohow plumoaqtr>id
diff~. The GI INCt i& llu. moot """"""n oil!> m MALT 111DPhom.a
(SO% of.,....) &lldlhen0D!.41d! i.e th.e_n...,_ Gl w(5S% of cua).
Table 13.8 dcoc:dbca ~ u.oc:ful lA d!mngnlehl . . MAll' lym•
phOil'A from cbR.olc gaotr1t1o (..l!lc. U.54). Go.auic MALT lymph""'" uo
typiully poo«iti.. tw GD20 (..V.., U.S$), CD~ CDll, &l1d GD3S, b'atn.cg·
- &.r GDS, Cli1Q, ADd GD23. Som.e pm;.: MAil" lympli.CIDIAI are CI>43
politi.. (..F;s. U.S'6). D~ of li8ht choin ramclicm is lu:lpful ill
divri..WID!g MAll' lymphoma fmm .-:Uvt .....,.. G:ar1zi<: MALT lym-
ph.oma.o boo"' bem 1uociaa:d with H. /11lari ia.foai.cm; bowevtt, the d,C!a' of
dttt.elio&H. nkr/dc=aaeowith~CD~bomo,...diOIIICIU'OJ'OO•
iii.. ~ will be~.. fo< fl. ~ ia hia!O!"tho. ~The
"""* pollIll
llltlll!lfc1'lltlr.
l""pl\olll folll<io
"'*•llrc:dontr.rtson
ln-lbl~r ~photy!al
MN.T IJIIII!G•
f""'U""
Mlybop-.,.nl
SINIto lnt.wmed11111n lb,
~
Mq
-
I>&-
S..ll•na round, n'lllura
fmolp.jlr n u - - .
~
Pl'odcmnont ..-nt In S!>ll110, uoual~llmbd to
~phold l>lklou!'od \rm~ loll-, do not
lrnslfolkullr- may 0001<1>-coo
-C043
T ~pllo<:jQo (poon"" lor Vlrkbla In nLITiber, K:IIUenlcl
-le. . bon-""'-
Pn!domNnt. dltflnoly '""'""
CD3) the krrina ~and
lntelfdli::IJ\ar • -
In nt.mbor, UIWIJt ~It ll"'''"N!!I, diiPu,.,Y
pn!ISI!II'It., tki!!tlunfna
lnJn.ltplll\llklm, -ll&trt proprll,llck 11atrt chlln
dloln- NllllrlciSon
Ron> ond '"""""""""'~ t11o
USIIol\r poo'lnont tho
111111:ra111& \rm!>hold .... lnlh- ~phold «>lh
uo B«>liund folm •ro T .....nd lnd~lt
........., ala!'od"'r distl1bWd, ~·"'
-...:tli>n- d - nol404dont
-pylori Miybop-...nl M1Jb6~
mk::nxqlnbml
ln111h11on of mUCJ\ali>
mu..,_ bY~Phold...,
212 I SECTION Ill: Gl TRACT
I. INTRODUCTION. The indications for cytologic sampling of the stomach include the
presence of an inflammatory process or a neoplasm. Mucosal lesions can be sampled
by endoscopic brushing cytology, and intramural lesions by endoscopic ultrasound-
guided fine needle aspiration (EUS-FNA). Brushing cytology for malignancy has
a sensitivity of 85% to 93% and a specificity of 99%, both comparable to the
sensitivity and specificity of endoscopic biopsy. However, brushing cytology and
biopsy are best considered complementary for detection of malignancy (Acta Cytol.
1988;32:461; Acta Cytol. 1990;34:217). EUS-FNA for malignancy has a lower
sensitivity and specificity (Gastroenterology. 1997;112:1087), which most likely
reflects limitations of the technique due to inadequate sampling.
II. INFLAMMATORY PROCESSES. Antral mucosa brushing cytology with Papanicolaou
stain is a sensitive, accurate, and simple procedure for investigating the presence
of H. pylori infection in cases of gastritis. The bacteria presents as curved and S-
shaped rods with basophilic staining properties (World] Gastroenterol. 2005;11:
2784).
Ill. NEOPLASMS
A. Adenocarcinoma.
1. The smear of intestinal-type adenocarcinoma is hypercellular, consisting of
haphazardly arranged three-dimensional cell groups and atypical single cells.
The malignant cells show nuclear enlargement, hyperchromasia, and irregu-
lar nuclear membrane contours (e-Fig. 13.57). A necrotic, dirty background
is often present.
2. The cytologic diagnosis of diffuse-type adenocarcinoma is difficult due to
scarcity of the malignant cells; when present, the characteristic signet ring
cells demonstrate an intracytoplasmic vacuole that indents the nucleus into a
concave shape (e-Fig. 13.58), with associated nuclear hyperchromasia. The
differential diagnosis of the atypical cells in diffuse-type adenocarcinoma
includes histiocytes and goblet cells (Diagn Cytopathol. 2006;34:177).
B. Gastrointestinal stromal tumor (GIST). The smear shows microfragments and
sheets of spindle cells with moderate to high cellularity (e-Fig. 13.59), intact
single spindle cells, and abundant stripped nuclei. The spindle cells have spindle
to oval nuclei, fine chromatin, and abundant delicate cytoplasm with indistinct
borders (e-Fig. 13.60). Nuclear atypia, mitosis, and necrosis may be identified
occasionally. The epithelioid variant demonstrates large epithelioid cells with
round nuclei and distinct cell borders. GIST cannot be graded based on cyto-
logic specimens. Immunostains are required for a definitive diagnosis to exclude
other submucosal spindle cell neoplasms that possess similar cytomorphology
(Cancer. 2001;93:269; Am] Clin Pathol. 2003;119:703 ).
C. Neuroendocrine Tumor G1 {Carcinoid). The cytomorphology of carcinoid tumor
(low-grade neuroendocrine carcinoma) is identical to that of the tumor at other
sites. The smears are cellular and composed of loosely cohesive clusters and
Chapter 13 • The Stomach I 2 13
I. NORMAL ANATOMY
A. The small intestine is 6 to 7 m long and divided into the duodenum, jejunum,
and ileum. It begins at the distal gastric pylorus and ends at the ileocecal valve
and is lined throughout its length by villous mucosa. The individual villus is a
slendet; fingerlike projection with a variable length-to-crypt ratio ranging from
3:1 to 5:1 (e-Fig. 14.1).* The epithelium consists predominantly oftall, colum-
nar absorptive cells that have basally situated nuclei, eosinophilic cytoplasm,
and an apical brush border. The absorptive cells rest on a visible, refractile
terminal bar. Other cell types of the intestine include goblet cells, crypt cells,
basal cells, Paneth cells, and endocrine cells; the granules of Paneth cells are
refractile, eosinophilic, and supranuclear, whereas those of endocrine cells are
smallet; eosinophilic but nonrefractile, and infranuclear. The lamina propria
contains mixed inflammatory cells including plasma cells, although neutrophils
are restricted to vascular channels. Peyer's patches, which are lymphoid aggre-
gates, are distributed throughout the small intestine mucosa. Intraepithelial
lymphocytes (IELs) are normally rare (no more than one per five enterocytes
at the tips of the villi), although an increased IEL density may be seen in epithe-
lium overlying lymphoid aggregates or Peyer's patches in the distal ileum where
the associated villi may be shortened or even flattened. Shortened and broad-
ened villi may also be seen in duodenal mucosa overlying Brunner's glands.
B. The large intestine, or colon, is 1 to 1.5 m long and consists of the right and
left colon. The right colon is further subdivided into the cecum, ascending,
and proximal transverse colon; the left colon consists of the distal trans-
verse, descending, and sigmoid colon, and the rectum. The mucosa contains
evenly spaced, nonbranching crypts arranged perpendicularly to the lumen
and extending from the surface to the muscularis mucosae (e-Fig. 14.2). Occa-
sional branching crypts or slight crypt architectural distortion may be seen in
the rectum and sigmoid colon, and in areas adjacent to lymphoid aggregates.
Paneth cells may be seen until the mid-transverse colon and, as noted earliet;
IELs can be prominent in epithelium overlying lymphoid aggregates. Howevet;
there are no villi, the lining epithelium has no microvilli and does not rest on
a terminal bar, and goblet cells are more numerous (particularly in the left
colon). The lamina propria components are similar to those of the small intes-
tine, although the lamina propria is denser in the right colon, and muciphages
(mucin-containing macrophages) are more common in the lamina propria of
the left colon.
The entire small and large bowel mucosa rest on muscularis mucosae. This
smooth muscle layer delineates the lamina propria from the submucosa which
comprises loose fatty tissue with a rich angiolymphatic supply. The inner and
outer layers of the muscularis propria lie below the submucosa and are sep-
arated by the ganglio-neuronal Auerbach plexus. The entire surface of the
*All e-figures are available online via the Solution Site Image Bank.
214
Chapter 14 • The Intestines, Appendix, and Anus I 2 15
excision (TME). The portion of resected bowel is oriented, and the length,
diameter (or circumference), and wall thickness are measured. The length
and diameter of the appendix and the dimensions of mesentery are also mea-
sured, if present. The external surface (serosa in most cases) of the bowel is
inspected for tumor involvement, perforation, adhesion, and fat wrapping.
For TME specimens, the grossly observable completeness of the mesorec-
tum is evaluated as "complete," "near complete," or "incomplete" before
opening the bowel (see College of American Pathologists Cancer Protocols
for Colon and Rectum at www.cap.org). The bowel is opened longitudinally
along the antimesenteric border, unless this would mean cutting through the
tumor.
The maximal size of the tumor and the distance to the proximal and
distal resection margins, or to the closest margin in unoriented specimens,
are documented. After fresh tissue is collected for biobanking (as needed),
the specimen is pinned out on a wax board (mucosal side up) and fixed
by submerging in 10% formalin overnight. The tumor is then sectioned
to assess the depth of invasion; blocks for microscopic examination are
taken to include the area of deepest penetration and the relationship to
adjacent, grossly nonneoplastic mucosa. Additional sections include prox-
imal and distal resection margins; if tumor approximates the margin, such
as in APR or LAR specimens, the margin should be inked and multiple
sections perpendicular to the margin submitted; if the inked radial margin
is not included in the tumor sections, one separate radial margin section
should be submitted. One random section from normal-appearing bowel,
and sections from any additional gross lesions (such as separate polyps),
should also be submitted. If the appendix is present, it is handled as an
appendectomy specimen as described later.
The mesentery and soft tissue are also dissected for lymph nodes (many
nodes are located along large vessels), and the number and size range of
identified nodes are recorded. Small lymph nodes can be submitted in toto
without sectioning. Larger nodes are serially sectioned and the cut surfaces
examined; if metastatic carcinoma is grossly appreciated, as evidenced by
a white and hard cut surface, the size of the metastatic deposit should be
recorded, and one representative section from each grossly positive node
should be submitted. If the cut surfaces of the nodes are tan, soft, homo-
geneous, and lack gross evidence of metastasis, the entire node should be
submitted for microscopic evaluation. Although a minimum of 12 nodes is
required by established staging criteria, all nodes that can be found should
be submitted. Fewer nodes may be acceptable for small specimens, for cases
that have received preoperative chemoradiation, and for APR or LAR spec-
imens (because lymph nodes are less numerous below the peritoneal reflec-
tion). However, when fewer than 12 nodes are identified, a second attempt
to dissect lymph nodes is strongly recommended (and should be documented
in the pathology report).
2. Nontumor bowel resections
a. For polyposis specimens, pinning and gross examination are similar as
for tumor specimens. Sampling focuses on the largest lesions, lesions with
a distinct or worrisome gross appearance (including firmness, ulceration,
and adherence to the wall), and flat and depressed areas of mucosa.
b. Resections for inflammatory bowel disease (IBD), particularly for ulcera-
tive colitis (UC), require sequential sections spaced every 10 em. The sec-
tions include transition regions between normal-appearing and diseased
segments, distal and proximal margins, and representative inflamma-
tory polyps. Any focal lesions (such as areas with raised mucosa), fistula
tracts, and strictures are sampled. The appendix, if present, is handled
Chapter 14 • The Intestines, Appendix, and Anus I 2 17
_,_.,_
Com'""" llllloblo mmunode11eloney
Vhi•-
GIIRI ~~
-··-
Foodel.....
Crolln'od'-
lt>II....--EIIIa1 .,.,drome
s,.lomlc: llltdmmu,. d -
O.m.ttttll hlrpollf'<IITIIl
--~~~lnlllrntniiiDIJ df11P
-P1fo/llnlocllon
Chapter 14 • The Intestines, Appendix, and Anus I 2 19
apparent. A complete lack of goblet cells and/or Paneth cells may be seen
in some cases. Although some patients have anti-enterocyte and/or anti-
goblet cell antibodies, serologic tests are not routinely employed. A clinical
response to steroids may help establish the diagnosis.
5. Eosinophilic gastroenteritis involving the small intestine exhibits histologic
features similar to those described for eosinophilic gastritis (e-Fig. 14.11).
There may or may not be villous blunting, but IELs are usually not increased.
Parasitic infestations, food allergy including cow's milk protein intolerance,
a drug reaction, connective tissue disorders, and a neoplasm should be
excluded.
6. Common variable immunodeficiency is characterized by the absence of lam-
ina propria plasma cells (e-Fig. 14.12). Other features may include a vari-
able degree of villous blunting, intraepitheliallymphocytosis, and lymphoid
aggregates. Infectious agents, particularly Giardia, should be searched for
in these biopsies.
7. Microvillus inclusion disease is a rare autosomal recessive disease causing
intractable diarrhea in infancy. The hallmark of the disease is the loss of
a normal brush border on the luminal surface of the enterocytes. Instead,
the brush border is incorporated into the cytoplasm as apical microvillus
inclusions. The microscopic features can be best demonstrated by periodic
acid-Schiff (PAS) stain, electron microscopy, and immunostains for car-
cinoembryogenic antigen, CD10, or villin. Diffuse villous atrophy is also
present, but an inflammatory response and intraepithelial lymphocytosis
are not evident.
8. Lymphangiectasia, either primary (congenital) or secondary (due to obstruc-
tion), may present as a localized mass lesion or diffusely involve the bowel
(e-Fig. 14.13). The presence of secondary lymphangiectasia is concerning
for an unsampled underlying mass lesion as the source of obstruction, which
should be mentioned in the report.
9. Abetalipoproteinemia features lipid accumulation in enterocytes giving rise
to a clear or foamy appearance. The normal villous architecture is well
preserved.
C. Infectious diseases
1. Tropical sprue and bacterial overgrowth simulate celiac disease but may
involve the entire small intestine with more severe disease distally. Clin-
ical history, including any travel history, is important in establishing the
diagnosis.
2. Giardiasis does not induce significant villous architectural change or an
inflammatory response. The diagnosis is based on the identification of pear-
shaped trophozoites at the luminal surface of normal-appearing mucosa (e-
Fig. 14.14), which can be mistaken as cytoplasmic debris. The organisms
can be highlighted by trichrome and Giemsa stains.
Whipple disease exhibits distended villi due to lamina propria accumula-
tion of foamy macrophages stuffed with the diastase-resistant, PAS-positive
(e-Fig. 14.15), rod-shaped bacterium Tropheryma whippelii. The microor-
ganisms can also be detected by polymerase chain reaction (PCR) analysis
and electron microscopy. Gomori's methenamine silver (GMS), acid-fast
bacilli (AFB), or Fite stains should be performed on these biopsies to rule
out fungal (histoplasmosis) or mycobacterial (due to Mycobacterium avium
intracellulare) infections because the morphology of these infections are
quite similar to Whipple disease on H&E stain.
3. Cryptosporidiosis is characterized by uniform, spherical, 2- to 4-~m bodies
attached to the brush border that appear bluishonH&E stain (e-Fig.14.16).
The organisms may be confused with mucin droplets.
220 I SECTION Ill: Gl TRACT
...
Galrl,...,.
L<el~ ~~-~IIz poplido1nd PPIPI'Y·"""'uotla NETs
..... ,.,...
Som-.pR>dulila NET
u-
Ltlo~
GulrolrboUnllalromal tumor
Ltlomyou"""""
Anp""""•
KaDCOI ..,.,....
Olton
a, Jb nn
8ultlt t.omplloma
8...,.1 bl'nDholllll, uhCIMolllel*>•...t~~-.oltllonmodiiiD botoo>on <lti'U.oo ..,., 8...,.1
~plloma and Bultlt bl'npilomo
Olll'ullo l a r p - ~phoma
Inrn_..,_ tmallln-11 d!w.M lh:Iuc!M ""h""'Ychaln d!Muo)
"'*"llr ~plloma
Mlr&NI"""' bl'nplloma o f m u - ~pldd tl&iwe (MALT ~pllomal
Mlnlle ooll.wntp""""
T....l bl'nDholllll
·-""-
E'IIWC~ ...,..loled T-<ooll,wntph,.... (E'ATI.)
221 I SECTION llh <II lAACT
I'1."! t! I
Ij I ~ lhlutallt (1'NIIO . . . kllemt hw tllllll lnttttflllll
"'-'a-m
lX Prm., t1.mo<..nnot be .......~
TO r i o - ol polmuy tvmor
11> Cor<*\Oino 1'1 .au
Tie 1\Jm« I'IO!Id•lomlna _,.
Tlb 'nJMOf fniJII!ldM 1\JbMI.IOO:I&t
T2 TumOf ln\lades muscuiwtl proplta
T3 Tum«-· tlirOI.IIIII the .......,IIJtl Pf'lllN lrbtlle su-..., «Into
nonpofllon- portnu...,ler U..uo ~or rdlo~l
lOti\_,.., 2tm « _..
T4 1\Jmor ~111o ..w:.tal """"*''"or dii"OOdl/ln¥0doo o111or «111/10
or_,... tN!ud• dller ""'"'ohtrtdl-, ,_,,,..
...,....._m em,
>2 lind ahdomlr'oll,... blf""Yol....,..;for
duodenum only, I n - of 1>11\C,.... Of bloducl)
...,....,.,.,. llllttlll)
KIC Relllonollfroph IIOdoo tonnot be ..,. "'.S
r«l rio..,.,.! tlmril n O l l e -
Nl -.llslnl-.lr..,_tlmrii-
N2 -.llsln4ormmr...,...tjmph-
. _..., ..... 011
MQ riodlllol!t m-Ilo
Ml ~nt rruaatu!a
III.P .......
SQIO 11> NO l,f.)
fllqol T1 NO l,f.)
T2 NO l,f.)
SlqtiiA T3 NO l,f.)
SlqtiiB T4 NO l,f.)
Slqj:IIIIA AnyT Nl l,f.)
SQ>I!IB AnyT N2 l,f.)
SQIIV AnyT M:tN Ml
,.-tt
........ ~IW!b::le.tt _.tll'dCI.6It t:Wtua !e. tarJej:unutn al'ld lteu"n. ofh tr "llef'itl\d, tot
53 3
..........
Ml
Slqj:IO
SQII n
Dtawrtrrw•ttWa.
111' NO
NO
1110
1110
SQIIIA '12 NO 1110
fllqo liB T3 NO 1110
SlqtiiiA T4 NO 1110
SlqtiiiB AnyT Nl 1110
Slqj:i IV AnyT AnyN Ml
I'1.1 I II
I J tit(. :nu•IDI.IInoelt .r Hlttcqfc Alldllp., M-lllcllemle
Ob&tltiNnlciJI!!clloa>llls
Eil!cn>hMIUihiiiJ<: Escllorlc/lir aol
NSA!Dd.,_
Crolill't<>Oik
Rlllio11Gnet>lilt
Oollqonou&-
228 I SECTION Ill: Gl TRACT
-
lll1IIU~r ul:ola. triable, ~IIY CoiJblooll:t\1'1.1
B"""IMII 'Tilt> 'Titlebnod "'norrnol
C!Mjllna m. .ntMc: fd Common
Sl!lclure U sultj•- ~.~.,..
-
RtdOI-11\Mt IOOlf. ~ISl!.
Anll-M'II 5-IO'lf. -75'lr.
Wei~--""""'
Tronwnunoll)mpllold ...,.,.....
Common
Common
210 I SECTION llh <II lAACT
D\OIO'oDiilc
Nudol
"'*
<Oih 1\ore
Sl:rdttfldld, UMI!!t iMI&I
''""'*''
FreaUOirt
- " ' · ..rb.l!; ....,.
Domon:o11Gnfl<m Sllarp Gradurrlthlnolllon
&lrrc&l ndfiW Ml.fctlll Mld~dbtatod Prom..,....ll'CIIIor:lliral dlllort!on
~"'""'of"""'""".,. No Yos
m""""
PFOII'Inont lom!FIO DI"<IC>rlo.
,.,..,uclerlr...,
PFOII'Inontlomlno Dr<~C>III +
nlllln>jlllb
PFOII'Inont pfi311y No
lm...,nohlllloch..-y
lloll.-ln
''I.I Illtl£!: 'I Hlttolotfc Gfldlltl'lf Coloftle C1'1ft ,_l·lbt Dr.ut (GVHD)
lladt ~IIMIIIflll: fNIIrtt
1 lhdWuolcol -~~~a IWt¢YilCO, eDOroo tlmi>IIO<:YII: 1'111-
2 AI>OIII* CIYI>ta. CIYI>t•bac"" ..• e1ld rnbtld lomlha I>I'OI>IIo 1'1111:ra>
$ Laud lndli'odual <rnJ~a, •reo of mu""""- oi"'YP'", fooll ulcet-.
4 Wldoip'eod ""'"'a)'l)11, and oulflueplllolllm wllll m._ldonudlllon and
ulcollllm
2J4 I SECTION llh <II lAACT
'' 1,1,! I lt!iU I WHO llltolollc Claullcalloft ol TUIIIOf'Uf * Coloft lnd Rectum
(tllloiiii-
Adonorno
Tubullr
VIlola
Tubub¥111ouo
----
ll)opi.Ua (1-pilllollllnooploslo), """•-
Diotc>I.Ua (1-lihollll IIOOI>Iooil), tqjl ll"ldf>
ltyp&IJ)iulle llOttP.IIII..,.IUiilypeo
'hdlttNIMii!lted
- .." - 00i>r'<1nllpolnl
liarniJ1x>lnololul
adertOII"'''Il
{!M->
~1111d PQin>
JUI'IIIIIIo PQin>
Pau~potn>
o..,.,.,...
""'-''*'"',.
C~blffonn
.. -~ a.rlen..,..l,..,.
MeWlilY <t~rdnotn~~
MlcraP«~>IIuyaul:lr,..,,
Mudftoute.rdftOI'nll
~., .... .,.,.,._
St:Nr•B:I ~rdnOil'liD
Mlood -..u-
Smoll<l!ll NEC
EC col-n-.~roduct\.1 N£T
.,..I""""'
_..,..._""
L cal, alu""''f\'h Pllllillund PP/PW~roclu~ N£T•
u-
U.iorn'J'>I!III
-t.Mlll'llllll,..,lll!.mot
I.e~
Anp,...,..
l<iopool..,..,.
&rwr~awa
lk.oll !ifnpi\Gm.. lll<iudllhl&, wllll _,,..lntsmw!ID bol>.loon d-lq&
B-alli !lmpi\Gma •nd Buillllt lfmJ:IIoma
Bullllt tjmpi\Gm&
Dlll'lloe IIIIo 8-col b'rn~homo
Marilo ... b'rnpllan.t
M&lfJNI"""' tjmpi\Gml dmi_H.,.,....... tjmphold-.., (MAI.T
t,'mpllome)
$tc IIJUWI
Chapter 14 • The Intestines, Appendix, and Anus I 2 35
-11tbl.lk1Dtllon-.
m>e~~ior
""'""'""'
1\Jbulll
VIlola
1\Jbulrw!lk>ol;
---
l>)opi.Ua {1-pillloiiiJIIIOpioslo), - · -
l>)opi.Ua {1-pillloiiiJIIIOpioslo), lilltl fi'IC!6
lf.IWP!Mdc I>O~P
Seoolooemotod~
'hdlttNIMii!lted adertOII"'''Il
c.rm.m.
Jlolonocarmom•
MucfnC~~~.atdanocarcfnoma
~~~~~~ mudnwsnooplonm
~n1...a Clldnomo
UN!IIo...,tii!Od .,.,_m•
rwr..ntloe!lrl& •"''>l&amt
-ntloellno tum" {N£1)
NETGI (clrdnold)
NETG2
Nelrcontloe!h<O.....,mo (NEI))
Uollll«>ll NEC
Smolleo!INEC
EC <Ill, -n-.i>rodU~ NET
Goblllco!ICJ-
~c:dl, l!luoapHI<e popl»o 1nd PPII'I'Y'flroduclnJ NET
_._,.........
1\Jbulll Cllolrdd
Nllrom•
u-
u..._....,.
~lom!omt
l<iii>Oll..,..,.
&rwr~awa
IK151FT.,_
1'1.' IIJtlflll
,,, , . . . . (!)
Clf"$JWI
1X Pltni/Y tumot' <:~nrd be a nllnid
TO No e\4i:lence d l>llh'IIIIY 111nor
..... C.-eln d:u: lntra.optilellalor lrwulon tl ~rnN PfOI>IIs'
Tl 1\rm« fnilll!ki'i!ll&UbrnuQO,Isll
l2 Tuma- fnillOICies muiCUirt& ~rta
l3 Tum .. tnllldosiiiiOII,III 1M mu~a~flrla pi'Ojrilln!D p o -
UIIuao
1\irnor - - 1 0 lllo ot.nce of.......-.J l>flltMourn. lncblha;
rnuel110114 Pl.....,.ltlrnorl«lln lllo r1&tlt lower QuadiOnt llldJOf
dlred:ty lrtw.dM ctl'lll!lr t:l!p nt or .strud:l.lr_.,"
1\rm~» pertl!lthlbl: 'ltloi!in.l ~ttlllllm, ftteUj!fW rnueftoua
petltlntwil
tum<rwllllln!M 1\!111- Quadrant
T4b Tumor dhdlt l - o r Is adhonnliD allllr _ . , mudlns
C.rcfllll
1X Pitn::l!ity 1:l.l"nnr Cl mot bll • see !llk'ld
TO NotMdenoo tl PflftWYl!l""'r
Tl
Tla
Tlb
Tum"~~ em In.,_"•test
Tumor 2 em or les$"
Clrsbn'
MQ Nodlslont ,...,..,... (no palllobfle MO,'""' elnlool M ll>"""P"*'
.........p)
Ml Dktlnl: m"'utnls
Mia ln1r""',_' rnallubula bo)oond rWrt_. qllldrant,ln-na
c-udorrf;xor•• J)eltonea
Non~lm_,.,
,....,....D)
~-nt I!'DIInb (no palllol>lll= MO• ...., elnlool M lbQOI!Ip'*
Ml ObeortmotaD.Gio
244 I SECTION llh <II lAACT
__.....,__
Slqj:IO n. 1'10 MO
SIQ:II n 1'10 MO
1'2 NO MO
fllqo I!A 13 NO MO
fllqo liB T4a NO MO
Slqti!C T~b NO MO
Slqti!IA n Nl MO
T2 I'll MO
13 Nl MO
T4 Nl MO
AnyT N2 MO
AnyT NO MlaGl
AnyT NO Mla G2,3
AnyT Anyl'l Mlb AnyG
AnyT N2 MlaAnr Q
AnyT AnrN Mlb AnyO
C..illid.lbp ........
SIQ:II n 1'10 MO
Slqj:ll! T2. T3 1'10 MO
Sla.ellll T4 NO MO
AnyT Nl MO
fllqo IV Any T AnyN Ml
C!Nipt• 14 • liM: ltrtMtines, ~i.l4 ud ANa I 2•a
,, I.I IlltjfU( WIIO Hlttoktlc Clllll'kalloft o!TIIIWIIf ol111t Mill C.nal
ljtlhlllll ~~
p..,...,ant-
Anll ~olol """'*Ill' (d)oj:losla), low .,..so
Anll~olll,.....~.hllhlll>do
a-nd.....
-IN
l'<rilnoll !qUIIMI&In1ruc>llfl...l noq>l&als
f'aeotd-
SquiJl"'IOU.l <::11 c:arc!nana
Verntcou> <Orcttorno
Ad""""'.....,m"
Mudnoutlldi8f\OC:IImotrtlll
Naurcondccmo """'""""
Naurcondccmo tumor (NET)
NET Gl (.."'*""'J
NETG2
NaUICOIIdccltto -""""' INECI
~MINEC
StnoiMINEC
·-""-
.... . ,. . . . .Ill
, •• I SECTION llh <II lAACT
T4 •«.........
Tumo< <>11rr, " " " ' " - ad)ll:ont ...,.n(s), urdllra. _ .
11&11111 ....... - Oil
~ R.,...ll,mJil n-com,. bu•SMd
NO No f1IP>nlilympll nodo moi&I-
Nl -lnpo-lymphncdo(t)
N2 Me_. In unlauraii!C«nolllloc•hdkr h!IINI IJmP. node!tl
IU ,.,_.In poltecul Olld "'"'""' tJrnl>ll nodaoond/llr blolerall.., U'tJ/o1
lniJillliOI tJrnpllnodet.
.......,1... (10
MO
Ml
~~qo .......
DIIWrt-·
No dlltantfl'<illlll ..
SlqtO 11l NO MO
SIQ:tl n NO MO
Slqj:lll T2 NO MO
T3 NO MO
StqoiiiA n Nl MO
T2 Nl MO
T3 Nl MO
T4 NO MO
Slqj:IIIIB T4 Nl MO
NIYT ~ MO
AnyT N:l MO
fllqo IV Any T Arr,N Ml
Chapter 14 • The Intestines, Appendix, and Anus I 24 7
I. NORMAL ANATOMY. The largest solid organ of the body, the mass of the adult liver
is 1200 to 1600 g. The right, left, and caudate lobes are subdivided into segments
on the basis of inflow blood supply (e-Fig. 15.1).* The liver has dual inflow supply,
with approximately two-thirds from the low pressure, low 0 2 portal vein, and
one-third from the systemic pressure, high 02 hepatic artery. Pressure equalization
occurs in the sinusoids, along with the nutrient and 02 gradient from portal tracts
to terminal hepatic venules. The venous return is via the left, right, and middle
hepatic veins which join to form the inferior vena cava as it enters the heart at the
right atrium. Bile duct blood supply is entirely from the hepatic artery plexuses.
Microscopically, the hepatic cords are lined by reticulin fibers and separated
by sinusoids. The parenchyma is subdivided into acinar units of Rappaport which
reflect an oxygen/nutrient gradient from most (zone 1) to least (zone 3 ), respec-
tively; zone 2 is an ill-defined area in between. The anatomy of Rappaport's units
underlies many pathologic processes. The lobule, often used interchangeably with
acinus, is a term based on the concept of the hexagon in which hepatic cords radi-
ate from the central vein toward the portal tracts. Each portal tract is a fibrous
matrix that contains a branch of the hepatic artery, a portal vein, a bile duct, and a
poorly visualized lymphatic (e-Fig. 15.2). Larger portal tracts contain autonomic
nerve fibers. Inflammatory cells are typically lacking, or are few in number. The
parenchyma is separated from the portal tract at the limiting plate.
II. GROSS EXAMINATION AND SPECIMEN HANDLING
A. Needle core biopsy. Specimen handling depends on the reason for liver biopsy.
After measuring and description of the number of cores, liver biopsies are
wrapped in lens paper and fixed overnight; use of sponge pads is strongly dis-
couraged because of the artifacts created during sectioning. Protocol "special"
stains and six additional unstained sections are recommended for medical
liver biopsies at initial preparation. Stains include three hematoxylin and eosin
(H&E), a stain for collagen (trichrome or picrosirius red), reticulin, periodic
acid-Schiff after diastase (PAS-d), and modified Perls' for iron. Additional stains
that must be available include copper or copper binding protein (rhodanine,
orcein or Victoria blue; orcein is also useful to differentiate passive septa of col-
lapse from active elastic fiber deposition in fibrosis); and Verhoef£ van Gieson
(VVG) for vessel wall architecture (Semin Diagn Pathol. 2006;23:190).
1. Tumor. Processing of biopsies for diagnosis of tumors includes three levels
for H&E and six unstained for possible additional immunohistochemistry
(IHC).
2. lmmunocompromised patients. Biopsies from immunocompromised patients
(typically solid organ or bone marrow transplant patients) may or may not
require "rush" processing; clear communication with the submitting clin-
icians is required in these cases and fixation, grossing, and processing are
tailored to the clinical needs.
3. Medical liver biopsy. The reason(s) for the liver biopsy, that is, diagnosis
or confirmation; grading and staging of hepatitis; or other possible medical
questions should be clearly understood prior to sign out.
*All e-figures are available online via the Solution Site Image Bank.
248
Chapter 15 • The Liver I 24 9
4. Frozen section. Indications for frozen sections of liver core or wedge biopsies
include donor liver evaluations for quantity of steatosis and/or portal inflam-
mation; acute fatty liver of pregnancy (AFLP) for microvesicular steatosis
detection by oil red 0 stain (from sections of liver biopsies at any stage of
processing prior to xylene clearing, cut onto charged slides). Evaluation of
intraoperatively encountered lesions is done from fresh tissue submitted on
saline-moisturized gauze; cores are best sectioned with as little handling as
possible, sectioned at 90 degrees to the long axis. Wedge biopsies may require
breadloafing before sectioning. Frozen artifact creates spaces that may be
challenging to distinguish from fat; thus conservative estimates of the degree
of steatosis are recommended from frozen sections. If electron microscopic
examination is expected for a potential metabolic disease, additional tissue
should be fixed in 3% buffered glutaraldehyde.
5. Miscellaneous. Iron and copper tissue quantitation can be performed in ref-
erence laboratories directly from tissue in the paraffin block.
B. Wedge biopsy Dr excision. Wedge biopsy is not typically recommended for the
evaluation of diffuse liver parenchymal diseases, as the subcapsular parenchyma
contains fibrous extensions for 3 to 5 mm that may mimic fibrosis (e-Fig. 15.3).
The subcapsular regions tend to show parenchymal collapse; elastosis may occur
in this location as well as a result of chronic ischemia. Wedge excisions for super-
ficial, circumscribed lesions are managed similarly to resections, as described
below.
C. Segmentectomy, lobectomy, or panial hepatectomy are performed for large lesions
that are not amenable to wedge excision. The surgery may or may not fol-
low anatomic boundaries, and thus before sectioning, it is important to under-
stand the procedure that was done; review of the imaging studies and reports
is invaluable. After the type of surgery, mass, and dimensions of the speci-
men are recorded, the resection margin is inked, the appearance of the capsule
noted, and the specimen is sliced in the axial plane at about 0.5 em incre-
ments. Gross examination of the lesion(s) and nonlesionalliver parenchyma
should include color(s) (nutmeg; tan; bile-stained; hemorrhagic; yellow), via-
bility (necrotic, nonnecrotic), texture (firm; hard; soft; spongy), and presence
of nodularity. If a tumor has been preoperatively embolized via transarterial
chemo-embolization (TACE), the percentage of tumor necrosis grossly should
be assessed; however, only after complete submission and evaluation micro-
scopically can it be adequately reported. Tumor sections (at least three) should
demonstrate relationship of lesions to liver parenchyma, grossly visible vessels
or ducts, margins (if close), and any variable areas within the tumor. Sections
of nonneoplastic liver and inked resection margin(s) are submitted to evaluate
underlying liver disease, vascular alterations, and margin status. One nontumor
section of normal liver should be submitted and evaluated by routine special
stains.
D. Total hepatectomy (explant}- performed for end-stage chronic liver disease, ful-
minant hepatic failure, or metabolic disorders- is followed by orthotopic liver
transplantation. Radiology reports must be consulted before processing the spec-
imen to ensure that radiographically detected lesions are sampled. The total
weight, dimensions of each lobe, and capsule appearance are recorded. The
hilum is completely removed, breadloafed, and submitted in toto proximally to
distally without dissection. If a TIPSS stent has been placed, no attempt should
be made to remove it as the spring-opened wires are not protected; rather, the
hilar tissue should be dissected away. The liver is then placed on the cutting
board facing up, and sectioned axially cephalad-caudad in about 0.5 em incre-
ments. Each slice is carefully examined fresh and after fixation (especially in
cirrhotic livers) for bulging, large, or discolored nodules (which are all gross
features of dysplastic nodules [DNs], or small HCC) that require documentation
2!0 I SECTION Ill: Gl TRACT
which includes location, number, size, color, and relation to the capsule or hilum.
In their absence, two random sections from both the left and the right lobes are
submitted. Only one section is necessary for routine special stains. The native
and donor gallbladders are submitted as for routine cholecystectomies.
Ill. DIAGNOSTIC FEATURES OF COMMON NONNEOPLASTIC CONDITIONS. In the approach
to liver biopsy, knowledge of the clinical information is essential. The adequacy of
the biopsy should be assessed, which must be judged on the basis of the nature of
the question(s) being asked. Grading and staging chronic hepatitis ideally involve
a 1.5 em core, and up to 11 portal tracts; <5 portal tracts is not optimal (Semin
Diagn Patbol. 2006;23:132) (see Figs. 15.1 and 15.2).
Grading and staging schema were initially developed for comparisons of treat-
ment for autoimmune and "nonAnonB hepatitis" trials, but they quickly tran-
sitioned to apply to chronic hepatitis (Hepatology. 2000;31:241). All systems
share the assessment of portal and lobular necroinflammation for grade and fibro-
sis for stage (e-Figs. 15.1 and 15.2). One published system (Am] Surg Patbol.
1995;19:1409) is simple to apply and communicate clinically; this system can be
applied for any form of chronic hepatitis, but is not meant for cholestatic or vas-
cular diseases, or alcoholic or nonalcoholic fatty liver diseases (NAFLD).
Patterns of collagen deposition and architectural remodeling are frequently sug-
gestive of the precedent injury: hepatitic, biliary, vascular, alcoholic, etc. Viral hep-
atitis and alcoholic hepatitis differ as the former is portal-based, and the latter is
centered in zone 3, is perisinusoidal initially, and results in nodules the size of the
acinus (e.g., micronodular cirrhosis). The portal-portal fibrosis of the chronic bil-
iary diseases often results in cirrhotic remodeling with maintenance of the terminal
hepatic venule in its central location; a "jig-saw" pattern is therefore suggestive of
biliary disease.
• •
A B
•
• •
• •
c
• •
•
•· •
•
..·e• •.•
• •• • • •
• • • •
• •
•
• • • •
• • •• • .. . . . . •
• •
~ •••
• • • •
•
•••••
Figure 15.1 Ludwig and Batts Grading of chronic hepatitis. A: Stage 1. B: Stage 2. C: Stage 3.
D: Stage 4. From: Am] Surg Pathol. 1995;19:1409. With permission.
Chapter 15 • The Liver I 2s1
•
-~
A B
• •
(it •
(;)
• • •
~
• -~
~ • •
• • • •
Figure 15.2 Ludwig and Batts Staging of chronic hepatitis. A: Grade 1. B: Grade 2. C: Grade 3.
D: Grade 4. From: Am] Surg Pathol. 1995;19:1409. With penni.ssion.
CWia•*---.....
_,,... l*ill~
1111111 tllll.lr ..1. . . .
0:<0% 0: ~· 0:-
1: ~ I: 4l21:b< 1llid 1: Mid, tow
2: ~'JI. 2: 2-4120x ~old 2: ~•kdld, ma~
$: ..&,'Jj, $: ><Cf.ll)ftold
URIIIIIIIII,r _... Ul N ( - + loblllor ~m- '+ lldconlnJI)
IIIMiolt .., . . , - . . , - •tc~•••mlll
.....-""'""""")Mitll-
-·*"""-lollllt.......
Do-
ho Nldt au~ ,......llllltlllolll, N ..lnll QlclnM tlllh
.Willi"' MU:
2 : - 3 ~·· ......... pootol ......
3ol~l.lllf
4oC-It
Chapter 15 • The Liver I 2 55
13. Cystic fibrosis has hepatic manifestations in about 20% of patients. CF may
present as a mimic of biliary atresia or neonatal hepatitis, or later as por-
tal hypertension, due to bile duct mucus plugging and fibrosis. The histo-
logic hallmarks are dense eosinophilic inspissated mucous in dilated ducts,
cholangitis, ductular reaction, chronic inflammation, and fibrosis (e-Fig.
15.36). Focal biliary fibrosis occurs in up to 70% of adults and may warrant
liver transplant.
14. Drug- and toxin-induced liver injury (DILl), commonly in the differential for
unexplained liver test elevations, can be direct (predictable, intrinsic) or indi-
rect (unpredictable, idiosyncratic). Direct toxicity involves agents known to
produce liver damage in a dose-dependent manner; methotrexate, antibi-
otics, and chemotherapeutic agents are examples. Indirect toxicity is immune-
mediated and dose-independent; granulomatous and eosinophilic inflamma-
tion typifies this type of injury. Acute injury may lead to cholestatic hepatitis,
bland cholestasis, interlobular duct damage, acute hepatitis, and massive
necrosis. Chronic injury may assume the form of chronic hepatitis, granulo-
matous hepatitis, steatosis, steatohepatitis, vascular injury, fibrosis, cirrhosis,
or neoplasia. Oxaliplatin injury is discussed below in sinusoidal obstruction
syndrome (SOS).
C. AIH and bile duct disorders Df the liver
1. Autoimmune hepatitis {AIH} can present in adolescent or postmenopausal
females, and is associated with hypergammaglobulinemia (lgG) and high
titers of antinuclear and antismooth muscle antibodies in adults, and anti-
liver-kidney microsomal type 1 antibodies in girls. The diagnosis should only
be made after other metabolic diseases have been excluded, and after neg-
ative viral serologies have been demonstrated (Hepatology. 2008;48:169).
In classic cases, there is a dense portal and lobular mononuclear cell infil-
trate enriched in plasma cells (e-Fig. 15.37). Marked interface hepatitis, cen-
trilobular confluent or bridging necrosis, and hepatitic resetting are present.
Advanced fibrosis may be found at presentation. Cholestasis is rare but sig-
nifies severity. Alli may rarely present as fulminant hepatic failure. Likewise,
mild chronic hepatitis and/or cirrhosis may be the initial findings.
2. Primary biliary cirrhosis (PBC}, a progressive cholestatic disease of middle-
aged women, results in the destruction of intrahepatic bile ducts. lgM and
serum cholesterol are elevated. The early stage (Table 15.2) has mixed portal
inflammatory infiltrates and the pathognomonic florid duct lesion consist-
ing of granulomatous or lymphocytic infiltration of duct epithelium (e-Fig.
15.38). The granulomas in PBC are epithelioid, may be portal or lobular, and
present in any stage of disease. Stains for fungal and acid-fast organisms are
appropriate at the time of initial diagnosis. The disease is inhomogenous, but
ductular reaction, interface hepatitis, chronic cholestasis, and biliary piece-
meal necrosis develop with progression, with eventual bridging necrosis, sep-
tal fibrosis, and biliary cirrhosis. Chronic cholestasis (cholate stasis) is char-
acterized by periportal edema, ductular reaction, MDBs, lobular foam cells,
copper (e-Fig. 15 .39), and cholestatic rosettes. Nodular regenerative hyper-
plasia (NRH)-like parenchymal features may occur in any stage of PBC and
may explain the clinical findings of noncirrhotic variceal bleeding.
Autoimmune cholangiopathy and anti-mitochondrial antibody (AMA)
negative PBC are synonymous terms for seronegative PBC that is otherwise
clinically and histologically identical to PBC (and is distinct from overlap
syndrome as discussed below). ANA testing is often positive in this setting.
3. Primary sclerosing cholangitis {PSC), a progressive fibre-obliterative disorder
primarily of young men, is of unknown etiology. PSC affects the extra- and
intrahepatic biliary tree leading to biliary strictures and ectasias, and cirrho-
sis. PSC is strongly associated with ulcerative colitis. The definitive diagnosis
ClllptlrlS•l!M:Li\re.t I 217
...........,.,,.,_••.,.nt.....
~ I~MM ~
1 Fbld duct-
Blo dlll:t-._
Poo1ll"""'
Ponll W.rnrndon
~ -~~--
Dueollu p,_allo,, &-nl PMPMII-
Eicpono!ad po!UI- Peolponolln!l~m . .llon ± plool,..l.-oolo
Pl'oiJof- du<lullll
PlooaiTIOII""""*
3 ScoiTinB SellCIII-..
FI110IIIt flbraUI&Ojlla
~.a~s clfduett. Bltil!lrc """""I&
4 Nodu~rctlflmll ~
4. Portal vein thrombosis (PVT} may result in subtle or gross parenchymal atrophy
characterized by approximation of vascular structures (i.e., the infarct of
Zahn). Biliopathy may also result from PVT.
5. Osler-Weber-Rendu syndrome (hereditary hemorrhagic telangiectasia; HHT}
is an autosomal dominant disorder that results in multisystem angiodys-
plasias. Of the four known genetic subtypes, liver lesions are in the I-n-IT 2
group of alk1 gene mutations on chromosome 12; final diagnosis rests with
fulfillment of three of five Curacao criteria, which include hepatic AVMs as
well as family history(] Med Genet. 2011;48:73). Nosebleeds are common.
The hepatic malformations may be insidious and range from ischemic bil-
iopathy, to focal nodular hyperplasia (FNH) with high output cardiac failure,
to noncirrhotic portal hypertension. Hepatic lesions include intraparenchy-
mal thick-walled veins with adherent arteries, scattered dilated sinusoids,
and abnormally sized portal tracts with extruded dilated vascular channels.
Commonly, grossly observed subcapsular enlarged vessels are present.
E. Miscellaneous
1. Granulomas of various sizes occur in the liver. Underlying etiologies are as
variable as the morphology of the granulomas. Stains for infection organ-
isms and evaluation under polarized light are useful in the evaluation of true
epithelioid types. Considerations specific to liver include PBC, sarcoidosis,
DILl, HCV, fungal or mycobacterial infections, foreign bodies, and hepa-
tocellular adenoma (HCA); rarely, granulomatous hepatitis is a bona fide
clinico-pathologic diagnosis.
2. Pregnancy is associated with various mitochondrial alterations. AFLP occurs
in the late third trimester and is potentially fatal to both mother and fetus;
emergent delivery is the treatment. While the histologic hallmark is zone 3
or diffuse microvesicular steatosis, oil red 0 stain on a frozen section may be
required since hepatitic features and extramedullary hematopoiesis may be
present. Endophlebitis is common. The affected hepatocytes appear swollen,
and the microsteatosis gives an appearance of cytoplasmic reticulation (e-Fig.
15.46). Preeclampsia/eclampsia and HELLP syndrome (hemolysis, elevated
liver enzymes, and low platelets) may cause zone 1 hemorrhage, necrosis,
and fibrin deposition.
3. Ductal plate malformation (DPM) results from developmental arrest with per-
sistence of the embryologic ductal plate, which assumes an anastomosing
ring-like structure lining the periphery of the portal tracts (e-Fig. 15.47).
DPM may manifest as von Meyenburg complexes, congenital hepatic fibro-
sis, Caroli's syndrome or disease, or polycystic liver disease.
a. Congenital hepatic fibrosis (e-Figs. 15.48 and 15.49) is a significant cause
of noncirrhotic portal hypertension. The abnormal portal tracts are
expanded, have an increased number of aberrant duct profiles, show
hypoplastic or absent portal veins, and numerous hypertrophic hepatic
artery branches. Bridging is noted but portal-central relationships are
maintained, as is synthetic function. Inspissated bile may be noted in
ectatic ducts.
b. Caroli's disease is characterized by segmental cystic dilatation of the larger
intrahepatic ducts, usually accompanied by recurrent bacterial cholangitis
and biliary lithiasis. When associated with congenital hepatic fibrosis, it
is termed Caroli's syndrome.
c. Polycystic liver disease is a debilitating process due to massive enlargement
with cystic replacement of the parenchyma (e-Fig. 15.50). Liver function
is maintained, but transplantation may be needed for quality of life. Berry
aneurysm of the middle cerebral artery is a significant complication.
4. Ascending cholangitis is diagnosed clinically. Histologically, intraluminal neu-
trophils are present within the interlobular bile ducts. Other features of
biliary obstruction are usually noted (e-Fig. 15.51).
260 I SECTION Ill: Gl TRACT
EM1J CI
o.cone- ""'-""-""~'«.
rN)>olty of dud>: E"<llln01>hllc
tnlnsfMT'IItlon of the ()'toplum;
o.·-
I.Jie Cl
diiiiiJlll&ln
,.,.Irlnallll&duds
l..oD In ~SO% of portll
II"'ICC1SS18d ni.IICtoer D~ulc tre.ctt
(Nft)) redo; 1111cioor hYI>oldl""'*'l'o;
.,_, nueklor Ol>l...., d-onty
PIJ1Wtt lnlld l!f ~IY 'l>iiii...I.,.Ik
Tonnll'lll ,_,x:
llle duct min .,S(}ll;
lntlm~mlnlllnti~J!'~mllon
of.,..,..-.. Fcall obiWJrtkln
-.enullt.-.dmna3 LJIIc"'"" 3 """"""' ....tlnflonmrtlon Verle"" lnfloml!llltion
-- n- Mid""'""""""' n-
~ Sovote(bfld&1111)fl-
P<:nolnot hei:ollc
.,.,.,,....>:iS" of
Oca.olonol looo I'MIM'l.l: dS" of 1>0rtol
'-'h\10Mng
Oit.or """"
St>allod- "'~""' 111>0111 S I - l l foam ..a
of hlpcloq!6s
G. Technical complications usually occur during the first few months after trans-
plantation. Hepatic artery or PVT or stricture may cause zone 3 hepatocyte
necrosis, but infarction is rare. Ischemic damage of the biliary tree is a sign
of hepatic artery complication, with protean manifestations including abscess,
cholestasis, obstructive changes, stricture, or loss of the bile ducts. Thus, CR
cannot be diagnosed without demonstration of a patent hepatic artery. Hepatic
vein thrombosis or stricture results in changes of venous outflow obstruction.
Stenosis or obstruction of the bile duct anastomosis causes morphologic changes
similar to those of biliary obstruction or biliary cirrhosis.
H. Recurrent diseases. Most diseases recur in the transplanted liver but the time-
frame and severity vary; diagnosis is complicated by the fact that recurrent dis-
eases share histopathologic features with rejection or technical complications.
Histologic evidence of recurrent HCV initially is acidophil bodies; portal
and lobular chronic inflammation occur later. Overlapping features with mild
acute rejection include bile duct damage, endotheliitis, and mixed infiltrates;
thus, a final diagnosis should include, if possible, description of the balance
of damage due to hepatitis versus rejection. HBV recurrence is documented by
protocol evaluation ofHB Sand C Ag testing on every follow-up allograft biopsy
(e-Fig. 15.56).
Fibrosing cholestatic hepatitis (FCH) (e-Fig. 15.57) is a rare but progressive
disease seen in both recurrent hepatitis B and C, with rapidly rising bilirubin
that may result in graft loss. FCH's histologic features include marked portal and
periportal perisinusoidal fibrosis with ductular reaction, canalicular cholestasis,
and nonzonal hepatocyte ballooning; the latter may be the earliest clue to diag-
nosis. Inflammatory changes are generally mild. In FCH-B, HB Sand C Ag are
highly expressed in reinfected hepatocytes.
PBC and PSC may recur several years after transplantation. Even with gran-
ulomatous duct lesions, recurrent PBC is difficult to diagnose. Recurrent PSC
needs to be distinguished from technical complications due to hepatic artery or
biliary stricture, or biliary obstruction. Recurrent steatosis and steatohepatitis
are a challenge to distinguish from de novo occurrence due to persistence of
the patient's metabolic syndrome and/or the medications utilized for allografts.
Recurrent HCC or cholangiocarcinoma are typically rapidly lethal complica-
tions.
I. Acute GVHD (e-Fig. 15.58) following bone marrow or stem cell transplantation
shows duct damage, and less frequently endotheliitis. Hepatitic forms of GVHD
rnay coexist. The differential diagnosis includes viral infection and DILl. Chronic
GVHD typically occurs after 100 days and simulates ischemia or ductopenic CR.
Skin and GI GVHD are commonly concurrent.
V. DIAGNOSTIC FEATURES OF COMMON NEOPLASTIC AND TUMOR-LIKE CONDITIONS. The
current World Health Organization (WHO) histologic classification of tumors of
the liver and intrahepatic bile ducts is given in Table 15.5. The 2010 American
Joint Committee on Cancer (AJCC) tumor, node, metastasis (TNM) staging is in
Table 15.6.
The most common tumor type in noncirrhotic livers is metastatic; neoplasms
that commonly metastasize to the liver include carcinomas of colorectal, pancreatic,
renal, pulmonary, and breast; melanoma; and neuroendocrine tumors. Metastases
usually present as multiple nodules, in contrast to the single nodules of primary
liver tumors. Metastases to a cirrhotic liver are very uncommon.
A. Epithelial tumors
1. Benign hepatocellular tumors
a. Focal nodular hyperplasia (FNH} is a common non-neoplastic lesion. It
is not caused by oral contraceptive use, but instead is a polyclonal
regenerative response to a local vascular injury. FNH has no malignant
potential. The nodularity of the lesion underlies the original moniker of
Clllptlr 15. liM: Li\re.t I 211
I' f.t:] I I I:I:JI WHO Hlttololfc Clltltflr:a'tbl•t 1-.. of tttt Lllrer lnclllrtriiMpdc
' _ Bile Duda
_.. .. ~tl.n::h...........
"""""
Ha~lll allonana
HNFI4r MUllllod
ji-CGrln~
lnftomm""'l\ 1!1>13> mUllllod
Un-
Rlcll nodul.lr hypltr:f 1'1
"'...,""'""'"'lodw~-.
lMJo coileh..., ~n·rulttt •d)oldoelo')
Slnol coileh..., ~n·rulttt •d)oldoelo')
~noduleo;
LM.,..,S.
HIJIIpe
"'....,,
HaDG>colllrlll Cllel"'""'
Ettlt; h"'"""'llular co"*'«no
He~luouel.,.,.,tb"""""l~r
Hll.,..liu a.rc!l'lll:.'f'IW, Klrmot..:
HI~ tiu carc!l"'a"'W, liln::omlllold
Ljm~-noma
Ha~ .. •JIII>olollllrlont
Undlll'ltlll'ldolod ..,...,.,...
ljlti IIIII tl_...llllor;r
"""""
B., duct-..ma (poo1blllry PO\d hemlllomo and~
Mlo!OO)'IIIII;
Bllory -·
edeho11br<ltna
~-
BIIo'Y lrl1:ruj>llll ...l n"''>iult, e"'d• 9 (811N-$)
lnt-II"''III:Y rwoplam \01111 ..,.. «lnllrm•llllo111'1dtlnlr1Mpilllollllnoopiall
lnt-II"''III:Y rwoplam \OIIIIIilll!....,.lnt71.o!>ltllollll noq>llda
Mudn..,.q'lllc """'*""will- "''-lol.o1P'Ido lntraJIIlllllolll """'laMa
"'....,,
Muctlouoq'lllc IIOOdoom wtil hllh-lrade lntrael>tilollll noolllooll
lntnoho1>'11C ektJioiiP>oudn..,.
lnt~l j)~Jiplltwy
Mucti,..Oflllc: """'*""will
llll....lottf ..... ,.._., 01\11
an-
1'160_,., 'Mth •n •IW'('il.ted II'IWISht&Otlltd'loma
ColciP,tl.r nooted ePiileloiGI.I'Oinol111n01
lnYuMI-norna
~
~blnodhi!I>O-ILW-<h~n..,.
Hl~taam, mbted•tth•1 ......~1
Mllpnt ~~~~- 1111""'
214 I SECTION llh <II lAACT
~
An~poma(PEQmaJ
cmmouo homonlllolna
Honllollotn~l
Hlom"""'Y ~mor
~pl\lli!IIOI!Il
~pllq1om
~~~~~~m
&>lloly~tn~~•111m«
llf/l&1>fllf
Anp-
Em~ •n:oma ( u n d - ...,....)
Eptthollold hoou~ollom•
Klpooi-
U.~n:oma
Rill~
...............
lilfliO¥\al..,.,.ma
.,.._
TotituTli
Yolt•olunv ~oltl"""...,.!)
a.: a,...._.
Clllptlr 15. liM: Li\re.t I 21&
TAIL£ 16.
f""": E41o SB. 81111 OR. (omll!m 0C. ot•l. edi.IIJCC Conw S1Wo1r -.1tll ed.- 'Iori<,
NY:ilcoi-21110.'--\0ih .... -
collision tumors which contain typical HCC and typical CCa separately in
the same liver. In contrast, in combined HCC-CCa, the elements are admixed
within a single tumor. Combined HCC-CCa is further subdivided into a
"classical type," in which all elements are typical by light microscopy and
rnc, and subtypes with stem-cell features including "typical," "intermedi-
ate," and "cholangiocellular." Notably, in the "typical" HCC-CCa, either
intermediate cells (with features of both hepatocellular and biliary differen-
tiation) or stem-like cells may be found at interfaces of the two components
of the HCC-CCa. The greater the biliary differentiation of HCC-CCa, the
more abundant the sclerotic stroma. The outcome of combined HCC-CCa is
worse than traditional HCC, but lack of uniform terminology has prohibited
large and comparative series.
2. Hepatoblastoma, a primary malignant blastomatous neoplasm of several cell
lineages, is rare but is nonetheless the most common liver tumor of children.
Up to 80% to 90% of cases present by 5 years, and prematurity and low
birth weight are associations. Most present as symptomatic masses in the
right lobe.
a. The wholly epithelial type consists of four subtypes: fetal, mixed fetal
and embryonal, macrotrabecular, and small cell undifferentiated (SCUD).
Fetal cells, resembling adult hepatocytes but smaller in size, contain vari-
able amounts of cytoplasmic fat and glycogen, giving rise to an alternating
light-and-dark pattern (e-Fig. 15.69). Embryonal cells have a higher N/C
ratio and may form nests, rosettes, or small tubules. The macrotrabecular
has a growth pattern reminiscent of HCC.
b. The mixed epithelial-mesenchymal (MEM) type, with or without teratoid
features, contains malignant mesenchymal components such as cartilage
and osteoid in addition to the epithelial elements.
c. Hepatoblastoma, NOS, is the final category.
The outcome of hepatoblastoma has improved significantly with
the improved ability to shrink tumors prior to surgical extirpation.
Chemotherapy induces a variety of predictable tumoral alterations. Hepa-
toblastoma staging systems are based on pretreatment features and postre-
section findings.
C. Tumors of the biliary origin
1. Benign
a. Biliary cysts, whether solitary or as components of the ductal plate malfor-
mation (DPM) (described previously), are lined by a single layer of benign,
flattened, cuboidal or columnar biliary-type epithelium, surrounded by a
variable amount of fibrous tissue.
b. The von Mayenburg complex (e-Fig. 15.70) represents persistence of the
ductal plate; it is a common finding in normal livers, and may dilate to be
the origin of solitary cysts or cysts of polycystic livers. The VMC is usually
adjacent to a portal tract and appears as dilated duct-like structures within
mature fibrous stroma.
c. Ciliated foregut cyst {e-Fig. 15.71), while rare, may be found by imaging
or incidentally during surgery. It is subcapsular and often in segment 4.
The lining is composed of ciliated pseudostratified columnar epithelium
lying on a basement membrane.
d. Bile duct adenoma/peribiliary gland hamartoma is composed of closely
packed, well-formed, and relatively uniform small ducts that form a 1
to 20 mm diametet; sharply demarcated nodule (e-Fig. 15.72). Careful
microscopic examination is necessary to exclude the angulated architec-
ture, desmoplastic stroma, cytologic atypia, mitotic activity, and evidence
of invasive growth that are indicative of cholangiocarcinoma or metastatic
adenocarcinoma.
Chapter 15 • The Liver I 269
2. Premalignant
a. Biliary intraepithelial neoplasia, grade 3 (BILIN3) is a precursor of intra-
hepatic cholangiocarcinoma (ICC) characterized as flat dysplasia with
multilayering of nuclei and micropapillary intraluminal projections. The
morphologic findings are identical to of extrahepatic ducts.
b. Intraductal papillary neoplasm (lPN) is another precursor of ICC. This cat-
egory of neoplasms has replaced lesion previously known as biliary papil-
loma and papillomatosis (e-Fig. 15.73). Synchronous or metachronous
IPNs may develop throughout the biliary system. IPNs are classified as
low, intermediate, or high grade on the basis of cellular and nuclear
features; the parallels with pancreatic intraductal lesions are many, and
up to one-third are mucin producing. Most of the intrahepatic papil-
lary neoplasms are lined by biliary epithelium, although intestinal, onco-
cytic, or gastric type may also be present. Ducts may be massively
expanded by the villous growth and distinction with MCN requires thor-
ough evaluation of the stroma (e-Fig. 15.74). Invasive carcinoma may
be found in association with lPN; perineural invasion is common. Col-
loid carcinoma is the type of malignancy that occurs with intestinal-type
lPN.
c. Mucinous cystic neoplasm, formerly known as biliary cystadenoma as in
the gallbladder and pancreas, occurs in women, does not communicate
with the biliary tree, and consists of cystic dilated structures lined by a
layer of mucin-producing columnar cells on a spindled-type mesenchy-
mal stroma (e-Fig. 15.75). It may additionally show low- or high-grade
intraepithelial neoplasia and is graded on the basis of the highest compo-
nent; thus, numerous sections need to be evaluated. Cases associated with
an invasive carcinoma occur in older patients.
3. Malignant Intrahepatic biliary neoplasms
a. Intrahepatic cholangiocarcinoma (ICC) is a nonencapsulated mucin-
secreting adenocarcinoma usually arising in noncirrhotic livers. Cirrhosis
or chronic liver disease cannot be used as evidence to exclude the diagno-
sis, however. The tumor is characterized by three growth patterns that may
coexist: mass-forming, periductal, and intraductal; it is the mass-forming
pattern that results in the prominent desmoplastic background with the
characteristic firm, white or gray gross appearance.
The tumor is an adenocarcinoma with a variety of architectural pat-
terns resembling canals of Hering, cholangioles, bile ducts of varying sizes,
or peribiliary glands. In larger ICC, the central areas may be mostly stroma
and paucicellular, while the peripheral regions may show a growth pattern
that replaces and incorporates portal tracts. Greater than 90% express
keratins 7 and 19, and about 40% also express keratin 20; most cases are
also positive for expression of CEA (using a monoclonal antibody) and
CA19-9. ICC may be indistinguishable from metastatic ductal adenocarci-
noma of the pancreas by histologic and immunohistochemical evaluation.
ICC involving only the peribiliary glands may be challenging to distinguish
from reactive atypia.
The prognosis of ICC is dismal due to the lack of effective treatments.
The exceptions are small peripheral ICCs found incidentally, the muci-
nous intraductal growth ICC that are completely resected, and the highly
selected cases of perihilar cholangiocarcinoma arising in PSC treated with
protocolized neoadjuvant chemotherapy and liver transplantation. The
2010 AJCC TNM Staging for carcinomas of the intrahepatic bile ducts is
in Table 15.7.
b. Cystadenocarcinoma occurs in men as commonly as in women, and no
spindled stroma is present. These tumors likely arise from lPN within a
2'10 I SECTION llh <II lAACT
..,.rlrtd'r ~ilm.l du.:t !lllhtr lh4l1 &om MCN. Cu<rc<rt d.u qport «
pMSilolil suporia.r to that cEICC.
I. 'Ill.all af -KIIJ•I artJII
1. M-IIJU r.wta• i.o .., "'"''"""""' d.1dhood t - , uouolly of tho
d;shllol><, ~ b:r • gdatiDoua......,.ofbllc duc1a !A • d..W pllllo
..-Ia; cyotic lll'"""lD & loooc, ~.....tl><a, Ot
paUCtD; III.CICIIC&ym&J ccllt;
""'Y"oid - with -dmi•~ clluttrt of liver c:dll; 411!<1 ~u!Wy
l>eouto~ (0'1".. l$.76), MH mt.Y rmllt io
"""'!'licati0111 tt IU>t ~ Thu!ilit!M mm ..,...,...,W
(1111~oial~)
.:u:¢1WJ'
..,.., !Ul!S) - ! r o t i& ...,..,""""'
2. canr.ua lll~~a.P.a !o-!'il- U.7'7), !be moct""""""' ~om;811 twDor «
the u....,bccomco cl!Alc&lly t<lcYut wb=. >4 em. ID. '"'"'mol
drm... s!oo.
~may iac:rwt Ot IUpQI<le ~1tring prq;11«11CJ. G<otllr, QI.IDj)t$
uc ill">"'!»'~ w appear wcD.~'W Mlcto~r. ~r·
ma~.....w..,. ""'"ot 1111('<1111,.., Otpniulioo """mnom .ntf. time.,
""""""" !rat molpm triiJIJ£cnmo&cicm do.. 11.0t"""""
3. Wnllll lllnlill1f,tlin (II) i.t the moct """"""" lomigD mrmxi,ymollm:t
tw110t of lll.flllta wcldldrat.. Glda atc.IDOt'C comnwUy dcctcd lhaA boyt;
.,....,.... m&y lll.clo.dc hCOit i'o1lutc or~ o1. odlcr - IH
Chapter 15 • The Liver I 271
For cases of suspected abscess, it is always necessary to rule out the pres-
ence of a necrotic or infected neoplasm by extensive sampling and careful cyto-
logic evaluation for the presence of viable tumor cells which may be obscured
by the inflammation. In the instance of suspected tumoral necrosis or well-
differentiated HCC, re-biopsy with attention toward the periphery of the lesion
should be recommended, such that the interface of the lesion and surrounding
normal parenchyma may be examined (Radiology. 1997;203:1 ).
B. In the case of hydatid cyst, the aspirated fluid may be clear or turbid. Lami-
nated cyst walls, scolices, and hooklets are observed; a neutrophilic background
is sometimes present. Although aspiration of a hydatid cyst poses a risk of
anaphylactic reaction, successful procedures are the norm (Diagn Cytopathol.
1995;12:173).
Ill. VASCULAR LESIONS
A. The aspirate of a hemangioma usually shows abundant blood. Scattered stro-
mal fragments with bland elongated spindle cells are characteristic (Diagn
Cytopathol. 1998;19:250). Cell block preparations are helpful to identify the
vascular channels.
B. Aspirates of epithelioid hemangioendothelioma are paucicellulat; containing
single cells and small tissue fragments, and display a spectrum of cytomorphol-
ogy from small bland-appearing epithelioid and spindle cells, to malignant large
tumor cells. The epithelioid cells have abundant cytoplasm and may contain
characteristic intracytoplasmic lumina or sharply defined intranuclear cytoplas-
mic inclusions (Acta Cytol. 1997;41:5).
C. In angiosarcoma, the aspirate shows abundant blood in which there are iso-
lated cells and loose clusters of cells. The malignant cells are spindle-shaped to
epithelioid, and have hyperchromatic nuclei and abundant but ill-defined cyto-
plasm. Necrosis is present. Scattered malignant cells may show hemosiderin-
laden cytoplasm or erythrophagocytosis (Diagn Cytopathol. 1988;18:208).
Vasoformative structures such as intracytoplasmic lumina, microacinar lumen
formation, and vascular channels are identified inconsistently (Anat Pathol.
2000;114:210).
IV. FNH AND HCA. These entities typically occur in distinct clinical scenarios as a solitary
nodule and as such require clinical, pathologic, and radiologic correlation for the
correct diagnosis. In both cases, the diagnosis rests on evaluation of the presence
or absence of cytologically benign liver elements; it is therefore critical that only
lesional tissue is sampled (World] Surg Oncol. 2004;2).
A. FNH. Aspirates of FNH show both abundant benign hepatocytes and benign
biliary epithelial cells (Acta Cytol. 1989;33:857).
B. HCA aspirates consist of benign hepatocytes without biliary epithelial cells (Acta
Cytol. 1989;33:857).
V. HEPATOCELLULAR CARCINOMA (HCC). The most characteristic and specific features
are thickened hepatocyte trabeculae rimmed by spindle-shaped endothelial cells
(e-Fig. 15.83), hepatocyte tissue fragments with well-defined traversing capil-
laries (e-Fig. 15.84), increased nuclear to cytoplasmic ratio (e-Fig. 15.85), and
frequent atypical naked nuclei (e-Fig. 15.86) (Diagn Cytopathol. 1999;21:370;
Cancer. 1999;87:270). Poorly differentiated HCC demonstrates loose nests, three
dimensional fragments, and occasional gland-like structures of malignant hepato-
cytes with marked pleomorphism, macronucleoli, necrosis, and numerous mitoses
(Cancer. 2004;102:247). Features that favor hepatocytic origin include polygonal
shaped cells with centrally placed nuclei, abundant granular cytoplasm, and bile
pigment (e-Fig. 15.86); however, the distinction from cholangiocarcinoma and
metastatic adenocarcinoma is challenging and may require immunostains (Arch
Pathol Lab Med. 2007;131).
The fibrolamellar variant of HCC has a distinct cytomorphology which
includes poorly cohesive clusters of cells and singly dispersed large monotonous
Chapter 15 • The Liver I 2 73
cells with abundant granular cytoplasm, a low nuclear to cytoplasmic ratio, promi-
nent nucleoli, and intracytoplasmic hyaline globules. Fragments of lamellar colla-
gen bands with benign spindle-shaped cells are present. The thickened trabeculae
typical of classic HCC are not identified (Diagn Cytopathol. 1999;21:180).
VI. CHOLANGIOCARCINOMA. The aspirate is composed of cells arranged in crowded
sheets, three-dimensional clusters, acinar structures, or as singly dispersed cells.
The malignant cells (e-Figs. 15.87 and 15.88) show a high nuclear to cytoplasmic
ratio, irregular nuclear membranes, prominent nucleoli, and occasional intracyto-
plasmic mucin (Cancer. 2005;105:220). Poorly differentiated carcinoma displays
marked nuclear pleomorphism and necrosis.
Combined hepatocellular-cholangiocarcinoma is a hi-phenotypic tumor arising
from the canal of Hering cells with features of HCC and cholangiocarcinoma. The
tumor bridges these two entities both morphologically and by immunohistochem-
istry. These tumors (e-Fig. 15.89) are cytologically malignant but usually require
immunostains for correct categorization as the malignant cells exhibit of spectrum
of differentiation from hepatoid to glandular (Acta Cytol. 1997;41:1269).
VII. METASTATIC MALIGNANCY. Metastasis from an extrahepatic primary tumor is the
most common malignancy of the liver (Diagn Cytopathol. 2000;23:326). The most
common primary tumors that metastasize to the liver are adenocarcinomas aris-
ing from the colon (e-Fig. 15.90), lung, pancreas, breast, and kidney. Comparison
with the primary malignancy in cases of metastases is essential for diagnosis, as is
appropriate immunohistochemical characterization. Carcinomas with polygonal
cell morphology, such as neuroendocrine tumors, renal cell carcinoma, adrenocor-
tical carcinoma, and others must be differentiated from HCC on the basis of their
immunoprofile (Arch Pathol Lab Med. 2007;131:1648).
The Gallbladder and
Extrahepatic Biliary
Tree
Ta-Chiang Liu and Elizabeth M. Brunt
I. NORMAL ANATOMY. The gallbladder, comprised by the fundus, body, and neck, is
covered by serosa, except the portion in the liver fossa which merges with liver
parenchyma. The lining mucosa, a layer of folded columnar epithelium and lam-
ina propria of loose connective tissue, directly rests on muscularis propria which
consists of longitudinally oriented, to irregularly arranged bundles of smooth mus-
cle with overlying subserosa and serosa. No muscularis mucosae or submucosa
are present. Secretory mucous glands in the neck and extrahepatic bile ducts are
arranged in a lobular pattern (e-Fig. 16.1)."'
The extrahepatic ducts include the right and left hepatic ducts, which join to
form the common hepatic duct in the porta hepatis; when the common hepatic
duct is joined by the cystic duct, the common bile duct is formed. A single layer of
columnar cells lines the ducts and rests directly on dense connective tissue; from
proximal to distal, there is a variable periductal smooth muscle fiber investment,
intermingled with collagen bundles.
II. GROSS EXAMINATION
A. Cholecystectomy is most commonly performed for cholelithiasis. After the
gallbladder is measured and opened longitudinally, the following should be
described: serosal, mural, and mucosal appearances; cystic duct integrity; and
consistency, quantity, and color of stones. Full-thickness sections should be sub-
mitted from the fundus, body, neck, and duct; the cystic duct margin should also
be submitted, as well as any lymph nodes. For a suspicious lesion, the overly-
ing serosal surface or hepatic bed should be inked, the lesion breadloafed, and
sections taken to demonstrate relevant anatomic relationships.
The gross finding that the gallbladder wall is uniformly firm with an asso-
ciated flattened mucosal surface suggests the diagnosis of a so-called porcelain
gallbladder. After the specimen is photographed, at least one section per em
should be submitted (if not the entire specimen) to exclude adenocarcinoma
(e-Figs. 16.2 and 16.3).
B. Biopsy of the common bile duct is performed for stricture or overt neoplasm
during endoscopic retrograde cholangiopancreatography (ERCP). The num-
ber and dimensions of specimens should be recorded to ensure that the biopsy
fragments are adequately represented; inking is not needed. At the time of ini-
tial histologic sectioning, preparation of three hematoxylin and eosin (H&E)
stained slides together with six additional unstained slides avoids resurfac-
ing the block if subsequent deeper levels or special stains are required for
diagnosis.
C. Frozen section. Evaluation of bile duct margins by frozen section during pan-
creatoduodenectomy, or liver resections for bile duct adenocarcinoma, is often
performed. The tissue should be frozen in its entirety, oriented in the frozen sec-
tion block to obtain enface sections, and cut deeply to obtain sections that
"'All e-figures are available online via the Solution Site Image Bank.
274
Chapter 16 • The Gallbladder and Extrahepatic Biliary Tree I 2 75
represent the entire margin so that small foci of tumor are not missed by
inadequate sampling. The tissue that remains after frozen section should be
submitted for evaluation by permanent sections, which helps assure adequate
sampling.
Ill. DIAGNOSTIC FEATURES OF COMMON NONNEOPLASTIC CONDITIONS
A. Cholecystitis is associated with cholelithiasis in > 90% of the cases. Acute chole-
cystitis is characterized by full thickness edema, congestion, and an associated
fibrinopurulent serosal exudate. Hemorrhage, transmural necrosis (gangrenous
cholecystitis), and/or perforation may occur (e-Figs. 16.4 and 16.5).
Chronic cholecystitis is variably characterized by mural hypertrophy or atro-
phy with fibrosis and chronic inflammation (e-Fig. 16.6). Intestinal, pyloric, or
foveolar surface metaplasia may occur. Rokitansky-Aschoff sinuses, which are
herniations of the lining mucosa into the muscle layers, are common. Adeno-
myoma represents exaggerated herniations in the fundus accompanied by mus-
cular hypertrophy and may appear as a gross deformity (e-Figs. 16.7 to 16.9).
Both xanthogranulomatous cholecystitis (due to rupture of Rokitansky-Aschoff
sinuses) or mucosal ulceration from stones may be transmural with associated
bile extravasation and accumulation of foamy macrophages. Acalculous chole-
cystitis may be acute or chronic. Follicular cholecystitis may be associated with
primary sclerosing cholangitis (PSC) (e-Fig. 16.10).
B. Cholesterolosis (strawberry gallbladder) is characterized by yellow mucosal
specks grossly and lipid-laden macrophages in the lamina propria microscopi-
cally (e-Fig. 16.11). It is an incidental finding of no clinical significance.
C. Choledochal cyst, a form of fibropolycystic disease, results in fusiform or spheri-
cal dilatation of the common bile duct. Following photographic documentation,
the entire lesion should be submitted for microscopic examination to exclude
biliary intraepithelial neoplasia (BillN) or adenocarcinoma (e-Fig. 16.12).
D. Biliary atresia is a congenital process in which the extrahepatic ducts and gall-
bladder may be completely absent, or replaced by fibrous cords with no or only
a very small lumen.
E. PSC involving the extrahepatic biliary system is an idiopathic disease diagnosed
by cholangiography (discussed in more detail in Chap. 15).
F. Secondary sclerosing cholangitis, histologically indistinguishable from PSC, has
a variety of obstructive and nonobstructive etiologies, including tumors, toxins,
ischemia, and infections (including AIDS cholangiopathy).
IV. DIAGNOSTIC FEATURES OF COMMON NEOPLASMS AND PRECURSOR LESIONS (Table
16.1)
A. Adenoma is a single, small, and incidentally found polypoid lesion, and is char-
acterized by a tubular, papillary, or tubulopapillary architecture. A pyloric or
intestinal type epithelium is more common than a biliary type epithelium; squa-
mous morules, Paneth cells, and neuroendocrine cells may be present. By def-
inition, all adenomas are low grade, but larger adenomas may harbor foci of
high-grade intraepithelial neoplasia or invasive carcinoma and thus should be
entirely submitted for microscopic examination.
B. Biliary intraepithelial neoplasia (BiliN) is a classification nomenclature intro-
duced in 2010 by the World Health Organization (WHO). BiliN-1 and BiliN-2
(low and intermediate grade lesions) are incidental and without established clin-
ical significance. BiliN-3 may be associated with invasive carcinoma, and thus if
present in the gallbladder, thorough sampling (including of the cystic duct and
margin of excision) is necessary to exclude invasive carcinoma. If no invasive
carcinoma is present, and the surgical margin of the cystic duct is not involved,
cholecystectomy is considered curative. A distinguishing characteristic between
BiliN-3 and reparative atypia is the abrupt transition noted in the former (e-Fig.
16.13) compared with the gradual alterations and heterogenous, widespread
2'J& I SECTION llh <II lAACT
,_"""''~' """*"'
MudnOU& OjOI!c •"''ll&am will low- cr lntenned~ lnll'Mplliollal....,plula
MudnOUI <;fllSc n"''llasm will hW!1P'I'I&I-~ ~
CM:txlma
""'-''*"""'
""'..,........,.. blllai)'IY!>O
-"''..,........., .. llll'llllc towdar IYl>O
""'"""".....,.. IMMlfnlltype
-.,.,.
C!Mr c:el adlnacan::!1"1£1'1W
_._......
Mudnow:.tdlnOQIJ'dnorna
AdDnOrllqL81'10111 ~]I'IQ'NI
~~ (ll!lbladdii'J or l'lndudlll (lilf>duoto) 1>01illoJY nD®Iaom Mil"" .....la1od
IMI&Iht&o.R:ftoma
MudnOU& OjOI!c •"''ll&am will en .....,lft!d - """'""""'
Squvnoua ail an:t.oma
_,..._
Undllallllllilled <:~ll:lr!oma
_._,.........
1\Jbulu welndd
......,......
&rwr~awa
1X
ro
P""'"'
No -
Q.mor <:On not be • -
... ol polmel)' lllnv
11> Cor<tlomol'letu
n Of
1\lm« ln'MdMiomlno ptap!lt mu..,ulor leyel
1\lm<r ln'MdMiomlno ptap!lt
Tumor-.. rrwcullrllyef
Tumor ln\lac:l6l peltnuiCUilr connecttYe tl.a:tue; no 8111Muion trw,tond
.....,.orlntol\lot
1\lm0fll0mra1Do111o- Mloore!IIO,....,urnl•ndlor d~ectJ
ln\llldoodl& lhwandkronolher ~OIIOh Of_,... oueh ..
th&ttomtl.eh, d~um, eolon, M ~I'MIJ ornt!lll'b.IMJ «
-Pill<: llll&ducll
T4 Tum«-.. moln polt.tl...., or hopd:lllol)'arln¥11d•lwoor m""'
Gldrlhapolfc: _ . .. slrudll,..
111111111 .... IIOdltOII
Nl( ~JiymP,f110dMaiiii\OtMM-
No ,..,.I tlmlil nod&"""'"+
--to
N0
Nl M - • t o nodos •li>r'C 1M ,.ai: .a.ct, common blodud, lleplllc
arlor.r. andlof portll,.!n
p o - . porlco\1111, ,..pootor
"'ll!tc •111l1Y""' Dh nodoo
''''"'*''"•lc
Brtlly. and/or
IIGrt••s•••t• (1111)
MO
Ml
No dlllanl m-••
llllllnl m-Ill
fllltii'M....
$QIO 11> NO MO
Slqa I Tl NO MO
Slqo II T2 NO MO
Slqo II~ T3 NO MO
5lqo I!IB Tl-3 Nl MO
5lqo IVA T4 ND-1 MO
sq, Ml AIIYT N2 MO
AnyT AnyN Ml
278 I SECTION Ill: Gl TRACT
Intrahepatic 10%.
Distal30%
Figure 16.1 Anatomic boundaries for AJCC classification of adenocarcinomas of the extrahepatic
bile ducts.
2. IPMNs of the extrahepatic bile ducts share the epithelial types characteristic
of pancreatic IPMNs, including pancreatobiliary, intestinal, oncocytic, or
gastric. Papillomatosis, a recurrent and potentially multicentric condition of
the entire biliary tree, is considered a subset of IPMN.
3. Mucinous cystic neoplasm (MCN) has replaced the term cystadenoma. More
common in the extrahepatic ducts than gallbladdex; MCN may grow to
20 em in maximal dimension. As in its hepatic counterpart, the lesion
contains estrogen receptor and progesterone receptor positive mesenchymal
stroma. As with papillary neoplasms, MCN lesions may be associated with
invasive adenocarcinoma.
D. The classification of adenocarcinomas of the gallbladder and cystic duct is shown
in Table 16.1. Tumors of both sites have similar staging schemes (Table 16.2).
1. Adenocarcinoma of the gallbladder may grossly result in mural indura-
tion and mimic chronic cholecystitis, or grow as an intraluminal poly-
poid lesion (e-Fig. 16.14). In order of decreasing frequency, the common
subtypes are biliary, intestinal (tubular or goblet), and gastric foveolar
(e-Fig. 16.15).
2. Biliary adenocarcinomas may contain mixed cell types including intestinal,
goblet, and neuroendocrine cells. The tumors composed of intestinal type
cells are usually 1<20 and CDX2 immunopositive; the goblet cell variant
may also have Paneth and neuroendocrine cells. The gastric foveolar type is
usually well-differentiated (e-Figs. 16.16 and 16.17).
E. Adenocarcinomas of extrahepatic bile ducts have been reclassified by the AJCC as
perihilar or distal (Fig. 16.1), and each has its own staging scheme (Tables 16.3
and 16.4). Perihilar tumors arise proximal to cystic duct and account for 60% of
the adenocarcinomas; the distal tumors arise between the junction of the cystic
duct and common bile duct and the ampulla of Vater, and account for 30% of the
adenocarcinomas (e-Fig. 16.18). Grossly, there may be only subtle thickening of
the duct wall, and thorough sampling is required to evaluate margins and local
extension.
Clt8pt• lS • 1M a..llbllldtlolrsnd EXtrtfua.,rtie 8il8y TIS I 2'!•
Mllln•n•••••
MQ Nodlsant m-..1&
..........
Ml
51qo0
SQII
DIIUii metutMJs
1b
Tl
NO
NO
MO
MO
Slqa II ~ NO MO
Slqa 1114 TS NO MO
Slqo 1118 Tl-.1 Nl MO
5lqo IVA T4 NOnl MO
51qr:!MI AnyT N2 MO
lvP.IT /vPJN Ml
'"""' ~SB,I!ynl 00,
••• Sptwet: 2010. ~- ....-
C>m"""
CC, .. •t, ed!I.IIJCC Cilnoof ~-7th ... - ......
I. IAWILAIIIIU
A. lnlllla!IIJ IIIII& In i-"'"
wlae ...,.,g,~ devdo111 imP •IO<>&J.
izzd """"""' upinolea CJPicollr do.ow a""""""" ...,.,g, in!lam..,any cell& wilb
~ acctolatl•-uo:ry dcbdo. Eno wb= .,. ah:aa ula 'lridllD •
-oc ~la&m, M!>Ua~ obl thow onlr F•eri<: ~ ..-.! chla it cao
210 I SECTION llh <II lAACT
Tu .....-....s
C.rdnM>O In otu
W>llle ble duet ~~!If
Tu...,..lnll&d• b<¥M !he Will oiiM blr.<llot
Tu..,..lnllldos l!l61011:iodclef, "'....., duodonLm,"' dlllr odjlclnl
O!plii\OIIhout tn-wrtof lilt coll.oc IOCb,"' lilt SllpMor
m-niDtlcllllay
...,...,. . ,.,_Oil
T~
ra
Tu...,..ln\10""" 11\e""llacdo. or111o.,IIOIIor ~lc ""'~Y
lesion is cystic, the cyst contents and the cysts relationship to the main duct
should be also documented.
Sections from the lesion should include samples that demonstrate the rela-
tion of the lesion to margins and uninvolved pancreas. If the pancreatic lesion
is cystic and small, it should be submitted in its entirety. For large lesions (e.g.,
>5 em), at least one section per centimeter is recommended, which should focus
on thickened, solid, or irregular areas. One or more perpendicular sections from
the inked posterior (retroperitoneal) soft tissue margin should be submitted. One
to two representative sections from the spleen are sufficient unless gross abnor-
malities are detected. Finally, the peripancreatic and splenic hilar soft tissue is
searched for lymph nodes; all identified lymph nodes should be submitted in
their entirety for microscopic examination.
C. The Whipple procedure, performed for tumors of the pancreatic head, common
bile duct, ampullary, or periampullary region, involves excision of a composite
specimen usually consisting of the pancreatic head, a portion of the common
bile duct, the duodenum, the distal stomach, and the gallbladder. Mter orien-
tation, the dimension of each organ should be measured. The various surgical
margins of the pancreas (pancreatic neck, posteriot; portal vein groove, and
uncinate) should be inked with different colors in the operating room with the
surgeon's aid; frozen section evaluation of the pancreatic neck and bile duct mar-
gins is almost always requested intraoperatively. The stomach is opened along
the greater curvature and the duodenum is opened along the aspect opposite
the pancreas to avoid the ampulla. The pancreas can be sectioned along the
plane defined by the pancreatic duct and bile duct using probes as a guide, or
cut perpendicularly. The size, location, and nature of the tumor are recorded, as
is the relation of the tumor to the margins. If the tumor is cystic, the cyst con-
tents and the relationship of the cyst to the main duct should be documented.
The specimen is then pinned out and fixed in 10% formalin before grossing. In
general, one section per centimeter of the tumor is submitted; if the tumor is an
intraductal papillary mucinous neoplasm (IPMN) or mucinous cystic neoplasm
(MCN), and is noninvasive in the initial sections, it is necessary to return to the
gross specimen and submit the entire lesion to ensure that invasive tumor is not
missed. Additional sections should demonstrate the relation of the tumor to the
various margins of excision, uninvolved pancreas, ducts, ampulla, duodenum,
and soft tissue; one section from the proximal gastric resection margin, and
one from distal duodenal resection margin should also be submitted. One sec-
tion from the uninvolved pancreas and one from uninvolved ampulla are also
submitted if not already sampled in the tumor sections. All identified lymph
nodes in the soft tissue should be submitted for microscopic examination; there
is no need to separate the nodes into groups because they are all considered
regional. Microscopic examination of a minimum of 10 to 15 nodes has been
recommended for Whipple specimens.
Ill. DIAGNOSTIC FEATURES OF PANCREATITIS
A. Acute pancreatitis is an inflammatory process in which pancreatic enzymes
autodigest the gland. It is most commonly associated with biliary tract disease
(such as gallstones) and alcohol abuse. The pancreas is swollen and edematous,
and hemorrhagic in more severe cases. Chalky white fat necrosis may be evi-
dent. Histologically, mild pancreatitis is characterized by interstitial edema and
leukocytic infiltration; the pancreatic parenchyma may be well preserved, with
only limited necrosis. In severe cases, extensive necrosis and hemorrhage are
seen (e-Fig. 17.1). * Calcification and secondary infection may occur.
*All e-figures are available online via the Solution Site Image Bank.
284 I SECTION Ill: Gl TRACT
1'1."!'
'
JDUI DfldleUon !ktwMn lttladiiCIIII ,.,. ,-IIIICIIIolllNH!II- (liMit)
_ _. IIIICI,_ Cydk; lleoplum (liCN)
r-.111.. 11'1111 ....
PollanI popWrllon Oldar _,.,. 1nd mm Pnlclomlnont!1 mlddi!Hpd-
Lo<ollon 1'11>1111<"'"" H""' Td or l>cdy
Oomrnulllco1lon IOllll du""l Yeo No
S>j'dlm
O..llln~P6- No I'M
Chapter 17 • The Pancreas I 2 87
-·~·
Prfmtl.llrpurt-
~· 3 (PoniN.Sl
PonerMlle llltl'Uj>.,.lol n110111u1t,
~uclll ..pllo(Y m u - n110111asm
~ucllllllb'*>PII>IIIIy ,..plum
MLdnwt O)'SI!o n110111awn
llfllll1>fM
ad""""'.....,m•
..-.
Duell!
AIJ~I.IM'I'IOUl t~~~n::!l'd'M
Coil* arcmma ( M J - noncysllc catlnan.ol
~
_,lilly <:Ordnoma
_._na<llloardnoma
Ulld1'1&111111o!ed ..,.....,m•
Ul'odi'I&IWIIII!Od "'rcmma \Oiih -ll·lbpntoolla
k!,... e.!I a.rettorr\1
klrwcoiiOJS\IOSorn<""'*"'*'"'cmoo,ma
-· .,.....,..,..."'hOODlum
&>ld·-.doi>OI>IIIIIY
~.....,.,
*'"'""
~l'let!MtSe r.&li'O&I1~ mk:tO:edeno/'118
twroon6oaNtumm, nonl\mcllonll(lr.tdo I, po 2)
twroon6oaNaorcmma 0..,. aoll, 11mll aoll)
EC oat -""'~'""'"'*" nouroondOQ'Ino t1.mor1
Goo-11\11
.,.,......
Glico....,.mo
........
S:orr.ltbltefi:IOI'nlll
VI Porno
.,.._
......_..lit
a.: a,..._.
F10m: llaomon f T , - F, Hrubon RH. Tldse ND. ods. - Ha/111
lllmwt><lflht: t:l/tfl:1ltM: ~ ~ IAAC Prao; 2010. UIOd olill ~.
,_,-of
~ from dlrOBic P"""Uiilif ut a h*!>lw:a<d ~ p.<!W'a
(Tahk 17.4), prri""'rol ilmuian (..l'ijJ. 17.l4), •......lar in...,;cm, WI d..oe
onro'"nwri•""' .,..,...,]u ......U.nm: (.,.Y'IJ- 17.2S). In m.oclo:ratdy clilfoxm.
Qmd~ th<.reiaaam.:..:.n,of~dw:t-like
w!.....Jl tllblllatgl..do of..n.hloollc wl.&bo,pc, ill.clo.dll!;s..,mc~r
formed a!ande Oibri£o:lm wi 1'-'~ -dl. ~ .... - ,....._,..,
Cl'l•ptw 17 • liM: PII/ICI"EEiii9. I 2I •
M-
T4 TumOfln- 111o odlao""'or thawJIIri>t ,_,.,.lc arlii(Y
Cunr-lllo pltna!'ftumorl
...,1111.,.. IIOdlltOU
~·~~ tjmph nod• ...... be
Nl(
NO
Nl
~ """""! bmph noclo mol•"•*
Rllf'onll tjmph node molo1alls
IWIIIrt•ltllladt OIQ
hOC Dlolant m-14 Cl/lld be ..,."""
ftql_,...
MO
Ml
SlqoO
~dlllant m•ua•
Oblont m-.ol&
111 NO MO
5lqo lA T1 NO MO
51qr:!IB T2 NO MO
$QIIIA 1'3 NO MO
Slqa liB Tl-3 Nl MO
Slqa Ill T4 AnyN MO
Slqo IV AnyT AnyN Ml
CK20 is typically negative or only focally positive. Other markers that are
expressed in PDAs include claudin-4, cyclooxygenase (COX)-2, mesothelin,
KOC (K homology domain containing protein overexpressed in cancer), SlOOP,
and maspin. These markers may be useful in distinguishing ductal from nonduc-
tal pancreatic neoplasms and from benign pancreatic ducts, but are less useful in
distinguishing adenocarcinomas of nonpancreatic origin. Approximately 55%
of PDAs harbor mutations in DPC4, but this is not specific as loss of DPC4
expression is also observed in cholangiocarcinoma (Hum Pathol. 2002;33:877)
and colonic adenocarcinoma (Mutat Res. 1999;406:71).
PDA is associated with a dismal prognosis, and the overallS -year survival is
only 3% to 5%. Resection improves survival as shown by 10% to 20% 5-year
survival in patients treated with curative resection, but only 10% to 20% of
tumors are resectable at the time of diagnosis.
B. Variants of ductal adenocarcinoma
1. Adenosquamous carcinoma consists of neoplastic components with both duc-
tal and squamous differentiation (e-Fig. 17.27). Diagnosis requires the pres-
ence of a squamous component exceeding 30% of the neoplasm. Patients
with adenosquamous carcinoma have a poorer prognosis than those with
pure adenocarcinoma.
2. Colloid carcinoma (mucinous noncystic adenocarcinoma) is almost always
associated with intestinal-type IPMN, and rarely with a MCN. Colloid car-
cinoma is believed to have a better prognosis than conventional ductal ade-
nocarcinoma. Histologically, the tumor is similar to mucinous carcinoma
in other locations and is defined by mucin pools comprising >80% of the
tumor.
3. Hepatoid carcinoma is an extremely rare tumor in the pancreas. It can
have a pure form or be associated with ductal adenocarcinoma (Am Surg.
2004;70:1030), acinar cell carcinoma, or a neuroendocrine tumor (Cancer.
2000;88:1582; Am] Surg Pathol. 2007;31:146; Gut Liver. 2010;4:98). Mor-
phologically, hepatoid carcinoma consists of large polygonal cells with abun-
dant cytoplasm (e-Fig. 17.28). Immunohistochemically, the tumor cells are
positive for hepatocyte-specific antigen (hepar 1); CD10 and pCEA highlight
a canalicular pattern, and most cases are also positive for alpha fetoprotein.
Data on the prognosis of hepatoid carcinoma are very limited. Hepatoid
carcinoma should be distinguished from hepatocellular carcinoma arising in
ectopic liver in the pancreas (Virchows Arch. 2007;450:225) and metastatic
hepatocellular carcinoma from liver.
4. Medullary carcinoma is characterized by its distinct morphology that fea-
tures poor differentiation with limited gland formation, a syncytial growth
pattern, and a pushing border (Am J Pathol. 1998;152:1501). Some cases
are also associated with infiltration by prominent CD3+ T lymphocytes.
Medullary carcinoma arises sporadically or in association with Lynch syn-
drome. Immunohistochemically, medullary carcinoma often shows loss of
at least one DNA mismatch repair protein (Hum Pathol. 2006;37:1498).
Prognostically, patients with medullary carcinoma do better than those with
ductal adenocarcinoma.
5. Signet-ring cell carcinoma is an extremely rare variant with an extremely poor
prognosis. Metastasis from a gastrointestinal tract or breast primary should
always be excluded.
6. Undifferentiated/anaplastic carcinoma histologically exhibits little epithelial
differentiation, although some or most of the tumor cells express cytoker-
atins by immunohistochemistry. Morphologically, three variants have been
described: anaplastic giant cell carcinoma (e-Fig. 17.29), sarcomatoid carci-
noma, and carcinosarcoma (e-Fig. 17.30). The prognosis for this tumor is
extremely poor; the average survival is only 5 months.
Chapter 17 • The Pancreas I 2 91
CIPIIN)
I Pill
Rtdlo!V"''hlc:oltf llal«t.oollo Yot
Orooot; ......lo Yflo
Orooot;.....,lo miiCin Yflo
Sl!aol- duct U•ullly >lOmm
Wtl~!- popliN
-llonwltha>lold
CII'CII'IOIIWI
"""
"""
294 I SECTION Ill: Gl TRACT
ultrasound-guided fine needle aspiration (EUS-FNA) has greatly expanded the num-
ber and type of cases seen. When an abnormality is identified or suspected, EUS
makes it possible to visualize the pancreas by ultrasound, directly visualize the
lesion, and then perform an FNA.
By imaging, pancreatic lesions can be broadly categorized into two main
categories, solid and cystic. However, heterogeneous lesions with mixed solid-
cystic imaging features and other confounding factors that obscure a clear
classification also occur. The clinical approach and differential diagnostic con-
siderations vary significantly between these broad categories, and thus it is
important to ascertain the imaging characteristics of a pancreatic lesion in
order to appropriately evaluate and diagnose EUS-FNA specimens from the
pancreas.
II. SOLID LESIONS
A. Inflammatory
1. Chronic pancreatitis can present a significant clinical and pathologic chal-
lenge. Aspirate smears will usually be scant to minimally cellular, show dense
fragments of connective tissue with bland spindle cells, and fragments of
bland ductal and acinar epithelial elements which typically have only minimal
reactive cellular changes (e-Fig. 17.48). The presence of underlying chronic
pancreatitis does not alter the cytomorphologic findings required for a diag-
nosis of malignancy (Diagn Cytopathol. 2005;32:65).
B. Neoplasms
1. Adenocarcinoma accounts for the vast majority of pancreatic neoplasms. The
majority of patients have disease at clinical presentation that is not resectable
and thus EUS-FNA provides the definitive diagnosis. The cytopathologic
features of adenocarcinoma vary on the basis of the degree of differentiation;
individual cases that have a spectrum of differentiation are also encoun-
tered.
a. Poorly differentiated adenocarcinoma is less cohesive with single cells and
small groups of cells with overlap and crowding. Nuclei are round to
oval with evidence of pleomorphism, and have large nucleoli. The cells
usually have a moderate amount of granular cytoplasm with only few
intracytoplasmic vacuoles (e-Fig. 17.49).
b. Moderately differentiated adenocarcinoma shows more cohesion and flat
sheets of cells, with fewer single cells. The sheets frequently show a disor-
dered "honeycomb" pattern. The cells possess nuclei with size and shape
variation as well as prominent nucleoli. The cytoplasm is more volumi-
nous with intracytoplasmic vacuoles (e-Fig. 17.50).
c. Well-differentiated adenocarcinoma accounts for roughly 20% of aspirates
and can be diagnostically challenging. Aspirates show large cohesive frag-
ments with very few background single cells. The main diagnostic features
are nuclear enlargement, nuclear membrane irregularities, anisonucleo-
sis (at least 4x size variation), and cellular crowding and overlap (e-Fig.
17.51 and 17.52). However, these features can vary within aspirate smears
(Cancer. 2003;99:44).
d. Mucinous noncystic carcinoma shows abundant extracellular mucin with
single cells and groups of epithelial cells that have the typical cytomor-
phology of moderately differentiated adenocarcinoma, but they can be
scantily represented (e-Fig. 17.53).
e. Signet-ring adenocarcinoma presents a predominance of single cells and
small groups of cells that have a large intracytoplasmic vacuole compress-
ing the nucleus (e-Fig. 17.54).
f. Undifferentiated {anaplastic} carcinoma is cellular with numerous dispersed
single cells that are epithelioid and show marked pleomorphism, with
scattered intermixed giant tumor cells (e-Fig. 17.55).
296 I SECTION Ill: Gl TRACT
SUGGESTED READINGS
Ali SZ, Erozan YS, Hruban RH. Atlas of Pancreatic Cytopathology with Histopathologic Corre-
lations. New York: Demos Medical Publishing, LLC; 2009.
Bosman FT, Carneiro F, Hruban RH, et al., eds. Tumors of the pancreas. In: Pathology and Genetics
of Tumors of Endocrine Organs (WHO Classification of Tumors). Lyon, France: IACR Press;
2010:279-334.
Centeno BA, Pitman MB. Fine Needle Aspiration Biopsy of the Pancreas. Woburn, MA:
Butterworth-Heinemann; 1998.
Chhieng DC, Stelow EB. Pancreatic Cytopathology. New York: Springer Science+ Business Media,
LLC; 1997.
Hruban RH, Pitman MB, Klimstra DS, eds. Tumors of the pancreas In: Atlas of Tumor Pathology,
Fourth Series. Washington, DC: American Registry of Pathology; 2007.
Thompson LD, Heffess CS. Pancreas. In: Mills SE, ed. Steinberg's Diagnostic Surgical Pathology.
5th ed. Philadelphia: Lippincott Williams & Wilkins; 2009:1432-1491.
SECTION IV
Breast
Breast Pathology
Souzan Sanati, Omar Hameed, Joshua I. Warrick,
and Craig Allred
Large Lumpectomy
Superior
B D F H J
C E G I K
Inferior
B D F
c E G K
Figure 18.1 Schematic representation of "bread-loafing,. and sampling of mass lesions. A:
Specimen was serially sectioned perpendicular to its long axis, and an irregular mass was iden-
tified in the center of the specimen. Sections A and L would be submitted as medial and lateral
margins, respectively, that are perpendicularly sectioned and entirely submitted. Given the size
of the specimen and the mass, submission of sections B through K would appear to adequately
represent the entire mass, as well as the immediately adjacent superior, inferior, anterior, and
posterior (deep) margins.
Small Lumpectomy
Superior
CD E F G
:.. .
......... .:.... . .
.·. .. .
• • • I • .•
Medial : Lateral
Inferior
8 c D
~ E F G
3. Ma11in and biopsy raaxcisians. These cases range from unorientated flat por-
tions of tissue that should be laid flat in a cassette (shave margin), where the
presence of any malignancy seen histologically would thus be indicative of a
positive margin; to larger, variably oriented specimens that should be inked
preferentially, bread-loafed perpendicular to the new margin (oriented), and
submitted in a manner to examine the new margin. For reexcisions follow-
ing invasive carcinoma, the status of gross residual disease should be evalu-
ated as well as its relationship to different excision margins; in the absence
of gross disease, the specimen need only to be representatively sampled.
For reexcisions following a diagnosis of in situ carcinoma, the specimen
may need to be more widely sampled to detect residual disease and/or asso-
ciated invasive carcinoma. Complete reexcision of the cavity created by a
prior lumpectomy may also be performed; these specimens should be inked
with respect to orientation, and serially sectioned as in a lumpectomy. The
blood-filled biopsy cavity should be entirely submitted, in particular with
respect to new margins to evaluate the residual disease and status of surgical
margm.s.
4. Mastectomy specimens range from simple skin-sparing mastectomy (which
removes the breast only covered by the nipple-areola complex and a nar-
row rim of surrounding skin), to simple mastectomy (mastectomy without
axillary tissue), to modified radical mastectomy (mastectomy with axillary
lymph nodes). Radical mastectomy (modified radical mastectomy with pec-
toral muscles), is rarely performed in current practice. Mastectomies may be
prophylactic or therapeutic. When the specimen is received for pathologic
examination, it should first be measured and weighed. Then, the specimen
should be oriented and the surgical margins inked; it should then be serially
sectioned perpendicular to the skin at 5 mm intervals from the posterior
aspect, packed with gauze, and fixed in formalin overnight. The size of any
Chapter 18 • Breast Pathology I 3 03
masses, including their growth pattern, location, and distance from all sur-
gical margins, should be recorded; the same principles discussed above (for
excisional biopsy/lumpectomy specimens also apply to grossing mastectomy
specimens. Masses should be entirely submitted if small, in a manner that
demonstrates the relationship to the surgical margin. If a mass is predomi-
nantly composed of ductal carcinoma in situ (DCIS) with focal microinva-
sion, submitting the entire mass is required to exclude a larger size of inva-
sive carcinoma. In addition, at least one section of nipple, grossly normal
tissue from each of the four quadrants of the breast, skin, and dermal scars
from previous biopsies should be submitted for microscopic examination.
If the specimen is a modified-radical mastectomy, the axillary tail should
be removed and dissected for lymph nodes, which should all be submitted
for microscopic evaluation (see below). A search for lymph nodes should
always be performed even in simple mastectomies and if lymph nodes are
found, they should be sampled accordingly. It is important to correlate the
gross and microscopic findings with imaging studies to optimize patient
care; for nonpalpable lesions, radiographing the specimen with submission
of the entire area of radiologic abnormality is recommended.
5. Mammary implants should be documented, photographed, and inspected
grossly for any evidence of leakage. One or two sections are usually suffi-
cient to evaluate the surrounding fibrous "capsule" that is usually submitted
with the implant(s).
6. Sentinel lymph nodes. The sentinel lymph node(s) is (are) the first lymph
node or group of lymph nodes that drain the breast into the axilla and in
most cases they are the first lymph node(s) involved in metastatic carci-
noma. Only rarely does cancer "skip" the sentinel node and metastasize to
a nonsentinellymph node. As such, specimens designated as sentinel lymph
nodes should be dissected carefully. Intraoperative evaluation (frozen sec-
tions and/or touch preparations) is usually limited to lymph nodes grossly
suspicious for malignancy; in these situations, intraoperative imprint cytol-
ogy of the lymph node can provide results at the time of surgery. For perma-
nent sections, each node is serially cut at 2 mm intervals and, in the absence
of gross evidence of metastatic carcinoma, entirely submitted for histologic
examination. There are many different protocols for microscopic evaluation
of sentinel lymph node, but one common approach involves examination
of three hematoxylin and eosin (H&E)-stained sections from each paraffin
block. The use of cytokeratin-stained sections-if initial H&E sections are
negative-is optimal, but not required. In the event of a positive sentinel
lymph node, axillary dissection is indicated.
7. Nonsentinel (axillary} lymph nodes. Axillary lymph node dissection may be
performed in conjunction with lumpectomy in a patient with a positive
sentinel lymph node, or may be performed as part of a modified radical
mastectomy (see above). After carefully dissecting grossly benign appear-
ing nodes from the fat, nodes <0.4 em can be submitted intact, whereas
larger nodes need to be bisected or trisected and submitted in a manner
that permits accurate enumeration (either by submitting them in individ-
ual cassettes or by differential inking). If possible, the size of the largest
grossly positive node or metastatic deposit should be measured, and the
presence of extranodal extension sampled in one section. It is recommended
that a minimum of 10 lymph nodes should be identified in an axillary
dissection (although this number is dependent on the surgical technique
and the extent of axillary lymph node dissection during surgery). Atten-
tion should also be directed to identification of possible intramammary
lymph nodes, which are usually identified in the upper outer quadrant of the
breast.
304 I SECTION IV: BREAST
the breast tissue are subtle in comparison with other sites such as endometrium.
The follicular phase of the menstrual cycle is characterized by simple acini and
collagenized stroma, while the luteal phase is characterized by apical snouting
of the epithelial cells, prominent vacuolization of myoepithelial cells, and loose
edematous stroma. The epithelial cells show peak mitotic activity in the late luteal
phase. During pregnancy, there is progressive epithelial cell proliferation resulting
in an increase in both the number and the size ofTDLUs. By late pregnancy, lobular
myoepithelial cells become inconspicuous while the cytoplasm of the luminal
epithelium becomes vacuolated as secretions accumulate in the expanded lobules.
After parturition, florid changes including the frequent presence of luminal cells
with atypical nuclei protruding into the lumen (hobnail cells) can be seen, which
can be alarming to the inexperienced observer (e-Fig.18.5). Gradually and slowly,
the lobules involute to their resting appearance.
In postmenopausal women, the lobules undergo involution and atrophy char-
acterized by reduction in size and complexity with an increase in fat (type 1
lobules) (e-Figs. 18.6 to 18.8).
In contrast to women, due to lack of hormonal stimulation, TDLUs do not
develop to a significant extent in men, and male breast consists of branching ducts
within a fibroadipose stroma.
IV. INVASIVE BREAST CARCINOMA. Establishing the diagnosis of invasive breast can-
cer (IBC) is the first and most critical responsibility of pathologists (Table 18.1).
Most invasive carcinomas present as a palpable mass and/or as a mammographic
abnormality. However, in some cases the primary tumor is occult, and the patient
may present with lymph node or distant metastasis. The purpose of the pathology
report is to communicate all the diagnostic, prognostic, and predictive findings to
a multidisciplinary team of surgeons, oncologists, and other specialists; some find-
ings are strong prognostic factors (histologic type, histologic grade, lymph node
status), some determine the likelihood of response to specific treatment (hormonal
therapy, Trastuzumab), and some determine the need for additional surgical pro-
cedures (margin status). Determination of pathologic stage (Table 18.2) is vital
to determine prognosis and to guide the therapy. In addition to pathologic stage,
the prognosis of breast carcinoma is greatly dependent on additional prognostic
and predictive factors that are mandatory and should be evaluated and reported
for all breast carcinomas (15), and are most significant in lymph node-negative
breast carcinoma.
A. Prognostic and predictive factors
1. Histologic subtype. Histologic typing remains the gold standard for classi-
fication of breast carcinoma and provides useful prognostic information.
Five major types of IBC are currently recognized. Four are characterized
by relatively unique/uniform histologic features and, thus, they are referred
to as "special" histologic types. Collectively, the special types account for
about 25% of all the IBCs, and include the so-called invasive lobular, tubu-
lar, mucinous, and medullary carcinomas (approximately 15%, 5%, 2%
to 3%, and 1% to 2%, respectively) (Breast Cancer Res. 2008;10:54). The
remaining ""'75% of the breast carcinomas are histologically and prognos-
tically very heterogeneous and are referred to as invasive ductal carcinomas
(IDCs), no special type, or, not otherwise specified. Except, perhaps, inva-
sive lobular carcinoma (ILC), all the special type carcinomas have more
favorable prognosis compared with IDC, although the degree of improved
outcome is variable for different types. In addition, some pathologists rec-
ognize other less common special types of invasive carcinoma that have
a favorable prognosis. These include invasive cribriform (1 %), papillary
(<1 %), and adenoid cystic (0.1 %) carcinomas. Different experts have used
variable criteria for diagnosing special type carcinomas, which is partially
IDS I SECTION IV. BREASl
Hemlllllorno
An&tomo!ooh
- m~-plleiY.,...I"""" Hem~
Fl---1\'0)
Apoctil'lll atdnoma l'llolldOlJIII>m-llnmllllypofpioala
,_ploo!Scca-u M)'aftbnli>-
Upld<fi::!ICW<:Inoma
SloctOCOIY <Or<*>orno lh1lo rnm~~o~y ln)0)1Ibloblu1lc 111m or
~cordftGtno u - 01\d Ol\lloii>OII'III
Adonold ejOIIe Olrell"""' Gn!iftu!.ar eel tumor
Aelnle eoll01rcmrna -bn>IM
~llch c!Mrcelatdnoma
~"'' Cllll'dnome
Hllmllllloly..- .
Lobular 1100Dioelo
Lobular cordftGtno lh du
-""""'
Anp"""""
UllOM"""""
Rho~
o.-rcomo
~ucal.,...,..,._ lesi<N IA!om)o>IM
lllwll-lllypofpiosla Ltlom)<loarama
All <lllfll\1111 aiJDII
Alnbl dudllllflMOplule AIM'IIf....lll . . . .
DCIS Flbroodenomo
Ml:toift..... ooi<b>l!lll PIIY!Iodootnnor
~ucal po~ry ,...ram, P•uti ucalllramal......,.ma, low fl'lld•
Paplloml MemiTIIry hi!NI!Dma
Alypbl Pll>loma
HroduebV..bl<)'lllc popllluyam:tloma -of-IWII
Ad0110111u N~I>IOlde•orna
11Jbulu
~.., ...,..
"""""""'
"-1M ulonomo
Sfltw>""""'•odOIIOI!III
f"''ll't'& d " -of !II& nlptlle
Ploomorpll~ odonom•
ilu<:tll od..,me
•uellfc..._
C - 1 8 . - l'rt!lololfi I 107
T<la
T4b
-bod !dow
·-n
Elilonoioll10 d!eot\lllil, nollnoludl'wmll' -111> mlllldo
Ellem.a (inell.ldlnJliJ&d• d!nlolill t l
IIIIIIa ol$1 nod""' ollll&!liln oflho b.....,"'
CMftnod tD tile 111!\6 -~~~~~-not mMI 1M
-~~~for lnllommololy ar<tlom1
T4<: Beth T4a and T4b
bil
T4d
,,.,..._(Jill'
lrrflemrnalorye~r<tloma
pNIXlrd+), 1\lnor
(--~""'lnr-)
...,d_
hllldoalc Olldlolll d-.1 b!f H&I «lmrnurdlllli>Cileln~
on!I' by IJQIIh molecl.lorftndlnp
DNlml
pNla
pN1b
pN1c
-nlqUOI)
"'"""""""' (>0.2 rnm •hd <2.0 rnml
""' - In 1.$&>1111Jy ljmpll nodoa (It loot OM
~, !et:h lnftlerrtl!ll mamtn~~~rynodM (rnkroor~··'"*l
rM->2.0 nvn)
DN3
pN2a
pN21>
·-of
Mrllnlllllln4-9oli1Jyljmpll • - ( I t lout.,.dr!pool >2mm)
Mrllnl11llln clnlclltt•ppnntlramll nwnmary tlfnph n-ln 1M
...,
Indlron ~11\ir. laiJIIII<>Id
..
--
, _ lubullr 5 Sptulrrlld m..., •moll""" M.l*fad ~-will Yos ER (+), P(R (+), HER2 (-)
.-,. llponol oncls, opon llmlno.
I~ miiCII'lOUO 2-$ W<lkl~<jjmorcd\od 1:>1>161bod 1\mor<lei&~"' POOh of Yeo ER l+l.I'I!R 1+1. HER2 H
.,..-,, m..., eolall"""""'-*
-"~'-
..
, _ nw!ulllly
.-,,
1-.1! ro..,_,.,
eaJ/foc$
-
wolld1:UI!'ISCl1bed ,..,
...
Synejtill..,-, Pill~
bonllll, tlmp/IOplosn\I<!Yife
Y•Ofp\1'8) ER (-), 1'11'1 H, HER2 (-);
(lrlplo nop1!\'rl)
..
1 - m"""'"'~Iflly
.-,.
1-.1! $lnlor t1> 100, NST
ln1!1reta, !\WI NICIII>r ll'lllll
T~~mr _ . , wtilln om~
100, NST 7G-'7S S!>tula18d muo. Wllllblo varlilbr&. ~ otOIIOCiel Vamblo varlilbr&
Mllllploob: .,.,.-,, tjp& -~- ...
<I l.arpr •~z~> T11110r tnonslo,_lo ~LN ER (-), 1'11'1 (-), HER2 (-);
noreond* (sq-111)
0< rr..... ocl>fmalolomonls
_.,......
m~~~C~ste . . (lrlplo ._tNo)
ER IUlrl 1'11'1 <On b8 ~
-" lnll!andu~r...,.rto
....
0
IDC, ~dl.dlltlteii'IOMII; HST, M:~l t,or.a.
110 I SECTION IV. BREASl
..-...........
hlodOIOIO U0...7Sll0) oftl.mOf 2
IJUio « IlOilo (<1010 of u.mor 3
--
Flolcl""" (mm2J O.l!i2
().0
6-10
>11
0.274
().Q
10.1!1
>20
().312
0.11
12-:!2
>ZI
I
2
3
Chapter 18 • Breast Pathology I 3 11
is a better tool to assess the prognosis in any given tumor (Pathol Oncol
Res. 2008;14:113).
c. Surgical margin. The distance of tumor from the margin of surgical exci-
sion is also important because it is inversely related to the likelihood
of local recurrence, especially in tumors removed by lumpectomy. Wide
margins with grossly distinct tumors can be adequately measured with a
ruler, whereas close margins require microscopic measurement. A mar-
gin is considered positive only if the ink that was applied to the margin
at the time of gross examination transects tumor cells. The distance of
the closest margin, as well as its location, should be reported. The extent
of margin involvement can be relayed as unifocal (<4 mm), multifocal
(more than one focus), or extensive (5 mm or greater).
d. Others
i. (MI) is a measure of the capability of the tumor cells to divide and
replicate and is determined by calculating the average number of
mitotic figures in a minimum of 10 consecutive high-power fields
(HPFs) in the most mitotically active part of the tumor, usually at the
periphery of the tumor. Only the clearly identifiable mitotic figures
should be counted.
ii. The presence of lymphvascular space invasion (e-Fig. 18.12) is an
important and independent prognostic factor, particularly in node-
negative patients with IBC. The presence of lymphatic channel inva-
sion recognizes a subgroup of node-negative patients who are at
increased risk of lymph node metastasis or distant metastasis (Ann
Oncol. 2005;16:1569; Diagn Pathol. 2011;13:18). There is a high
degree of interobserver variability in diagnosing lymphvascular inva-
sion (LVI); therefore, adherence to strict criteria is recommended in
order to reduce this interobserver variability. It is recommended that
the focus of LVI should be outside the borders of invasive carcinoma.
In addition, identification of an endothelial lining, and lack of con-
formation of tumor emboli to the shape of vascular space, help to
differentiate a vascular space from artifactual stromal clefting. LVI
present within lymphatic spaces in the dermis is often correlated with
the clinical features of inflammatory carcinoma (diffuse erythema and
edema involving one-third or more of the breast). In the absence of
the clinical features of inflammatory carcinoma, this finding remains
a poor prognostic factor but is insufficient to classify a cancer as
inflammatory carcinoma.
iii. Skin and nipple involvement. Skin can directly be involved by under-
lying invasive carcinoma (with or without ulceration). This usually
is seen in association with large tumors, or in tumors that are small
but very superficial. In addition, tumor cells from underlying lact-
iferous ducts of the nipple that are involved by DCIS can percolate
through the epidermis without breaking through the basement mem-
brane (Paget's disease).
Recently, gene expression profiling studies have classified breast
tumors on the basis of their gene transcription patterns into differ-
ent types or classes with different prognostic implications (Nature.
2000;406:747). These new technologies provide important informa-
tion that can be integrated into routine patient care (see below), but
histologic typing of breast tumors still remains the gold standard
for classification of breast carcinoma and provides useful prognostic
information.
B. IDC, no special type is the most common subtype of breast cancer, comprising
70% to 75% of the IBCs. This subtype is composed of a heterogeneous mixture
312 I SECTION IV: BREAST
not overexpress the HER2 gene product, except the pleomorphic variant,
which may show HER2 overexpression.
2. Tubular carcinoma. This tumor type constitutes 5% of the invasive
carcinomas, even higher on screening detected tumors (] Clin Oncol.
1999;17:1442), and is associated with limited metastatic potential and an
excellent prognosis. As a result of increased use of screening mammogra-
phy, these tumors present as a nonpalpable mammographic abnormality,
or are incidentally found on biopsies for other abnormalities. Grossly, they
form a spiculated mass. Microscopically, these tumors are characterized by
a haphazard arrangement of angulated tubules with tapering ends and open
lumens. The tubules are lined by a single layer of epithelial cells that often
display eosinophilic to amphophilic cytoplasm with apical snouts and oval
nuclei, with only mild to moderate nuclear pleomorphism (e-Figs. 18.35 and
18.36). A cellulat; often, fibroelastotic stroma is characteristic. The lack of
a myoepithelial layer is a helpful feature in distinguishing this tumor from
benign sclerosing lesions of the breast (sclerosing adenosis, radial scat; com-
plex sclerosing lesions). Estrogen and progesterone receptors are expressed
in >90% of these tumors (] Clin Oncol. 1999;17:1442), but they rarely, if
ever, overexpress HER2. Axillary lymph node metastasis is uncommon, and
if present, occurs only with larger tumors and is limited to the low-axillary
lymph nodes; axillary lymph node is not associated with the same adverse
outcome as IDC, NST tumors(] Clin Oncol. 1999;17:1442).
3. Mucinous carcinoma. Depending on the series and stringency of diagnos-
tic criteria, this variant comprises 1% to 6% of all the invasive carcino-
mas. This tumor is also associated with a favorable outcome. Grossly, the
tumor is well circumscribed and lobulated, with a soft gelatinous consis-
tency, and a glistening cut surface. Microscopically, the tumor is composed
of tumor cell clusters and trabeculae floating within lakes of extracellular
mucin (e-Fig. 18.37). Nuclear grade characteristically is low to interme-
diate. These tumors typically are immunopositive for ER (>90% of the
tumors) and PgR biomarkers, and negative for HER2 overexpression (]
Clin Oncol. 1999;17:1442). A significant proportion of mucinous carcino-
mas show neuroendocrine differentiation (Mod Pathol. 2004;17:568).
Mucinous carcinoma should be differentiated from other mucin-
producing lesions of the breast. Mucocele-like lesions are characterized by
mucin filled benign ducts and cysts that often rupture and result in extrava-
sation of mucin into surrounding stroma. Sometimes, the ducts show pro-
liferative changes, and portions of ductal epithelium may become detached
and float within the mucin pool. Howevet; the linear configuration of epithe-
lial cells and presence of myoepithelial cells favor a diagnosis of mucocele-
like lesion. Whenever the distinction between the two is not possible, espe-
cially in core needle biopsies, excision should be recommended for definitive
evaluation(] Clin Pathol. 2008;61:11).
4. Medullary carcinoma. This tumor comprises 1% to 2% of all the invasive
carcinomas. There is an association between medullary carcinomas and
carcinomas with medullary features with familial mutations in the BRCA1
DNA repair gene. Medullary carcinoma is usually diagnosed in younger
women and presents with a palpable mass. On mammogram, the tumor is
seen as a well-circumscribed mass usually without calcifications. On gross
examination, the tumor is a well-circumscribed, soft, tan-brown to gray
masses, with a bulging cut surface. Hemorrhage, necrosis, or cystic change
may be noted. Microscopically, these tumors are characterized by a syncy-
tial growth pattern in> 75% of the tumot; an intense lymphoplasmacytic
infiltrate, pushing borders, highly pleomorphic nuclei (nuclear grade 2 to
3), and a lack of glandular differentiation (e-Fig. 18.38). Tumors that show
314 I SECTION IV: BREAST
most but not all of the above criteria are recognized as variant medullary car-
cinomas. Medullary carcinomas are generally are negative for all biomark-
ers (triple-negative tumors). Pure medullary carcinomas have a favorable
outcome, however, variant types do not share this prognostic advantage.
5. Other rare, but clinically significant subtypes
a. Invasive papillary (predominantly micropapillary) carcinoma. In a "pure"
form (> 75% of the tumor), invasive micropapillary carcinoma accounts
for < 1% of the invasive carcinoma; however, it is much more frequently
a minor component of IDC, NST tumors. Histologically, this variant is
composed of small solid clusters of malignant cells floating within clear
stromal spaces (e-Fig. 18.39). These cell clusters lack true fibrovascu-
lar cores and show reverse polarity (the apical surfaces of the cells are
polarized to the outside) (e-Fig. 18.40). The importance of recogniz-
ing this variant of invasive carcinoma (as well as mixed tumors with a
micropapillary component) is the associated high incidence of axillary
lymph node metastasis. It should be noted, however, that when matched
for stage, this variant does not necessarily have a worse prognosis than
IDC-NST (Mod Pathol. 1993;6:660; Am ] Surg Pathol. 2009;33:202;
Breast. 2010;19:231).
b. Metaplastic carcinomas are rare and comprise< 1% of the breast carcino-
mas. They include a heterogeneous group of tumors in which a portion of
the malignant cells have undergone transformation into a different cell
type: nonglandular epithelial (squamous), or mesenchymal cell (chon-
droid, osseous, muscle, spindle cell) types. These tumors are similar to
IDCs of no special type with regard to clinical presentation except that
they are usually larger at the time of presentation.
Tumors with squamous differentiation show a spectrum of differen-
tiation from well to poorly differentiated. Low-grade adenosquamous
carcinoma, frequently associated with papillary and sclerosing lesions,
is characterized by angulated glands embedded in a cellular stroma com-
posed of cells with low-grade cytologic atypia and focal squamous differ-
entiation (e-Fig.18.41). In contrast to higher-grade tumors that resemble
adenosquamous carcinoma elsewhere, low-grade adenosquamous carci-
nomas rarely metastasize. Metaplastic spindle cell carcinoma includes
carcinomas with abundant spindle cell transformation (e-Fig. 18.42).
The diagnosis is relatively straightforward when glandular elements are
evident, but when absent, diagnosis may only be made by demonstrat-
ing cytokeratin immunoreactivity in the spindle cells. It should be noted
that spindle cell carcinoma can be histologically bland and show min-
imal cytologic atypia (such as in the so-called fibromatosis-like spindle
cell carcinoma). The key to correct diagnosis is wide sampling of the
tumor to identify areas with clear glandular elements, and/or the use of
a panel of immunohistochemical markers, particularly high-molecular
weight/basal cytokeratins, myoepithelial markers, and p63 to demon-
strate the epithelial nature of the tumor (e-Fig. 18.43) (Histopathology.
2008;52:45). Metaplastic carcinomas with heterologous differentiation
are most commonly composed of foci of chondroid or osseous differ-
entiation; however, other types of mesenchymal differentiation such as
fibrosarcoma, rhabdomyosarcoma, and other sarcomas can also be seen
(e-Figs. 18.44 to 18.46). Despite the lower relative rate of lymph node
metastasis (considering their larger size at the time of diagnosis), meta-
plastic nonetheless carcinomas have a high rate of LVI and a poor prog-
nosis overall. ER, PgR, and HER2 are usually negative, but can be focally
expressed in the glandular component.
c. Inflammatory carcinoma is a clinical-pathologic entity characterized by
diffuse erythema, induration, and tenderness of breast skin associated
Chapter 18 • Breast Pathology I 3 15
with non pitting edema (peu d'orange) involving one-third or more of the
skin of breast. A breast mass may or may not be evident. Histologically,
there is extensive dermal lymphatic invasion by tumor emboli, which
is believed to be the underlying mechanism for the clinical picture. A
tissue diagnosis is necessary to demonstrate invasive carcinoma in the
underlying breast tissue. The presence of dermal lymphatic tumor emboli
without the skin changes does not qualify as inflammatory carcinoma.
These tumors are classified as T4d by the TNM staging system.
d. Microinvasive carcinoma is defined as extension of cancer cells beyond the
basement membrane of terminal duct-lobular units to an extent :::;1 mm.
Microinvasion is usually seen in association with high-grade DCIS. In this
setting, the presence of a dense periductal stromal reaction and periductal
lymphoplasmacytic infiltration should prompt the pathologist to search
for foci of microinvasion that occurs as single cells or small clusters infil-
trating outside the confines of TDLUs (e-Figs. 18.47 and 18.48). The
focus of invasion should be obvious and care should be taken to avoid
interpretation of DCIS involving the lobules (a so-called cancerization
of lobules), or DCIS involving sclerosing lesions, as microinvasion. In
this setting, immunostains for myoepithelial markers and examination
of deeper levels of the block can be extremely helpful. Of note, if there
are multiple foci of microinvasion, the number of foci and the size of
the largest focus should be noted; separate individual foci of microinva-
sion should not be added together (http://www.cap.org/appsldocslcom
mittees/cancer/cancer_protocols/2009/InvasiveBreast-09protocol.pdf).
e. Other rare types of carcinoma include, among others, adenoid cystic carci-
noma, invasive cribriform carcinoma, apocrine carcinoma, myoepithe-
lial carcinoma, sebaceous carcinoma, lipid-rich carcinoma, clear celV
glycogen-rich carcinoma, acinic cell carcinoma, and oncocytic carci-
noma.
V. PATHOLOGY OF TREATED INVASIVE BREAST CANCER. Neoadjuvant chemotherapy
and radiation are increasingly being used in the management of breast cancer,
and so it is important to be familiar with radiation-induced changes in normal
breast to avoid interpreting them as malignant or premalignant disease. Likewise,
familiarity with chemotherapy/radiation-induced changes in tumors is required
to evaluate response to treatment.
Posttreatment changes in normal breast tissue usually include lobular atro-
phy, hyalinization, and nuclear atypia with degenerative changes, both nuclear
and cytoplasmic. Response to treatment is often categorized clinically as com-
plete, partial, or no response, and is a strong prognostic factor for disease-free
and overall survival. During gross examination, careful attention to identifying
and evaluating the tumor bed is necessary. In addition, exhaustive sampling of
the tumor bed region is necessary before making a diagnosis of complete treat-
ment response. Treatment may affect the morphology of the primary tumor and
lymph nodes involved by metastatic disease producing various histologic changes
in tumor morphology including necrosis, chronic inflammation, foamy histiocytic
collections, decreased cellularity, cytoplasmic eosinophilia, nuclear alterations,
fibrosis, and hemosiderin deposition (e-Figs. 18.49 to 18.51). Invasive carcino-
mas with a minor response may show little change in size; with greater degrees of
response, the carcinoma shows decreased cellularity or may show multiple foci
of invasion scattered over a larger tumor bed. When evaluating treated carcino-
mas, the size of the tumor is determined by the largest focus of contiguous tumor
and/or the number of foci involved over the tumor bed, not by the area of tumor
bed. Most carcinomas are of the same grade after treatment, but in some cases a
change in the grade of tumor occurs. Changes in biomarker status rarely occur
after treatment. Many systems have been developed for grading of pathological
response to treatment (Breast. 2003;12:320;] Clin Oncol. 2007;25:4414).
316 I SECTION IV: BREAST
Lymph nodes with treatment response usually show fibrous scarring and
foamy histiocytic aggregates in the place of previous tumor (complete response),
or may show small tumor deposits within an area of fibrosis. A lymph node with
complete response should not be reported as "positive" for carcinoma.
VI. NONINVASIVE CARCINOMA. Noninvasive carcinomas have historically been divided
into two major categories: ductal and lobula:r; on the basis of the misconception
that they arise from ducts versus lobules, respectively. However, now it is recog-
nized that both of these lesions arise from TDLUs.
A. Ductal carcinoma in situ (DCIS) encompasses a heterogeneous group of lesions
with highly variable clinical presentations, histologic findings, biomarker pro-
files, genetic abnormalities, and clinical potential. They have in common a mon-
oclonal proliferation of neoplastic epithelial cells confined to the ductal-lobular
system, typically in a segmental distribution, without extension through the
basement membrane. DCIS is a nonobligate precursor of invasive cancer. It
expands and unfolds TDLUs, and may grow into larger ducts or into adja-
cent lobules, giving an appearance referred to as cancerization of lobules, or
may even grow further and expand into spheres, a source of the misnomer
"ductal" carcinoma. The most common clinical presentation ofDCIS is as cal-
cifications seen on mammogram (e-Figs.18.52 and 18.53) although up to 30%
of the detected DCIS is not associated with calcifications; in this latter scenario,
DCIS is seen as densities and architectural distortions on mammogram. Less
commonly, DCIS may present as a mass.
Grossly, most cases of DCIS are not visible. If palpable, they may form a
mass with cords of tissue extruding a paste-like material from their cut sur-
faces (a gross correlation to the material seen on histologic sections as comedo
necrosis, see below). Morphologically, DCIS is seen as proliferation of neoplas-
tic cells with variable nuclear cytology, and in a variety of growth patterns,
which usually grow over and obliterate the luminal space of the ducts and
TDLUs. High-grade DCIS may incite a desmoplastic stromal response mixed
with chronic inflammation which raises the concern for IBC (e-Fig. 18.54).
Myoepithelial markers may be useful in such cases as they highlight the myoep-
itheliallaye:r; thus providing evidence that proliferation has not yet breached
the ductal-lobular system. DCIS may become displaced during previous nee-
dle biopsy, leading to carcinoma within a biopsy tract that may be confused
with IBC in the subsequent excisional specimen. DOS may show a variety
of morphologies including apocrine (e-Figs. 18.55 and 18.56) and clear cell
(e-Fig. 18.57) differentiation. While familiarity with these morphologic pat-
terns is helpful in recognizing the lesion, most of these morphologic variants
do not bear clinical significance independent of their nuclear grade.
For clinical purposes, a few prognostic and/or diagnostic characteristics of
in situ carcinomas should be reported in a pathology report. This information
rna y affect the clinical decision making.
1. Size/extent. If the lesion is only present on one slide, microscopic mea-
surement of the focus of DOS will be the most accurate measurement. If
the lesion is present on multiple slides, the most accurate measurement is
achieved by correlating the microscopic sections containing DCIS with the
gross diagram of the breast specimen showing location and relationship of
different sections within the specimen (see e-Appendix 18.2). By correlat-
ing the two, an estimate size of the lesion can be provided. In rare cases
where an accurate size estimate cannot be given, extent of the lesion can be
reported as the fraction of slides involved by DOS.
2. Nuclear grade. On the basis of nuclear features, DCIS is classified into three
grades by SBR criteria: low, intermediate, or high grade (Table 18.5). Low-
grade DCIS consists of a proliferation of small, monomorphic, and evenly
spaced luminal epithelial cells with homogeneous chromatin distribution
..... !
,(In'S A' I ~tall) l,.,alOI&t)
Nu- -m•<IIIIID """""'~ ploomorpillo
Dioo<O<tlilllm
Nucioorolze 1.~1too> 111& olreof. 2.Sltnoo 11\o olrD of rod
blood oe!l or"""'"
111111 bb:ld oe!l « l'll:lrrrtill
U:t.tlopi!Niol nudiL& duc:l.llopi!NI~I nudol
Ch,....Un
dlllrlbutl:ln
Dl!IWa, flnoly clo!>t- .,_""' VMcuiw, lrrt&Uiu
<hiOI!IItin d\1111-n
,......
Nu-l
Oolollt01!Gn
~I
Pclulr>d -rd ttl&
Promlnont, mu._,lo
M/(t 1>0 hO<IUOIIt
Not~-rd
llmlhol_... lt.mlnol-
318 I SECTION IV: BREAST
B. Lobular neoplasia. This term refers to the entire spectrum of atypical epithelial
proliferations arising in TDLUs composed of loosely cohesive uniform cells
with small nuclei and indistinct nucleoli, which expand TDLUs and grow in a
Pagetoid fashion underneath the epithelial layer. Traditionally, LCIS and atypi-
cal lobular hyperplasia only differ in the degree of involvement and expansion
of the lobules; however, most authorities now consider such lobular prolif-
erations under the rubric of "lobular neoplasia" or "lobular intraepithelial
neoplasia." Many studies show that there is a direct relationship between the
extent of the disease and risk of developing IBC.
1. Lobular carcinoma in situ (LCIS} is characterized by a neoplastic proliferation
of small, loosely cohesive, uniform cells, with homogeneous chromatin and
inconspicuous nucleoli. The cells may contain intracytoplasmic vacuoles
containing eosinophilic mucin globules (e-Figs. 18.83 and 18.84). The pro-
liferating cells fill (and often distend) most of the lobules (>50%) in the
involved TDLUs. Although Pagetoid extension of neoplastic cells into the
major ductal system is usually seen with LCIS, it can also be seen in associa-
tion with DCIS; such extension undermines the normal epithelial layer and
produces a clover leaf-like appearance (e-Figs. 18.85 and 18.86). Differen-
tiating LCIS from intermediate and high-grade DCIS is usually straightfor-
ward. However, distinguishing LCIS from low-grade DCIS--particularly
with a solid growth pattern-can be challenging. In contrast to solid LG-
DCIS, the cells of LCIS are usually small and discohesive, and often display
intracytoplasmic lumina. The loss of membranous expression of E-cadherin
protein by immunohistochemistry has been recognized to be characteristic
of lobular carcinoma (cytoplasmic expression may still be evident) (e-Fig.
18.87), and retention of expression is characteristic of ductal carcinoma.
However, about 10% to 15% of the cases of lobular carcinomas retain E-
cadherin expression and a minority of ductal carcinomas lose E-cadherin
expression; therefore, the two should be distinguished mainly on histologic
grounds (Am J Surg Pathol. 2010;34:1472). Most cases of classic LCIS are
managed by steroid hormone receptor antagonists, and most data in the lit-
erature do not support reexcision for LCIS present at the surgical margins.
2. Pleomorphic LCIS is a morphologically distinct type of LCIS which shares
with classic LCIS distension of the TDLUs by discohesive malignant cells;
however, as the name indicates, it is composed of poorly differentiated cells
with significant pleomorphism that show two- to threefold nuclear size
variation, nuclear membrane irregularity, and prominent nucleoli. Central
comedo necrosis and calcifications may be observed (e-Fig. 18.88). The
main differential diagnosis for pleomorphic LCIS is intermediate- or high-
grade DCIS, a differential confounded by the not infrequent presence of
central comedo-like necrosis in all these lesions. The discohesive nature of
neoplastic cells can be very useful in confirming the diagnosis. Similar to
classic LCIS, pleomorphic LCIS can also spread along the duct system in
a Pagetoid fashion. Given the clinical, radiologic, and pathologic similar-
ities of pleomorphic LCIS to DCIS, including the frequent extension into
larger ducts, it is managed in a manner similar to DCIS; nevertheless, it is
still important to correctly identify pleomorphic LCIS due to the occasional
coexistence of ILC that can be focal and quite subtle (e-Fig. 18.28), espe-
cially on needle biopsy. In such difficult cases, the use of E-cadherin can be
potentially useful (Future Oncol. 2009;5:233). Intraepithelial macrophages
can mimic lobular neoplasia; however, microscopic examination at higher
magnification can resolve these issues (e-Fig. 18.89).
VII. THE ROLE OF BIOMARKERS IN BREAST CANCER. The role of biomarkers in estab-
lishing the prognosis and the management of patients with breast cancer can-
not be overemphasized. Established biomarkers such as estrogen (ER) and
320 I SECTION IV: BREAST
progesterone (PgR) receptors have been used both as prognostic factors and
as predictors of response to endocrine therapy in patients with breast cancer.
More recently, human epidermal growth factor receptor 2 (HER2) was added
to this list as a prognostic factor (marker of poor outcome), and as a predic-
tor of response to certain chemotherapeutic regimens including trastuzumab (a
monoclonal antibody against the HER2 receptor). Currently, evaluation of these
biomarkers in patients with invasive carcinoma is considered standard of care.
Additional biomarkers have changed breast cancer treatment in the past decade
and have the potential of enabling individualized therapies to different molecular
subgroups. The shift toward an earlier diagnosis of breast cancer due to improved
imaging methods and screening programs highlights the need for biomarker dis-
covery to quantify the residual risk of patients to indicate the potential value of
novel treatment strategies. With the introduction of high-throughput technolo-
gies, numerous multigene signatures have been identified that have the potential
to outperform traditional markers (Endocr Relat Cancer. 2010;17:R245).
A. Estrogen and progesterone receptors. The estrogen receptor belongs to a fam-
ily of nuclear hormone receptors that function as transcription factors when
they are bound to their respective ligands. Estrogen and progesterone recep-
tors are parts of complex signaling pathways, which interact with multiple
survival and proliferation pathways in the cell and play a critical role in the
development and progression of breast cancer. They have proven usefulness as
prognostic factors, and more importantly as predictive factors, in the clinical
management of breast cancer. There is growing evidence that patients with
endocrine-responsive breast cancers benefit less from adjuvant chemotherapy.
Assessment of ER and PgR status is an important task in the evaluation of
breast cancer and is mandated in every primary carcinoma, as well as metastatic
tumors if the result could influence decision making. Approximately 70% to
80% of the breast carcinomas are ER/PgR positive. Immunohistochemistry
is the first-line attempt in evaluating hormone-receptor status, although it
can be highly affected by a variety of preanalytic factors, including time of
tissue fixation and the antigen retrieval method(] Clin Oneal. 2010;28:2784).
The expression of PgR is strongly dependent on the presence of ER and is
reflective of a functioning ER pathway. Tumors expressing PgR but not the ER
are uncommon and represent < 1% of all the breast cancer cases in some large
series; therefore, retesting of the ER status in this setting is recommended to
eliminate false ER negativity. In rare cases of solely PgR-positive tumors, the
patients still benefit from endocrine therapy. Recently, quantitative methods
for RNA-based assays (21-gene assay) have been established for ER and PgR
quantification. These quantitative assays have potential advantages compared
to rnc methods and may become the assays of choice in the future.
Several studies have shown excellent concordance rate between evaluation
of biomarkers on needle core tissue and excisional specimen (Acta Oneal.
2008;47:38; Ann Oncol. 2009;20:1948; Clin Breast Cancer. 2010;10:154;
Cancer Sci. 2010;101:2074). Because of better fixation of the needle core tis-
sue, and the potential for guiding subsequent therapy, immunohistochemical
assessment of hormone receptors and HER2 status is best performed on needle
biopsy material if available. In cases of negative hormone receptors on core
biopsy material, reevaluation of the excisional specimen may be warranted to
exclude a negative result due to tumor heterogeneity. To minimize the effect of
preanalytic variables on test results, the ASCO/CAP recommended using only
10% NBF as a fixative and controlling the formalin fixation time between 6
and 48 hours.
The ASCO/CAP has recommended an algorithmic approach to assess hor-
monal status. To standardize immunohistochemical evaluation of breast car-
cinoma, several scoring systems incorporating both intensity and percentage
Chapter 18 • Breast Pathology I 321
general, compared with other subtypes, they have a better prognosis. Some of
the luminal B cancers may overexpress HER2. Both luminal A and B cancers
tend to respond to hormonal therapy, but luminal B cancers show a better
response to chemotherapeutic agents than luminal A.
HER2-associated cancers (accounting for 15% of the breast cancers) show
high expression of HER2 and low expression of ER and ER-regulated genes.
They are usually ER and PgR-negative and are more likely to be high grade
and involve axillary lymph nodes.
Basal-like cancers show high expression of basal epithelial genes (basal
cytokeratins such as CK 5/6 and CK 17) and low expression of ER, PgR, and
HER2 genes, the reason for calling these tumors triple-negative carcinomas.
This subtype is especially common in Mrican-American women, has a poor
prognosis, and is the most common phenotype in BRCA1-associated breast
cancers. These tumors are not amenable to treatment by endocrine therapy or
trastuzumab (Nature. 2000;406:747).
These results are significant in the sense that they show, although breast
cancer shows significant heterogeneity, from a biologic point of view many
tumors can be classified into particular groups on the basis of genetic similar-
ities, with gene signatures that correlate with clinical outcome and response
to chemotherapy. On the basis of genomic profiling data, several genomic
tests have been developed with the intent of providing even stronger prog-
nostic information. A 70-gene signature has been developed to predict the
risk of recurrence within 5 years in node-negative, ER-positive, or negative
patients; this test is able to accurately classify tumors into good or poor prog-
nostic categories (Nature. 2002;415:530; N Engl] Med. 2002;347:1999), is
performed on fresh frozen tissue, and has FDA approval for clinical use as
a prognostic test. Similarly, a 21-gene signature assay has been designed to
predict the risk of distant recurrence in patients withER-positive early breast
cancer (stage I and ll) who are receiving tamoxifen; it also predicts the benefit
from chemotherapy treatment in node-negative, ER-positive patients. This test
is done on formalin-fixed paraffin-embedded tissue and is based on real-time
polymerase chain reaction (PCR) measurement of the expression of 16 genes
with known significance in breast cancer; the results are used to calculate a
recurrence score (RS) that is predictive of overall survival independent of age
and tumor size, which classifies patients into groups with a low (< 10% ), inter-
mediate (10% to 30%), or high (>30%) risk of 10-year distant recurrence.
Another 76-gene assay has been developed for use on node-negative breast
cancers (PNAS. 2003;100:10393; Expert Rev Mol Diagn. 2004;4:169; N Eng
] Med. 2009;360:790) the 76 genes used in the assay do not overlap with the
genes used in 70-gene or 21-gene assays.
The above assays can only be performed by specific companies that have
patented the test, and quality assurance must be maintained within the com-
pany. Nonetheless, in current practice, histologic typing of breast tumors still
remains the gold standard for classification of breast carcinoma.
VIII. INTRADUCTAL PROLIFERATIVE LESIONS. Intraductal proliferative lesions that carry
an increased risk include atypical ductal hyperplasia (ADH) and atypical lobule
hyperplasia.
A.
1. Atypical ductal hyperplasia (ADH) is a term used to describe an intraductal
proliferation with some of the cytologic and/or architectural features of
low-grade DCIS, which qualitatively or quantitatively falls short of diag-
nosis of DCIS. Examples of ADH include a duct partially involved by a
uniform cell population resembling LG-DCIS (e-Figs. 18.95 and 18.96), or
a duct that is focally expanded by a uniform cell population with geometric
Chapter 18 • Breast Pathology I 3 23
spaces (e-Figs. 18.97 and 18.98). Quantitatively, there are a variety of arbi-
trary criteria for distinction of ADH from LG-DCIS (<2 mm or <2 duct
spaces) (Am] Surg Pathol. 1992;16:1133; Hum Pathol. 1992;23:1095).
While there is agreement on the importance of the extent of the lesion for
this distinction, there is no widely accepted size cutoff (e-Fig. 18.99). While
a diagnosis of DCIS confers a 10-fold risk of later developing IBC, and
ADH confers a four- to fivefold risk, they are both on a continuum of the
same neoplastic process. A diagnosis of ADH made on a core needle biopsy
is usually followed by excision, as studies have shown that depending on
the technique of the biopsy and size of the needle, a follow-up excision is
associated with an in situ or invasive carcinoma in 20% to 40% of these
cases (Adv Anat Pathol. 2003;10:113; Breast. 2011;20:50). It should be
noted that a diagnosis of ADH is only conferred to lesions with low-grade
nuclear cytology; intermediate and high-grade DCIS should be diagnosed
as such regardless of their size.
2. Aypicallobular hyperplasia (ALH) differs from LCIS with regard to the degree
of lobular involvement and expansion (e-Figs. 18.100 to 18.102). ALH
and LCIS are also on a continuum of the same neoplastic process. While
ALH is associated with sixfold increased risk of invasive carcinoma, LCIS
is associated with 12-fold increase in risk.
B. Several intraductal proliferative lesions do not carry an increased risk.
1. Usual ductal hyperplasia (UDH) is characterized by proliferation of a het-
erogeneous epithelial cell population with a tendency to bridge across, fill,
and distend duct lumens. Haphazard placement of cells of variable size
and shape (both epithelial and myoepithelial), variable spacing of the cells
often with prominent streaming and/or swirling, indistinct cell borders, and
irregular and usually peripheral secondary spaces are all features of UDH
that distinguish it from low-grade DCIS (e-Fig. 18.103). Bridges formed
in UDH are usually not rigid, and show stretching with a central attenua-
tion. Immunohistochemically, UDH is usually positive for high molecular
weight keratin (e-Fig. 18.104) unlike most examples of DCIS. UDH is not
a direct precursor of breast cancer; however, if florid, it is a marker of low
increased risk of breast cancer (1.5- to 2-fold), and it is usually not acted
upon clinically.
2. Columnar cell hyperplasia (CCH). The enlargement of normal TDLUs by
hyperplastic epithelial cells is one of the most common abnormalities of
growth in the adult female human breast (Adv Anat Pathol. 2003;10:113;
SeminDiagnPathol. 2004;21:18;AmJ Pathol. 2007;171:252;Histopathol-
ogy. 2008;52:11). These lesions have been called by many names over the
years (Semin Diagn Pathol. 2004;21:18), but currently they are most com-
monly referred to as columnar cell lesions (CCLs) or CCH. CCHs are often
multifocal, bilateral, and up to 100-fold larger than the TDLUs they evolve
from. The majority of CCH are lined by one or two layers of monotonous,
crowded columnar epithelial cells (e-Figs. 18.105 to 18.107), but many
exhibit more diverse histologic features contributing to the complex termi-
nology that has evolved to describe them.
It is currently unknown whether the cytologic atypia occasionally
observed in CCH is associated with significantly higher risk for developing
breast cancer than the majority of cases that are without atypia, although
in some preliminary studies up to 20% of the cases with atypia identified on
core biopsies are associated with cancer in follow-up excisions, a worrisome
association that has lead some authors to advocate follow-up excisions in
this setting (Am] Surg Pathol. 2005;29:734; Histopathology. 2008;52:11).
However, not all studies find a significant relationship between CCH with
atypia and cancer. It seems likely that some CCH may indeed represent
324 I SECTION IV: BREAST
I. MEDICAL RENAL BIOPSY HANDLING AND PROCESSING. This chapter covers med-
ical renal biopsies; biopsy for diagnosis of renal masses is covered in Chap-
ter 20. Renal allograft biopsy pathology is added at the end of this chapter.
Renal biopsies are usually received in transport media, allowing distribution of
tissue for light microscopy (LM), immunofluorescence (IF), and electron micros-
copy (EM).
A. Light microscopy. A minimum of three hematoxylin and eosin (H&E), one
trichrome, two periodic acid-Schiff (PAS) stains, and one Jones silver-stained
section is required. A minimum of seven glomeruli and one artery is required
for adequate LM evaluation.
B. Immunofluorescence. A minimum of two glomeruli is required. A direct IF
method is routinely used employing a panel of antibodies, including anti-IgG,
IgA, IgM, C3, Cl q, fibrinogen, albumin, and kappa and lambda light chains. It
is important to document the staining pattern (linear vs. granular) and distribu-
tion (mesangial, loop, or combined).
C. Electron microscopy. Ultrastructural evaluation of two glomeruli is recom-
mended. EM allows for evaluation of cellular and extracellular abnormalities in
the glomeruli, tubules, interstitium, and vessels. EM is very useful in confirming
the presence and distribution of electron-dense deposits, which can be located
in the mesangium or the glomerular capillary basement membrane. In the base-
ment membrane, electron-dense deposits can be subepithelial, subendothelial,
or intramembranous (surrounded by basement membrane).
II. GLOMERULAR DISEASES. Glomerular diseases may be primary or secondary (associ-
ated with systemic diseases). Patients can be grouped into those that present with
nephrotic syndrome (>3 g urine protein/day), nephritic syndrome (proteinuria+
hematuria), or isolated hematuria. It is important for the pathologist to have access
to patient•s clinical and laboratory data. For example, minimal change disease
(MCD), focal segmental glomerulosclerosis, and membranous glomerulonephri-
tis (GN) usually present with nephrotic syndrome. In contrast, postinfectious GN
usually presents with nephritic syndrome.
A. MCDifocal segmental glomerulosclerosis (FSGS). MCD and FSGS are the most
common causes of nephrotic syndrome in children and are also common in
adults, affecting about 10% to 20% of patients with kidney disease. Primary
329
330 I SECTION V: URINARY TRACT
*All e-figures are available online via the Solution Site Image Bank.
I"IIJ!I Jli!U llljof81emiNirPalbou•- - l o i i X . . , -. . IJii.M IIIII:-
-
llnl•l clllope
Mlll'roii<N,_
RIS
PltndiYNOS
seooriC!aJYNOS
ll'lllillu1IIIJ
·~lllfCilillliiJ
IIA-IiSP. MCMSGS
I.IJDUO
'nldlllofe
Mamtni'QI.l
D~-
,__k
MPGN
liS!' -·-
QN
c--
Prll~n
>li(W,.
«oo<:erd:
llldillrl!lllm
Dlallallo
Am,.,_
MPON
.PC.
'lllln momtni'IO Cei!L61r""" 1Do11'10Dhi'OI*IIY AIDOlt
~
Lupus Lupuo
HSP
.olOll'.•
-ON
E!Jtj """'IliA
E&J¥mld
..-.-
-!lor
COI!Ipsq
Clq ,.pii/'Oj)llhy
QA:G P>m"'*'J)ftlly
MIDI>
E!Jtjclo-
..-
332 I SECTION V: URINARY TRACT
the receptor-associated protein. expressed in the tubular brush border and the
basal surface of the visceral epithelial cells.
LM: Membranous GN is a diffuse process in which the glomeruli are not
hypercellular but usually exhibit thickening of the capillary basement membrane
while maintaining luminal patency. In Jones silver-stained sections, the base-
ment membrane can show "spikes" projecting from the epithelial side of the
basement membrane. Spikes result from the presence of subepithelial electron-
dense deposits (silver stain negative) and deposition of basement membrane-like
material on the sides of the deposits (e-Fig. 19.5). Glomeruli appear essentially
normal in early cases.
IF: Diffuse, granular staining for IgG and C3 is present along the glomerular
capillary loops by IF.. Other immunoglobulins can be present but have lower
intensity staining.
EM: At early stages, the electron-dense deposits are subepithelial. As the
disease progresses, deposition of basement membrane-like material at the sides
of the electron- dense deposits occurs so that with time, the electron-dense
deposits are surrounded by basement membrane and thus becomes intramem-
branous. The deposits eventually become electron-lucent, suggesting resolution
(e-Fig. 19.5).
E. Postinfectious glomerulonephritis {PIGN). PIGN is a classic complication of strep-
tococcal pharyngitis and presents acutely with nephritic syndrome. However,
classic PIGN is currently infrequent; most cases follow staphylococcal skin infec-
tions and other bacterial, viral, fungal, or parasitic infections, and are more
frequently chronic or atypical (Hum Pathol. 2003;34:3).
LM: The pathology is unique, characterized by white cells in the
glomeruli (predominantly neutrophils in the acute phase), and lymphocytes or
macrophages in chronic cases.
IF: There are large granular IgG and C3 deposits along capillary loops (e-
Fig. 19.6). Occasionally, deposits are located predominantly in the mesangium
(instead of in the loops) and are C3 or lgA/IgM instead of IgG (Semin Diagn
Pathol. 2002;19:146).
EM: Shows characteristic bell-shaped deposits (humps). Erythrogenic toxin
type B is thought to be the target antigen for immune complexes that are
implanted in the GBM.
F. Membranoproliferative glomerulonephritis {MPGN). Patients with MPGN can
present with features of nephrotic and/or nephritic syndrome, and most patients
have a low serum C3level. Although MPGN can affect patients of all ages, it is
more common in children. Three types of MPGN have been described; because
all types can have similar histologic findings, EM is used to differentiate them.
Type I is the most common type, followed by types ll and Ill. It is important
to note that MPGN type I can be associated with other diseases such as viral
hepatitis, so patients should be worked up for secondary causes of MPGN when
the pathologic diagnosis is established.
LM: MPGN is a diffuse glomerulopathy with endocapillary proliferation
that results in lobular accentuation (e-Fig. 19.7). The GBMs are thick, and the
capillary lumens are not evident. The Jones silver stain reveals double GBM con-
tours, also known as "tram-tracking." These findings are more commonly seen
in MPGN type I. MPGN type II tends to have a less consistent histologic pattern.
IF: A strong and diffuse granular staining for C3 is observed along the
glomerular capillary walls and the mesangium. Approximately 60% of type
I MPGN also exhibit IgG and/or C1q immunostain. Negative immunostains for
immunoglobulins and Clq are usually observed in MPGN type II.
EM: This is the most useful tool for differentiating MPGN types. MPGN
type I shows mesangial interposition (extension of mesangial cell cytoplasm
into the capillary wall) and subendothelial electron-dense deposits. When the
mesangial cell cytoplasm extends into the glomerular capillary wall, basement
334 I SECTION V: URINARY TRACT
membrane-like material is laid down by the mesangial and endothelial cells, cre-
ating a second "new" basement membrane. This process results in the "tram-
tracking" observed by LM. MPGN type II is also known as dense deposit disease
because it has ribbon-like electron-dense deposits along the capillary walls, often
replacing the lamina densa. These deposits are not necessarily present in all cap-
illary loops and may only be present in some segments and the mesangium. The
composition of these electron-dense deposits remains uncertain. The ultrastruc-
tural findings of MPGN type III are similar to those of MPGN type I, but with
subendothelial and subepithelial electron-dense deposits (e-Fig. 19.7).
G. Crescentic glomerulonephritis (crescentic GN}. The term "crescentic GN" refers
to the presence of cellular crescents in >50% of glomeruli available in a renal
biopsy. Its usual clinical presentation is that of rapidly progressive GN. Patients
can have a renal limited or systemic disease.
LM: Cellular crescents are identified in >50% of glomeruli (e-Fig. 19.8).
Necrotizing glomerular lesions are commonly seen. Additional morphologic
findings are dependent on the type of renal limited or systemic disease.
Immunofluorescence is used to further classify crescentic GN.
IF: In anti-GBM disease or Goodpasture•s disease, glomeruli exhibit smooth
linear IgG staining along the capillary basement membrane. Immune-complex
GN has a granular staining pattern for one or more immunoglobulins. Pauci-
immune GN has negative immunostain for all immunoglobulins.
EM: The smooth linear IgG staining of anti-GMB or Goodpasture•s dis-
ease does not have a morphologic correlation that can be detected by EM. The
glomerular changes noted by EM correspond to the necrosis, disruption of the
GBM, and crescents that are seen in all crescentic GN. Similar findings are
also identified in pauci-immune GN. Cases of immune-complex GN will show
electron-dense deposits (e-Fig. 19.8).
H. Lupus nephritis (LN}. Systemic lupus erythematosus (SLE) is a multisystemic
autoimmune disorder with a peak incidence in the second and third decades of
life and a female predominance (male-to-female ratio of 1:9). It is more common
in African-Americans. The clinical diagnosis of lupus is based on clinical and
laboratory criteria established by the American Rheumatism Association. Renal
involvement by the disease is relatively common; approximately half of lupus
patients develop lupus nephritis during the first year of the disease. Although
this chapter emphasizes the glomerular findings of (LN), interstitial, tubular, and
vascular lesions can also accompany the glomerular changes. Use of the ISN/RPS
classification of LN, which is a modification of the WHO classification (Kidney
Int. 2004;65:521), is recommended.
LM: LN can present with various light microscopic patterns (Tables 19.2
and 19.3). With time and/or treatment, the renal lesion can evolve into a dif-
ferent class. Membranous LN can occur in combination with Class lli or IY.
The following are considered glomerular active lesions: endocapillary hypercel-
lularity with/without leukocyte infiltration and with substantial luminal reduc-
tion; karyorrhexis; fibrinoid necrosis; rupture of the GBM; cellular or fibro-
cellular crescents; wire loops; hyaline thrombi (e-Fig. 19.9). The glomerular
chronic lesions are segmental or global glomerulosclerosis, fibrous adhesions,
and fibrous crescents.
IF: A glomerular IgG-positive immunostain is almost universal in LN; the
term "full house" is used when IgG, IgA, and IgM immunostains are positive.
C3 and C1q are usually present.
I. Diabetes mellitus (DM}. DM is a disorder of carbohydrate metabolism that affects
multiple organ systems. In the kidney, it increases the propensity to pyelonephri-
tis, papillary necrosis, arteriosclerosis, and glomerular disease. Hyperglycemia
in these patients induces biochemical changes in the GBM, nonenzymatic glyco-
sylation of proteins, and hemodynamic changes with glomerular hypertrophy.
Cl't8ptet 19 • Medical OieeM:Ieeot th& Kid""Y I II&
-·
Claal
Claall
Mlrlmal mmnJPI LN
-..,11"'1,.,_ LN
f<><111 LN ~of <!iOlCo
!IleAl
Ill we!
Ill (C)
"""'""I)
fotOI D10llfera1MI LN (DUrei/-IGolonel
Fa pmlllar.o1No and .llderoalng LN (..- ond
dlronle IMiona)
Fa -.uwLN (chronic:-- pmo!Wor
sail)
Claal¥ Dlluoo LN (Aoltfa Oflne- dlluto, ""'"'"""" , _ pobel end-lilly
01\d/ot -li!IBIYQN m.:Mn1 ~or m010 abnoru!D
IV-$ lAlor IV-<l CAl Dlllll.. -GMIII « eSoi:lol pr<lferat.'o LN
IV-S(M:l«IV-GWCl Oll!llaa~Ofgjabelp--and
111-S(C)«IV-G(C) scllrooirc LN
CltftWa ~"' alab<l sdoroolna LN
Mamb111nou.t LN
A<h11>011d ocloroslrc LN (!10!!; or m«e J,bnorUI l)ol>oltyocla.-)
II
Ill lna--
,.,.....,..,ldj
-rc•I .. ·~ ~~~lt:yond
EnOo<:o!illuy and/or
-~~~~IJ pr<lfcnllon In
<50% of alamerull
~+
~and loop+ ,..,.,..! and
tuban-olal d.,....
polymerized proteins forming beta-pleated sheets and stains red with Congo
red.
LM: Varies from GBM thickening to nodular or segmental sclerosis. Amor-
phous material is often apparent in the mesangium or the capillary loops. A
Congo red stain is diagnostic revealing apple green fluorescence under polariz-
able filters (e-Fig. 19.11).
EM: This shows characteristic randomly arranged fibrils of 4 to 12 nm
(e-Fig. 19.11C). It shows either finely granular or filamentous deposits in the
mesangium or the GBM (Semin Diagn Pathol. 2002;19:116).
IF: May show K or l chains. Some deposits of fragments of IgG, IgM, or
lgA immunoglobulin heavy chains or light chains do not form amyloid; in these
cases, a Congo red stain is negative and the term "congo red negative amyloi-
dosis" is used, or the equivalent term "monoclonal immunoglobulin deposition
disease" (MIDD).
The differential diagnosis of MIDD includes other nodular GN such as
diabetes and fibrillary and immunotactoid glomerulopathy. The latter may
have overlapping IF, but EM findings are distinct from amyloid (Kidney Int.
2002;62:1764). The other pathologic findings in MIDD are as follows:
LM: In MIDD, glomeruli may appear nodular or have nonspecific changes.
Diagnosis is usually (but not always) made by IF.
IF: Identifies the specific type of immunoglobulin fragment as either heavy
or light chain UAm Soc Nephrol. 2001;12:1482) (e-Fig. 19.12).
K. AI port syndrome/thin basement membrane disease. Classic X-linked (X-L) Alport
syndrome presents with microscopic or gross hematuria and deafness. Deafness
and/or proteinuria are indications of severe disease. Such symptoms are usually
absent in young children. Mutations in COLIVAS cause X-L AIport; COLIVA4
mutations cause autosomal dominant (AD) and COLIVA3 mutations cause
autosomal recessive (AR) Alport. As many as lS% of X-L Alport patients have
no family history and are thought to represent a new mutation. Patients with
AR Alport are clinically similar to X-L Alport; however, AD Alport is rare.
The involved organs in Alport syndrome reflect the sites where these collagen
IVa chains are normally expressed (a3, a4, and aS are exclusively found in
the kidney, eye, and ear). Renal biopsy is performed for diagnosis as well as
assessment of disease progression.
LM: This is not specific; it varies from unremarkable glomeruli, to FSGS,
to chronic scarring. Foamy interstitial cells were once thought characteristic of
Alport, but they are in fact seen in many types of proteinuria.
IF: Routine stains are negative. Diagnosis of Alport is facilitated by collagen
a3-S chain immunostaining. The aS chain of collagen IV is distinctly absent
in X-L Alport, and it is accompanied by the absence of a3 and a4 chains in
glomeruli. (Collagen IV monomers are incorporated into basement membrane
as triple helices to form the structural meshwork [Kidney Int. 2004;6S:1109];
when one chain is mutated, all three chains may degenerate because they become
susceptible to enzymatic proteolysis.) Typically, women with X-L Alport have
mosaic linear staining with collagen IVaS in the glomeruli or skin (e-Fig. 19.13)
(Hum Pathol. 2002;33:836). However, some women may have positive stain-
ing, making it difficult to distinguish from thin membrane disease (TMD). A
screening test for families with potential X-L Alport is skin biopsy (IVaS is the
only chain found in skin); the diagnostic pattern is absence of collagen IVaS in
men and mosaic staining (linear interrupted positivity) in X-L women carriers
(e-Fig. 19.13B-D).
EM: The cardinal pathologic findings ofX-LAlport are seen on EM and con-
sist of abnormal splitting, widening, or thinning of the GBM with degeneration
of collagen IV in the lamina densa known as "bread crumbs" (e-Fig. 19.13).
The lesions may involve tubular basement membranes as well. However, the
Chapter 19 • Medical Diseases of the Kidney I 3 37
C. Allergic interstitial nephritis. Most cases of allergic interstitial nephritis are drug
related; renal manifestations develop "'2 weeks after drug exposure. The typ-
ical presentation includes feve.t;, skin rash, and eosinophilia, but this triad is
only present in approximately one-third of the patients. Interstitial eosinophils
are the most important histologic clue for diagnosis. Eosinophils are accompa-
nied by other inflammatory cells, mainly lymphocytes, and may only be seen
focally (e-Fig. 19.15). Tubulitis and tubular injury usually accompany the inter-
stitial changes. Nonsteroidal anti-inflammatory drugs (NSAIDs) can also cause
allergic interstitial nephritis, but interstitial eosinophils are usually rare. More-
over, patients taking NSAID can also develop nephrotic syndrome, and their
glomeruli exhibit changes indistinguishable from MCD.
D. Cast nephropathy. Cast nephropathy is a renal complication of multiple
myeloma. Casts are usually located in the collecting ducts, but due to retro-
grade filling, casts can even be seen in the proximal tubules. Casts are composed
of light chains and Tamm-Horsfall protein, and tend to have a fractured
appearance. A granulomatous response with multinucleated macrophages and
reactive epithelial cells is typically found at the periphery of myeloma casts
(e-Fig. 19.16); other inflammatory cells, such as neutrophils and lymphocytes,
can also be part of the inflammatory process. Nonspecific changes such as
tubular atrophy and interstitial fibrosis are commonly present. IF studies
exhibit a monoclonal light chain in the casts in most cases.
IV. VASCULAR DISEASES
A. Vasculitides. The kidney has a dense arterial, venous, and lymphatic network.
Vasculitides affect all these vessels, but most commonly the arteries. They are
usually classified as large, medium, and small vessel vasculitides on the basis
of the predominant vessel size affected, but although the classification is con-
venient, it is controversial because there is variability in the size of affected
vessels. Vasculitides are also classified by the underlying pathogenetic mecha-
nism as either immune-complex mediated or pauci-immune (absence of immune
deposits). The following are the most common vasculitides affecting the kidney,
grouped by size:
1. Takayasu's aortitis affects large branches of the abdominal aorta and the renal
arteries. Mural inflammation with giant cells is the distinct feature.
2. Polyarteritis nodosa, scleroderma, lupus, rheumatoid arthritis, and Wegener's
granulomatosis affect medium- to small-sized vessels. These are systemic dis-
eases and, with the exception of lupus, are pauci-immune systemic vasculi-
tis. Kidney pathology is variable and includes interstitial granulomas, arte-
rial acute inflammation, fibrinoid necrosis, and thrombotic occlusion. The
inflammation can lead to aneurysm formation or fibrous wall thickening.
Involvement of glomerular capillaries leads to crescentic GN. The diagnosis
largely depends on the clinical syndrome, not the pathological findings. For
example, Wegener•s granulomatosis is associated with serum autoantibodies
against components of neutrophils known as circulating antineutrophil cyto-
plasmic antibodies (ANCA). These antibodies correlate with disease activity
and underscore the pathogenesis of vascular injury (see Chap. 9).
3. Of the small vessel systemic vasculitides, some are referred to as thrombotic
microangiopathies(TMA). A typical presentation is with hemolytic anemia
and thrombocytopenia, known as hemolytic uremic syndrome (HUS). Enti-
ties included under TMA are diarrhea associated (classic HUS), thrombotic
thrombocytopenic purpura, eclampsia, pre-eclampsia, thrombosis due to
drugs (oral contraceptives, chemotherapy drugs such as bleomycin), cryo-
globulinemia, idiopathic hypereosinophilia, antiphospholipid syndrome,
hereditary deficiency of blood clotting factors, and malignant HTN (] Am
Soc Nephrol. 2003;14:1072). Renal biopsy shows arterial intimal thicken-
ing (called mucoid degeneration) and luminal fibrin thrombi (e-Fig. 19.17A).
Chapter 19 • Medical Diseases of the Kidney I 339
All of these entities may also show characteristic arteriolar thickening (also
known as onion skinning, e-Fig. 19 .17B), endothelial swelling, and/or lumi-
nal thrombosis.
B. Systemic HTN. Vascular diseases of the kidney are often associated with systemic
HTN. The most common cause of HTN is idiopathic, which, despite modern
pharmacologic therapies, remains a primary cause of renal failure. High blood
pressure damages parenchymal arteries causing wall thickening; the arterioles
show characteristic acellular eosinophilic material due to increased intravascular
pressure and endothelial injury that allows leaking and deposition of plasma pro-
teins. Glomeruli beyond damaged arterioles undergo sclerosis (e-Fig. 19.17B).
1. Malignant HTN, defined asdiastolic pressure> 120 mmHg, complicates ""10%
of patients with benign HTN, presents acutely with papilledema, nau-
sea, vomiting, convulsions, and coma. Renal function declines rapidly and
patients develop gross hematuria, albuminuria, and hemolytic anemia. Onion
skinning is characteristic (e-Fig. 19.17C).
2. A small fraction (5% to 10%) of benign HTN is secondary to specific
causes, some of which may be amenable to surgical therapy, that together
are known as renovascular HTN. Most common is renal artery stenosis
due to atherosclerotic aneurysms in older mostly diabetic men. Patients with
abdominal or renal artery atherosclerosis may present with acute renal failure
or proteinuria due to cholesterol embolism in parenchymal arteries (e-Fig.
19.17D). Fibromuscular dysplasia, an idiopathic mural thickening of the
renal artery, is seen in young women (Am] Kidney Dis. 1997;29:167).
V. PATHOLOGY OF THE ALLOGRAFT KIDNEY
A. Processing of renal allograft biopsies. Renal biopsy is a key component of man-
aging transplant recipients since clinical diagnosis is changed in rv40%, and
therapy in ""60% of cases following biopsy (Am] Kidney Dis. 1998:31:515).
Transplant biopsies in most centers are performed when clinically indicated
(indication biopsies) or at standardized time intervals (protocol biopsies). Crite-
ria for indication biopsies include proteinuria, acute renal failure, and increased
creatinine levels, among others. Adequacy criteria of the renal allograft biopsy
were established at the BANFF 1997 Consensus Conference (Kidney Int.
1999:55:713).
It is recommended that two 16- or 18-gauge needle core biopsies are sub-
mitted for pathologic evaluation: one for LM and the other divided for IF and
EM. The core for LM is submitted in 10% buffered formalin; the IF core should
be submitted in refrigerated transport media; and a small fragment from cortex
should be submitted for EM fixed in 2% glutaraldehyde. For LM, 10 glomeruli
and 2 arteries are required for definitive diagnosis; 7 to 10 glomeruli and 1
artery are considered marginal, while <7 glomeruli and no arteries are an inad-
equate sample. The paraffin blocks should be sectioned at 3 to 4 Jtm thickness;
seven slides including three H&E stains, three PAS or silver stains, and one
trichrome stain should be prepared (Kidney Int. 1999;55:713). For IF, a min-
imum of two glomeruli is required for the evaluation of glomerular disease;
however, IF evaluation for IgG, IgA, IgM, C3, fibrinogen, albumin, and C4d is
standard. For electron microscopic evaluation, at least one glomerulus is rec-
ommended. See Table 19.4 for BANFF diagnostic categories for renal allograft
biopsies.
B. Transplant rejection. Major complications with specific pathologic findings in
the allograft biopsy are acute and chronic rejection (which are subdivided into
cellular and humoral rejection), and recurrent and de novo disease (see Table
19.2).
1. Acute T-cell-mediated rejection. Tubules, arteries, and the interstitium are
the principle targets of acute T-cell-mediated rejection. The BANFF 2005-
updated classification of renal allograft rejection (see Table 19.2) is based on
aco I SECTION v. URINARY TRAel
1'1,1,!' JttS 1~;:.w ~ cu,or~u for ~~o~n~~ Alh~'lft llloP'IIeHI-" ' "
1.-1
2. Arili>ody<nod- ro)od!on
Quo lo clocumon1Dd ...-n., ..tlbocl;r, C4it 1nd .,q1lll prll>i>l:v
kl/1» •Mibo<tr'-rnodllllod reJedloh
'l'fl>oo (plo)
I. ATN-II<.t m!rlmalln!lam-, C4<1+ In portll.JbiJ~r ..plliulo!; ()"'''C)
11. C.pililly and!« P>mot\llu ln1!11!1mrllon anc!lof llin>m-., 04d+ In PTC
Ill. Al1oulll- .e, C4<l+ In f'TC
Chronic anlllloctl-- ,.,_,
04d+, p.-ceol d1Wo1fnuh1Hioll« •hllbodilo, ..,.Illlor dou'*> -ll'e•hdlor
DOII!ubulorCIIDiaiY- memb11110 muhllo>iol1,. arod/of ln1orodlol ~brooM!Jbular
....plly. .d/orlbiOI.Illr-.t""l thldcln!ngln •~ta~lol&
a llml•lht ella- •swpl&:lcus" ftlf""""' TGiklwfllled r<ljeclbn
Thl& ...,....,.. uNd.m., no lnllmala-1& p,_nt, but lllor•a1'6fad ofblb~ll Ql, 12, «
13) 101111 mlnllf ln1Dmlllollnflomtnlltion (10 Of II)"' ln1Drlllllol!nllomrn<11on will mill tubuli>
lUl.tl <4, 12•4-10, G;e: 10intoeokh..,lt,m~pertobularo,_-.
4. N.ob T....a.l!lfdo1ed relodbo
'l'fl>oo (plo)
lA. SfVII!aonl 1-llnllllmDI(>2S%!>'~1,12orla)and!Odai..-111A
blbullla (12)
18. _,lloant ln1Dmlllollnfllrwllon (>20% pnndrjmo.l2« G) •nd fad of -..tllb!Jlll&
!13)
IIA. Mid'> 111Cdemeln1imo10Mtll4 Ml
liB. s - . 1 - •"'ltii<Om1Jflst\& >25% ollte klmlnal"""' (.:!)
Ill. 'IIINmurol-ltllftlditlfanonallbo!nold ""'•IIO•rod .-otmedlal.srnooth m.-
CIIIIIwttll .....,poft)1ngl.lmplloq1lc !nlllmrnlllon (v3)
Chronic T.....modiii1Dd r$CIIOn
~blooll.- · .,_ol
Chronic aJoa7of\ artcrlo!oallr.r (u!crlellnllmolfllmibwttll monon....., ... lnfl-ln
---·-··
Tubulllo
......
-~olllll)
fSGS
D~
•-•
r,. mjillnt' '""rulopctlly
fSGS
--
Gbnonlllll IJU>Uo D~ta.
""l1dlol ht1111nma1Soh l.i!'A Ami-GBM
MPGN
Momtnnouo Momlnl"""
V...Cullla (ANCA, ...u.GBM) -
Am~Wrtc:Ntt
--
doi>OOitlon <loou&
~•lie C4i II'NIIIIIIIeil 1\Diloll_,..
I.-!ln1\al\'ll"don 1.1)4sionll <Sit Mpl\~ Throm- mlc,..rclopotlty
Intlm1111b~ (di\IIJind.-)
In.-r11brosll
Ctlo-.1 ombolorn
Reno! erteiY 11\rombooh
Ronii•Inlllrombas&
AnU~ rejodlon
Tl..tUDli-..ttlil
Ro<:um>l'lt HUS A<altain11- •~~>hi1111
Chronic: Wllltr d11.111..:ilon (oof!J.
111.21)
O>fliota<t't'Oialo
CU:Iurn phool>ila1o <tW.olo
....'"""
ATN
M_,lllt tJrnDhcmo
f>TU>
Chapter 19 • Medical Diseases of the Kidney I 34 3
NORMAL ANATOMY
In the adult, the normal kidney weighs about 115 to 155 g in women and 125 to
170 g in men. Anatomically, the kidneys are composed of an outer cortex and an
inner medulla that has 8 to 18 pyramids. The base of each pyramid is at the corti-
comedullary junction, and the apex forms a papilla where the collecting ducts open
into the renal pelvis. The minor calyces receive the papillae and in turn join to form the
major calyces that are the dilated upper portion of the ureter in the renal pelvis. Histo-
logically, the components of the kidney include glomeruli, tubules, blood vessels, and
interstitium.
Maldevelopment and
Nonneoplastic Cystic Diseases
Johann D. Hertel, Peter A. Humphrey, and Helen Liapis
The developmental abnormalities and benign cystic diseases of the kidney that are most
likely to be seen by the surgical pathologist are presented here.
I. DEVELOPMENT ABNORMALITIES
A. Renal dysplasia is seen in malformed kidneys where there is abnormal differentia-
tion of metanephric elements. Most cases are unilateral and sporadic, but a wide
variety of genetic diseases, malformation syndromes, and chromosomal disor-
ders have been linked to renal dysplasia (Stocker ]T, Dehner LP, Husain AN,
eds. Stocker and Dehner's Pediatric Pathology, 3rd ed. Philadelphia: Wolters
Kluwer/Lippincott Williams and Wilkins, 2011). Bilateral renal dysplasia is less
frequent and is associated with renal failure at birth. Renal dysplasia is a com-
mon cause of an abdominal mass mimicking neoplasia in children < 1 year of age
but can also be diagnosed in older children and adults. There is an association
with congenital urinary tract obstruction in about 50% of cases.
B. Grossly, multicystic dysplasia is characterized by a slightly enlarged kidney, or
a small and irregularly cystic kidney (e-Fig. 20.1)* Aplastic dysplasia is typified
by a small, solid remnant of the kidney. Segmental dysplasia occurs when the
collecting system is duplicated; microscopically, there are immature tubules or
ducts surrounded by collarettes of condensed mesenchyme (e-Fig. 20.2), and
islands of immature-appearing cartilage (e-Fig. 20.3 ), cysts of varying sizes that
have a flattened epithelial lining, and islands of normal glomeruli and renal
tubules between the dysplastic areas. Rare cases of Wilms tumor and renal cell
carcinoma (RCC) have been reported in multicystic dysplastic kidneys.
*All e-figures are available online via the Solution Site Image Bank.
344
Chapter 20 • Surgical Diseases of the Kidney I 34 5
SUGGESTED READINGS
Agarwal MM, Hemal AK. Surgical management of renal cystic disease. Curr Urol Rep. 2011;12:
3-10.
Bonsib SM. The classification of renal cystic diseases and other congenital malformations of the
kidney and urinary tract. Arch Pathol Lab Med. 2010;134:554-568.
Eknoyan G. A clinical view of simple and complex renal cysts. JAm Soc Nephrol. 2009;20:1874-
1876.
Pediatric Renal Neoplasms
J.son A. J~rzemboW'Ski 1111d Fl'llnCC$ V. White
...""'""""''
......
......,
Clalllo • • II Ill
CIUJllri'IIIDMMI:fe
. . . ~.~~
7d
4mo
-~~- 00
I
3
... .rottllllf• (J)
C>-2
().2
-
nepfl It
IIIII ...... -~~~--
18,.., 2 I).$
•• u t ..
2y
36.5 mo !bol'l)
4
...
~..,.-9yol
'--·--
lla"' C>-18+
lOy <5 1-18+
,.'""" .:c Wide,.,.. <5 1-18+
.........
-·--..a·-
~
~.--
....
41y
221
SOy
llano
llano
llaro
IS-Ill+
11-18+
17-111+
tumors; howevet; intraoperative biopsies are discouraged due to the risk of tumor
spillage unless the diagnosis will alter operative management.
Pediatric renal tumors are often large, friable tumors that bulge beyond the nor-
mal renal contour. The capsule should be carefully examined for sites of rupture
and inked before incised. The initial plane of section is taken to demonstrate the
relationship of tumor to the capsule and renal sinus. At this point, fresh tissue from
each tumor nodule, nephrogenic rests, and normal kidney is snap frozen for proto-
col studies. Tissue may also be taken for cytogenetics, flow cytometry, and electron
microscopy, depending on clinical history. Following initial cuts to facilitate fix-
ation, overnight fixation is recommended before histologic sampling to decrease
tumor friability and capsule retraction. Tissue for histology should be mainly taken
from the tumor's periphery, as histology of the interface between tumor and renal
parenchyma is often important in identifying the type of tumor. Each separate
tumor nodule should be sampled, with at least one section per centimeter of tumor
diameter. Sections should also demonstrate the relationship of tumor to the capsule
and the renal sinus. The hilum is a common route of tumor spread, necessitating
adequate sampling of this area as well. Ureteral and vascular margins, hilar lymph
nodes, nephrogenic rests, and normal kidney are also sampled. Tumor sections are
mapped, using either a diagram or photograph of the specimen; mapping is nec-
essary for nephroblastoma, as the presence of diffuse versus focal anaplasia alters
type of treatment (Arch Pathol Lab Med. 2003;127:1280).
Ill. DIAGNOSTIC FEATURES OF PEDIATRIC RENAL TUMORS. Pediatric renal neoplasms are
notorious for their variable histologic patterns, which often show overlap between
the various entities; thus, there should be careful examination of the tumor-kidney
interface for pattern of infiltration, which is characteristic for each tumor type.
Correct diagnosis depends on knowledge of both classic and variant histology,
along with adequate sampling and correlation of the microscopic findings with
patient age and other clinical information. While immunohistochemical studies
may be useful for establishing the diagnosis (e.g., for INI1 in rhabdoid tumor) or
for excluding a diagnosis in specific cases, immunohistochemical stains are usu-
ally not needed. Molecular studies can also be used to confirm specific diagnoses
(Table 20.2).
A. Nephroblastoma (Wilms tumor) and nephrogenic rests. Nephroblastoma (Wilms
tumor) accounts for 85% of pediatric renal tumors. Most patients present before
10 years of age, with a peak between 2 and 5 years. Rare examples have been
reported in adults (] Clin Oncol. 2004;22:4500). The tumor is more common
in children of African descent than of other races and is slightly more common
in girls than boys.
Although genetic abnormalities have been identified in only a minority of
nephroblastomas, multiple genetic abnormalities and syndromes are associated
with the tumor. Abnormalities of WT1 on chromosome 11p 13, a gene involved
in renal and gonadal development, occurs in aniridia and genital anomalies
syndrome (WAGR), Denys-Drash, and Frasier syndromes, all associated with a
high risk for nephroblastoma. Molecular alterations of imprinted genes at the
WT2 locus on chromosome 11p15 are associated with Beckwith-Wiedemann
syndrome, which has an increased risk for nephroblastoma. Both WT1 and WT2
locus gene alterations are found in a minority of sporadic tumors. About 1%
of patients with nephroblastoma have a family history of nephroblastoma; two
familial genes, FWT1 and FWT2, have been identified in this setting in which the
disease shows autosomal dominant transmission with variable penetrance (Cu"
Opin Pediatr. 2002;14:5). Other molecular alterations that are associated with
tumor progression or aggressiveness have been identified; TP53 is implicated in
progression to anaplastic nephroblastoma, and loss of heterozygosity of 1 p and
16q is associated with poor prognosis in favorable histology nephroblastoma
(] Clin Oncol. 2005;23:7312 and] Clin Oncol. 2006;24:2352).
I~!.HI
,._
K_,.MIIIIt•
crt.MIIe ...lftl!IIJ
li~IS,llplS,orl!llll.l
""""' ......
El•!J! c:.omm.. Mol111111r ~lflllltl II Ptcllltlle IIMIII NiloplllrM
l'lallll ft . . .
Dlliliii:>Mor ~no '""'~I lm. WT.i?. lm. 1m( 1JIIC:·ft- DNI\-bhlftl-1"'
llm>rSII~--
I POX tlr WT1
Tl-·· •••-ct I
22q11.2
t(l(;l)(p112;q34)
t(l(;I) (p112;q25)
ASPI.·TI'f3
PSF·11'f3
~~~-pllonol ~·
T111'111C1fptionoi-B ~·
~111LIPOXIorTFE
•nd~K
t(X; l)(p11.2.-q21) Pf!CC·TI'f3
t!6:11){~1;ql2) Ntihi>-TFEB
.... ....,...,ewclru• C<l\ldullonolll~IS.S HBB Hatrlllllloli1 $ IPOXIoriNII
m - . ; po!lllblo 22ql1.2 hSNF'S/IN/1 Cllromllt! ramoclolnrn
AieL ;c.,._ ~.
-.- 16!113.3, 5q mW!tw l!CI, 1SC2
hl'llltlfpti>r!ol ~·
Tumor tuppraaor...,... ~llof)'Oiypo
....
IPQX, 111111- RT-PCR, ........ cnu•<flpll:>h1101J1ro-cl!eln- ASH, n u - l n ol\u hol>l1di.tlrll:>n; PNEI", l'fl.-.......-.nal 11/mor:
CI:Sll. oloor<OI ..""'""'<llltirot. COn\plod hom Reloton"" InS\/...,.-~
.......
350 I SECTION V: URINARY TRACT
m-
Tl.mor ..,..,. of •ny• - " ' dlwpllon ocallfiJ11Iilloreor duJti.ltuiPY
TJ.mor ••ncl'llad In mora Uwl one pb::e
Sllpsy ~ atrt """'11>"""""'1
H HllnOI-OUO m...a ... ~rw. IYof.l>ono. lnl11, *-l
cymph nOISe nvst•WM ~ ttt•.abdOI'I'IIIf'l (lr l)f!IMl
'I S-renoltl-atd"""""*
sr..,.- --rllli)P
ModlledfiOili_Dtv,_~,
"'*"·-·-
M- "'""""will foil into tho·~· .,U. w:ich lllllrt<n.tl"""""'"' aad
do.-lc U'f:p.bulc d......, to oboctmllll tlo: .rcn>•lnfn;s vlohlo-, !~Ada- .....
tc111 ptOtocol.o, thcoo pctlc.atol'CCC!R u adcl!doaal 6 '19\lCb <i ch~ ...PJ.
Lt. ~IJCD!Ir. I\I!XI.Ort -111it1W (l()!Xlpk(c _ . . , ;, whi<:h cut tuq;ical
lftlldio11""" bepafo..-.1 w:itbautadJiiticm.dcho:moth.erApy. JW:.psy~
4em ODitlati.q WOuisome histntotPc fu.tu:ra such AI prt.dunin ID '* f1l 'b:l.o..naDaJ
o(...,.,,. or -luio oftc:r iaili&lllU1m:llt ""' nri1che4 to """'' -mitt
dl..c::m.Dcherapeu1ic ngi ram'
t. lllliiiMplrfo 111•11 arc wcl.hl!ffuclllltcd IICI'hto!llu1lc - """''''"'"'
nryiog proportioaf of q>iche&l ....t ~ cdla. ~ ll:!m<Xrt ue
beftigll; ~bod! ~bluci>D14 and p•pillaq> RCC ht.vc beta rcpottcd
iD ll:!mOrt with t:pith<lial elm-.
1. M ••JIIIe llr.aJ III•DFI 001'1) CCC1Il' t!uov,shov;t childl.ood, lro.c-
""""""l'ly ill iDfaa<y (..Pis- 20.13}. lfill»zi<>ally, tlu:se """""' ......, .,..,.
a!dctcd to be """"'h.Wdc .D.ePlu:omu (MN) tdloco....t later) bm uc .D.aW
..,.cbod a dlrtiDct ca1!ty, MST It ......U:, aollluy ....t c:xltl!da out flom.
cht; ~ ~ U .,_ ~·-Pf'!llatcd 111U1 tht.t.ln9' be aolid Ot eyt1i(:.
Tbc 1M""""""' i& li!m .....! " ' f - ~p;.:..!ly, the <IIDIJ>r i& .,..,.
poud.ofspilldled .,..,.u... ..U. w:ich jdisrinct.,.,.plum. U.ader l.,...p....,
mX:!ooa>py, cht; - hu. diwri!l<X Jll>da1ar · - clue ... .Jtem.t-
lllg atel.t <i hn-oo:Jlidodty and ~. 11lc tumotl.t · - -
latcd, and bolldo of t11m0t cello Cl:la>d outwud to ccttap adjaOCIIt ~
w cub'alea, ru'll&iagi». cyN"""q>i!belid~ 4IDII ~
352 I SECTION V: URINARY TRACT
-
lint Jtll••..t 10 Vlll>l&lumor rnusl..,l)lla»113 of tilt tumor mtn
At lllalt213of ll11ulolilo lllnv oonslsb d -ma
011\et 00111~ of IIOI>hrobiMiornoll'lll!' be ltllOOirt In
""llln1 p,po-
10
......
Tl.mOilrllmOII.IIr1ibli 1116 usual Cl1t.wl.t of,...lor dlfflll&
Tumor,
cortex and Tumor and renal
perirenal fat ~~-:---=--!=----pelvis ± renal sinus
to include
radial margin
or Gerota's
fascia Transverse
section of
hilar vessels
·- - - Section of distal
l==~ ureter
T2b
···r··- -
>10cm
_ _ L_ _ _
Figure 20.2 RCC pathologic primary tumor (pT) stages. (Modified from: Edge SB, Byrd DR,
Compton CC, Fritz AG, Greene FL, Trotti A, eds. AJCC Cancer Staging Manual. 7th ed. New
York, NY: Springer; 2010.)
for tumor involvement of the perirenal soft tissue are inked selectively. Occa-
sionally, lymph nodes are found in the hilar region.
The kidney is then sectioned sagittally. Tumor size, location (upper or lower
pole, cortex, or medulla), involvement of calyceal or pelvic mucosa, invasion
of the capsule or perirenal soft tissue, and involvement of the adrenal gland
are recorded. The uninvolved parenchyma is also examined: color, cortical
thickness, additional focal lesions, and renal pelvis are described. One section
per centimeter of tumor, demonstrating its relationship to adjacent capsule,
renal parenchyma, and pelvis are submitted, as well as sections of any addi-
tional lesions, uninvolved renal parenchyma, and adrenal gland (Fig. 20.2).
Nephroureterectomy for urothelial carcinomas is described in Chapter 21.
II. DIAGNOSTIC FEATURES OF COMMON TUMORS OF THE ADULT KIDNEY
A. Carcinoma. RCC arises from the epithelium of the renal tubules and represents
.....90% of all renal malignancies in adults.
1. Risk factors. The most important risk factor is tobacco smoking. Additional
risk factors include obesity, hypertension, unopposed estrogens, and expo-
sure to arsenic, asbestos, cadmium, organic solvents, pesticides, and fungal
toxins. Patients with tuberous sclerosis, chronic renal failure, and acquired
cystic disease of the kidney have an increased incidence of RCC. About 5% of
patients with acquired cystic disease of the kidney develop renal cell tumors
(see section on Acquired Cystic Disease-associated Renal Cell Carcinoma).
Most RCCs are sporadic, although there are several inherited cancer syn-
dromes that affect the kidney (N Eng J Med. 2005;353:2477), as follows.
a. Yon Hippel4.indau {YHL) disease is inherited in an autosomal dominant
manner and is characterized by hemangioblastomas of the cerebellum and
retina, clear cell RCC, pheochromocytoma, pancreatic cysts, and inner ear
tumors. Typically, renal tumors are multiple and bilateral (e-Fig. 20.20).
Numerous renal cysts, lined by neoplastic clear cells (e-Fig. 20.21), are
also characteristic. This disease is caused by germline mutations of the
VHL tumor suppressor gene on chromosome 3p25.3.
360 I SECTION V: URINARY TRACT
_.......
~ i (J:] I J t•I:JI WHO Hlttololfc Cllutflr:l.tbl•t 1-.. of tM IUcfMJ
CIMrcolroNicoiClldnoma
MLIIIoculor <lolr col r<Nicol Clldnoma
llliill,pwrt ftlln>u& hlsllat)1oma
H..,.nJIIopellcytorill
O&oMn:ome.
PoDIIo!Yrenll col corel1101110 Ali.l.toln'l'>l-.ol!lll
Ch1011101>11o1Jo llflll col CI/Cinoma ~llold .........,,..1)'<11,_
a..mma ofllle edledlng duel:a d Bo!lllnl
R. .l """'*'III:Ycardnoma
...............
H""'npmo
)(plllrln-n - - ~plionpma
a..mma- \llln nauroblutoml J~~rcoltlmo<
Muclnouotlbulor •nd Olindb"'ll"'"*'..,• Ronomoclulluyl-lcoltlmo<
R""'l "'llco"*>oono.lll<luollod Sci\WoN\01110
-....,.,...-brtma
l'oDiio!Y-..
~·
Mecln01>hllc edehol!lll
Sclbry ftbrtaiULmor
lllllol- ................._
C::,.UO nap/roms
lllxad el>tilel~l•hd otrcmollllmct
.....,.... _..
Meoln01Jhllc syr....w ..rcomo
Meoln01Jhllc ltiOrnol tumor
"'••I
Quclnold
;Mt.cn
_
Cia", .. ...,.~. ~pllomo
c::loorcol-• l.aukeml!.
Rhobdoldtumor
Q>npnllol "'""'blulle Mj:t\IUM
(lai'Jing nwltlmtr or .ml!la
.......
'"'''""·-""
Tai'III:Hne.
IIIIo •lii""'""*-
Ldomyos~"""""(lnQidi'1Jnrlllwln)
Olo-11'1011'11
An....._....
Rhobdo.,.,.,.._,.
au I SECTION Vt URINARY TRACl
,...,
~i(.1:!1Jt•I.:JI .,..DpMnQrt ofbnaJ TUrMfS
llllbr
ROCMo
PAX2
CDIQ
- -
CINr c.ll
+
+
+
+
+
+
care-
f)lillbe l.CC
-1+
-1+
-1+
-
OKGI:f·
+
llf11
+
+
Ill*
+1-
+
0
ICC
ti/A
HIA
has been proposed that multilocular cystic RCC is a subtype of dear cell
RCC (Mod Pathol. 2010;23:931). No recurrences or metastasis have been
reported for this tumor type.
c. Papillary RCC comprises about 10% of RCCs. Grossly, the tumor can be
cystic, hemorrhagic, and necrotic, with a fibrous pseudocapsule (e-Fig.
20.33). Multifocality and bilaterality are more common than for other
RCCs. Microscopically, there is a papillary or tubulopapillary architecture
composed of papillae with thin fibrovascular cores which may harbor
aggregates of foamy macrophages (e-Fig. 20.34) and cholesterol crystals.
There are two types of papillary RCC. Type I tumors have papillae
lined by a single layer of cuboidal cells with scant cytoplasm (e-Fig. 20.34)
that has a basophilic quality. Type II tumors have taller cells with more
abundant eosinophilic cytoplasm, higher nuclear grade, and pseudostrat-
ified nuclei (e-Fig. 20.35). Fuhrman grading may be applied (Am] Clin
Pathol. 2002;118:877), but nucleolar grading may also be useful (Am]
Surg Pathol. 2006;30:1091). Type I papillary RCC is often positive for
vimentin, keratins detected by AE1/AE3 antibodies, CK7, AMACR,
and RCC Ma; immunostains that are usually negative include CD117,
kidney-specific cadherin, and parvalbumin. Type II RCCs have a vari-
able immunophenotype (Arch Pathol Lab Med. 2011;135:92). Geneti-
cally, there is characteristic trisomy of chromosomes 7 and 17, with loss
of chromosome 4, but chromosomal analysis is not used diagnostically.
Papillary RCC, especially type I, has a better outcome than clear cell RCC.
d. Chromophobe RCC accounts for ""5% of RCCs. Grossly, chromophobe
RCCs are well circumscribed, solid, and beige to light brown (e-Fig.
20.36). Tumors are composed of pale polygonal cells with prominent
cell borders, often wrinkled nuclear membranes, and perinuclear halos
(e-Fig. 20.37). The cytoplasm can be finely granular or flocculent (e-Fig.
20.38) to eosinophilic (e-Fig. 20.39). The use of Fuhrman grading is con-
troversial (Am] Surg Pathol. 2007;31:957; Mod Pathol. 2009;22:524).
Diffuse cytoplasmic positivity with Hale's colloidal iron is characteristic,
and these tumors show extensive aneusomy with loss of chromosomes 1,
2, 6, 10, 13, 17, and 21 which can be detected by FISH (Mod Pathol.
2005;18:320).
Hale's colloidal iron and FISH can be used to address the differential
diagnosis of the eosinophilic variant of chromophobe RCC versus onco-
cytoma. For the differential diagnosis with clear cell and papillary RCC,
immunostains can be helpful, although H&E slides are typically diagnos-
tic; immunopositivity for kidney-specific cadherin, parvalbumin, CD117,
EMA, keratins (detected by AE1/AE3 antibodies), and CK7 is typical,
with negative immunostains for vimentin, carbonic anhydrase IX, and
AMACR (Arch Pathol Lab Med. 2011;135:92). Chromophobe RCC has
a much better prognosis than clear cell RCC.
e. Collecting duct carcinoma {CDC) is thought to arise from the collecting
ducts of Bellini. It accounts for < 1% of renal cell tumors. These tumors
are centered in the medulla, have tubular or tubulopapillary architecture,
and have a surrounding desmoplastic reaction (e-Fig. 20.40). The tumor
cells are typically of high nuclear grade and can have hobnail cytology. Use
of immunostains in diagnosis has limitations; when needed, a-methylacyl
coenzyme A racemase (AMACR), CD10, and RCC Ma negativity would
favor CDC over papillary RCC, which figures prominently in the differen-
tial diagnosis, and which would usually be positive for these three markers
(Semin Diag Pathol. 2005;22:51). Immunopositivity for PAX2 and PAX8,
with negative immunostains for p63 and uroplakin Ill, would favor CDC
364 I SECTION V: URINARY TRACT
single central nucleoli (e-Fig. 20.54 ). Scattered larger cells with a higher
nuclear/cytoplasmic ratio and hyperchromatic nuclei can be seen (e-Fig.
20.55). Smaller cells with scant cytoplasm (known as oncoblasts) can also
be noted in some cases (e-Fig. 20.56). A hyalinized or edematous stromal
background is frequent. Microscopic (but not gross) extension into perire-
nal fat and vessels has been described. Rare cases of numerous oncocytic
tumors (oncocytosis) have been described. Chromosomal abnormalities in
oncocytoma include t(5;11) and loss of chromosomes 1 and 14, but assess-
ment for these abnormalities is not usually necessary. No cases of death due
to metastatic disease have been reported.
C. Metanephric tumors (see also the section on pediatric renal neoplasm)
1. Metanephric adenoma occurs in children, and in adults during the fifth and
sixth decades. It is more frequent in women. About 50% of cases are inci-
dental. Tumors are usually well circumscribed, gray to tan to yellow, and
firm or soft. Hemorrhage, necrosis, calcification, and cyst formation are
common. Microscopically, metanephric adenomas are composed of small,
uniform round acini with small lumens (e-Fig. 20.57). The cells are uni-
form with small nuclei and inconspicuous nucleoli. Branching and tubular
configurations are common, as well as a papillary architecture with numer-
ous psammoma bodies. In the differential diagnosis with papillary RCC,
immunostains that are positive for WT1, negative for EMA, and focally pos-
itive for CK7 favor metanephric adenoma.
2. Metanephric adenofibroma is more common in men. Grossly, it is solitary and
partially cystic. Histologically, its cells are similar to those of a metanephric
adenoma but are embedded in a stroma of fibroblast-like spindle cells. Psam-
moma bodies are also common.
D. Mesenchymal tumors
1. Leiomyosarcoma is the most common renal sarcoma; it occurs mainly in adults
and affects women and men equally. They can arise from the renal capsule,
parenchyma, pelvis muscularis, or the renal vein. They are solid, gray-white,
and focally necrotic. Histologically, these tumors are composed of spindle
cells with a fascicular growth pattern (e-Fig. 20.58). Necrosis, nuclear pleo-
morphism, and numerous mitotic figures indicate malignancy. Leiomyosar-
coma is an aggressive tumor with a 5-year survival rate of 29% to 36%, and
most patients die within 1 year of diagnosis. Sites of metastasis include lung,
liver, and bone. Sarcomatoid RCC is in the differential diagnosis and is more
common, and so must be excluded; diffuse desmin and smooth muscle actin
immunoreactivity would favor leiomyosarcoma.
2. Rare primary sarcomas include RMS, angiosarcoma, malignant fibrous histio-
cytoma, chondrosarcoma, low-grade fibromyxoid sarcoma, malignant mes-
enchymoma, and osteosarcoma.
3. Angiomyolipoma is a benign clonal mesenchymal neoplasm tumor com-
posed of thick-walled blood vessels, smooth muscle cells, and adipose tissue
(e-Fig. 20.59). This tumor belongs to the perivascular epithelioid cell tumor
(PEComa) family. The mean age at presentation is 45 to 55 years for patients
without tuberous sclerosis, and 25 to 35 years for patients with tuber-
ous sclerosis. Patients with tuberous sclerosis tend to have multiple, bilat-
eral renal tumors and can have associated pulmonary lymphangioleiomy-
omatosis. Tumors are nonencapsulated, yellow to pink masses (e-Fig. 20.60)
depending on the content of the tissue components. Rarely, angiomyolipo-
mas can extend into the renal vein or the vena cava. Vascular invasion and
lymph node involvement can be present; however, these features are consid-
ered to be evidence of direct extension and multifocality, respectively, rather
than metastatic disease.
Chapter 20 • Surgical Diseases of the Kidney I 3 67
Microscopically, the limits between the tumor and the kidney are well
defined. The smooth muscle cells are generally spindled (e-Fig. 20.61) but can
appear round or epithelioid in some cases (e-Fig. 20.62). The smooth muscle
cells often appear to radiate from the outer aspect of the thick-walled, hyalin-
ized blood vessels (e-Fig. 20.63). Nuclear atypia can be present (e-Fig. 20.64).
The amount of fat is variable and can be very focal (e-Fig. 20.62). Uncom-
mon to rare histologic variants include fat-predominant, smooth muscle-
predominant, lymphangioleiomyomatous (e-Fig. 20.65), oncocytoma-like,
sclerosing type, and angiomyolipoma with epithelial cysts (e-Fig. 20.66).
Characteristically, angiomyolipomas coexpress melanocytic markers such
as HMB45 and smooth muscle markers such as smooth muscle actin and
muscle-specific actin. Epithelial markers including cytokeratin are always
negative. Angiomyolipomas can be diagnosed in needle biopsy tissue (e-Fig.
20.67). Classic angiomyolipomas are benign.
4. Epithelioid angiomyolipoma is a potentially malignant mesenchymal neoplasm
that presents more commonly in patients with tuberous sclerosis. It is much
less common than usual angiomyolipoma. Grossly, these tumors are usually
large with tan-gray or hemorrhagic cut surfaces and necrosis. The cells are
epithelioid with abundant granular cytoplasm. Multinucleated cells, nuclear
pleomorphism, mitotic activity, vascular invasion, necrosis (e-Fig. 20.68),
and involvement of perinephric fat can be present. These tumors express
melanocytic markers with variable expression of smooth muscle markers.
These tumors can metastasize to lymph nodes, live.r; lungs, and spine.
5. Leiomyoma is a benign smooth muscle neoplasm that can arise from the
renal capsule, the muscularis of the renal pelvis, or from cortical vascular
smooth muscle. Most are found incidentally. Grossly, they are firm well-
defined masses, although calcification and cysts can be present. Necrosis
should be absent. Leiomyomas are composed of spindled cells arranged in
fascicles, with minimal nuclear pleomorphism and no mitotic activity. They
demonstrate a smooth muscle immunophenotype, with actin and desmin
immunopositivity. So-called capsulomas that look like leiomyomas originat-
ing from the capsule that are HMB-45 positive are thought to be monophasic
leiomyomatous angiomyolipomas.
6. Hemangioma is a benign vascular tumor that presents in young and middle-
aged adults. The tumor is usually unilateral and single, with a red spongy
gross appearance. Microscopically, the lesion is characterized by irregular
blood-filled spaces lined by a single layer of endothelial cells. No mitoses or
nuclear pleomorphism is present.
7. Lymphangiomas are more common in adults and can represent a lymphatic
malformation or can develop secondary to urinary tract infections. Grossly,
they are cystic, encapsulated lesions that can overgrow the entire renal
parenchyma. The cysts are filled with clear fluid and lined by a single layer
of flat endothelium.
8. Juxtaglomerular cell tumors are benign renin-secreting tumors that occur in
younger individuals and are more common in women. Clinically, the tumor
manifests with severe hypertension and hypokalemia. The tumors are solid,
well circumscribed, and composed of sheets of polygonal or spindled cells
with central regular nuclei, well-defined borders, and granular eosinophilic
cytoplasm. Mast cells, hyalinized vessels, and tubular elements are common.
The tumor cells are immunoreactive for renin, actin, vimentin, and CD34.
9. Renomedullary interstitial cell tumors are commonly found during autopsy.
They are present in about 50% of men and women, and frequently they
are multifocal. They are 1 to 5 mm in diamete.r; white or gray, and located
within the renal pyramids. Histologically, they contain small polygonal cells
368 I SECTION V: URINARY TRACT
B. For a core needle biopsy. report the histologic type, histologic grade (Fuhrman
nuclear grade), and any additional pathologic findings (such as inflammation
and glomerular disease).
C. For a nephrectomy, partial or radical, report the tumor site (upper pole, mid-
dle pole, lower pole), focality (unifocal or multifocal), tumor size (largest, if
multiple) in greatest dimension, macroscopic extent of tumor (tumor limited
to kidney, tumor extension into perinephric tissues, tumor extension into renal
sinus, tumor extension beyond Gerota•s fascia, tumor extension into adrenal
gland, tumor extension into major veins [intraluminal, with or without vein
wall invasion], tumor extension into pelvicaliceal system, histologic type, sarco-
matoid features [and percentage if present], histologic grade [Fuhrman nuclear
grade], pathologic TNM stage, and margin status [cannot be assessed, margins
uninvolved by invasive carcinoma, margins involved by invasive carcinoma]).
The site(s) of margin positivity should be specified. For partial nephrectomy
specimens, the renal parenchymal margin, renal capsular margin, and per-
inephric fat margin should be assessed. For radical nephrectomy specimens,
Gerota•s fascial margin, renal vein margin, and ureteral margin should be
addressed. The adrenal gland, if present, should be reported as uninvolved by
tumor, involved by direct invasion, or involved by metastasis. Lymph-vascular
invasion should be designated as absent, present, or indeterminate. Additional
pathologic findings in nonneoplastic tissue 5 mm or greater from the mass
should be recorded, including glomerular disease (type), tubulointerstitial dis-
ease (type), and vascular disease (type). Optional reporting includes cyst(s) and
tubulopapillary adenoma(s).
VI. NONNEOPLASTIC TUMOROUS CONDITIONS include the maldevelopment and nonneo-
plastic cystic diseases discussed earlier. Another important pseudoneoplastic cate-
gory is inflammatory masses such as xanthogranulomatous pyelonephritis (XGP),
renal malakoplakia, and tuberculosis (TB).
A. xanthogranulomatous pyelonephritis (XGP) is a subacute to chronic, unilateral
inflammatory process that can form a mass in the kidney mimicking RCC clini-
cally, radiographically, grossly, and histologically. This disease most commonly
occurs in women from the fourth to the sixth decades of life. XGP typically
presents with fever, flank pain, or a tender flank mass and is frequently compli-
cated by nephrolithiasis. Urine cultures show common urinary tract pathogens,
such as Escherichia coli and Proteus mirabilis. in up to 70% of cases. If the
kidney itself is cultured, an organism can be isolated in 95% of cases.
Macroscopically, XGP can either be confined to the kidney or extend into
the surrounding soft tissue. XGP is typically composed of yellow nodules of
varying size replacing the normal renal parenchyma (e-Fig. 20.75); the nod-
ules can range in size from a few millimeters to several centimeters. Micro-
scopically, the nodules are composed of lipid-laden macrophages (e-Fig. 20.76)
that can mimic low-grade dear cell RCC, especially in small tissue samples
such as needle biopsy specimens. The lesion may also contain reactive and even
multinucleated fibroblasts that can mimic a sarcomatoid component in RCC.
However, XGP lacks the vascularity typically seen in RCC, and moreover, XGP
often exhibits neutrophils, lymphocytes, foreign body giant cells, plasma cells,
cholesterol clefts, and microabscesses. If there is concern for RCC, a panel of
immunohistochemical stains can be helpful: the clear cells in XGP will be pos-
itive for CD68 and vimentin, and negative for epithelial markers such as EMA
and pan-cytokeratins.
B. Renal malakoplakia is uncommon but can form masses simulating a primary
renal neoplasm. Most patients are women who often have extrarenal malako-
plakia in the urinary bladder, ureter, or retroperitoneum. The lesion is bilateral
in about one-quarter of cases. Grossly, there may be diffuse multinodular cor-
tical enlargement or a large yellow mass. Abscesses and cystic spaces may also
Chapter 20 • Surgical Diseases of the Kidney I 371
Cytopathology of Renal
Neoplasms
Souzan Sanati
I. INTRODUCTION. The role of fine needle aspiration (FNA) in diagnosis of renal masses
is limited in comparison with many other deep-seated organs, due in part to the suc-
cess of imaging techniques in correct classification of most renal masses as benign
or malignant. FNA is therefore usually considered only when imaging studies show
equivocal results. Since most renal cysts with equivocal radiologic findings usually
require extensive tissue sampling, the role of FNA of renal cysts is limited and often
renders negative results; while FNA can be useful when malignant cellular features
are identified, cytologically negative cyst fluid does not exclude a malignant pro-
cess and thus it has been recommended that these specimens should be reported as
nondiagnostic. FNA sampling of pediatric renal masses has not been advocated.
Despite these limitations, FNA of renal masses, under radiologic guidance, can
be used as an alternative to core biopsy and can be useful in evaluating metastatic
tumors, benign masses (eliminating the need for a surgical procedure), masses diag-
nosed in patients who are poor surgical candidates, and in masses occurring in
candidates for a tumor ablation procedure.
When the renal pelvis is involved by tumo.r; cytologic sampling can be per-
formed endoscopically. Since the endoscopic biopsies are usually small, cell block
processing is advocated, particularly in low-grade lesions where the architectural
features are of diagnostic importance. Cytologic evaluation of ureteral catheteri-
zation samples is also useful for detecting high-grade urotheliallesion of the upper
urinary tract(/ Urol. 2000;164:1901). In the case ofrenal abscesses, FNA can have
a therapeutic as well as diagnostic role.
II. CYTOPATHOLOGY OF RENAL NEOPLASMS
A. Benign neoplasms
1. Angiomyolipoma. Mature adipose tissue, smooth muscle, and blood vessels
are the main components of this tumor (e-Fig. 20.78). Identification of adi-
pose tissue in the FNA biopsy is an important clue for diagnosis; however,
most angiomyolipomas subject to cytological sampling have a low fat con-
tent. The smooth muscle component may have an epithelioid appearance
with significant nuclear atypia, and it is important not to misinterpret this
finding as a malignant process. Immunostains for HMB45 and smooth mus-
cle actin are used to differentiate angiomyolipoma from RCC (Acta Cytol.
2006;50:466).
2. Oncocytoma can be solitary or multifocal and usually has a characteristic
radiologic appearance that includes good demarcation, a central scat; and
372 I SECTION V: URINARY TRACT
I. NORMAL ANATOMY. The upper tract of the urinary collecting system is composed of
the renal calyces, pelves, and ureters. Renal papillae protrude into the minor calyces,
which expand into two or three major calyces, which in turn are outpouchings of
the renal pelvis, a sac-like expansion of the upper ureter.
The mucosa is normally arranged in folds. The urothelium of the renal pelvis
is three to five cell layers thick, and five to seven cell layers thick in the ureter.
The lamina propria is composed of highly vascularized connective tissue without
a muscularis mucosa; it is absent beneath the urothelium lining the renal papillae
and is thinned along the minor calyces. The thickness and amount of muscularis
propria in the collecting system within the renal sinus fat can be variable; ureteral
muscularis propria is composed of interlacing bundles of smooth muscle, without
inner or outer layers (Mills SE, ed. Histology for Pathologists. 2nd ed. Philadelphia:
Lippincott Williams and Wilkins; 2007:839-907).
II. GROSS EXAMINATION AND TISSUE SAMPLING. Tissue samples include ureteroscopic
biopsies, needle biopsies, pyeloplasty specimens, segmental ureterectomy speci-
mens, radical cystectomy/cystoprostatectomy specimens, and radical nephroureter-
ectomy with urinary bladder cuff resection specimens.
A. Ureteroscopic biopsies are entirely submitted. Because these are often minute in
size, one approach to processing is to submit the biopsy sample for cytology cell
block preparation (Urology. 1997;50:117).
B. Needle core biopsies of renal masses, including urothelial carcinoma involving
the kidney, should be completely submitted. For microscopic examination, it is
recommended that three levels on each of three hematoxylin and eosin (H&E)-
stained slides be produced.
C. Pyeloplasty specimens for ureteropelvic junction obstruction consist of a portion
of distal pelvis with a short segment of ureter attached to it. If the specimen is
intact, it will be funnel-shaped, and narrowing, and/or angulation of the ureter
may be evident. Usually, however, the specimen is longitudinally splayed open
by the surgeon and consists of a flat, fan-shaped pelvis with nub of opened ureter
at the "handle" of the fan. The mucosa should be examined to rule out rare mass
lesions. Measurements include mural thickness of the pelvis and ureter, and both
external and internal diameter of the ureter (the latter at its narrowest point).
If present, narrowing and angulation of the ureter should be noted. Sections
include serial cross sections of ureter and distal pelvis.
D. Segmental ureterectomy is performed for tumors of the proximal or mid-ureter.
The length and diameter of the intact ureter is recorded, with a search for a
mass by palpation and visual inspection. Proximal and distal cross-sectional
margins are taken, and the outer aspect of the ureter is inked. The ureter is then
opened longitudinally and assessed for mucosal abnormalities. After overnight
fixation in 10% formalin, sections are taken to demonstrate the deepest inva-
sion of any lesion(s). At least one section of uninvolved ureter should also be
submitted.
E. Radical cystectomy/cystoprostatectomy with segment Df ureters. Ureteral margin(s)
may be submitted for frozen section for evaluation of carcinoma (and particu-
larly carcinoma in situ). These are shaved margins and should be submitted as
cross sections.
373
374 I SECTION V: URINARY TRACT
*All e-figures are available online via the Solution Site Image Bank.
Chapter 21 • Renal Pelvis and Ureter I 3 75
of Pathology; 2004 ); and tend to present with higher histologic grade and at
higher stage. Also, biopsy of upper tract tumors is more difficult than lower
tract tumors. In about one of four cases, small ureteroscopic biopsies of the
upper tract will be nondiagnostic due to inadequate tissue (Am] Surg Pathol.
2009;33:1540).
1. Risk factors. The main risk factor, as for urinary bladder malignancies, is
smoking. Other risk factors are analgesics (as noted earlier); occupation
in chemical, petrochemical, or plastics industries; exposure to tar, coal,
or asphalt; papillary necrosis; Balkan nephropathy; thorium contrast expo-
sure; hereditary nonpolyposis colorectal cancer (HNPCC) syndrome (Lynch
syndrome II) UUrol. 2011;185:1627); and urinary tract infections or stones
(Eble JN, Sauter G, Epstein ]1, Sesterhenn lA, eds. Tumours of the urinary
system and male genital organs. Lyon: IARC Press; 2004).
2. Clinical features. Most patients are around 70 years of age, and the chief pre-
senting symptoms are hematuria and flank pain. In the majority of patients,
there is a prior history of a bladder cancer, which means that many upper
tract tumors are detected during the course of clinical surveillance after
diagnosis of a bladder tumor.
3. Histologic typing and diagnosis of upper tract tumors are accomplished by
examination of H&E-stained sections. Typing of upper tract urothelial neo-
plasia is the same as that for the urinary bladder as defined in the 2004 World
Health Organization (WHO) classification of neoplasms (see Table 22.1).
4. Urothelial carcinoma is by far the most common type of upper tract
tumor.
a. Gross diagnosis is possible in resection specimens. Patterns of growth
include papillary, polypoid, nodular, ulcerative, and infiltrative. Exo-
phytictumors can fill and distend the pelvis (e-Fig. 21.4), with or without
associated hydronephrosis and stones. High-grade invasive tumors can
grossly involve soft tissue and/or renal parenchyma (e-Fig. 21.5). Exten-
sive renal parenchymal involvement can mimic a primary renal parenchy-
mal neoplasm; in these cases there is often a request for an intraoperative
consultation to determine whether the mass is a urothelial carcinoma or
a renal cell carcinoma (RCC) because the distinction alters the extent of
surgery. The correct diagnosis can usually be determined by straightfor-
ward gross examination alone, but in some cases a frozen section may be
required. The average size of renal pelvic tumors is just under 4 em, with
a range of 0.3 to 9 em (Am] Surg Pathol. 2004;28:1545). Multifocality
in the pelvis and ureter is seen in about one-quarter of cases. In the ureter,
the tumor may be associated with a stricture and hydroureter.
b. Microscopically, the full range of urothelial carcinoma may be seen from
flat intraepithelial neoplasia, including carcinoma in situ (e-Fig. 21.6);
to noninvasive papillary neoplasia, including papillary urothelial neo-
plasm of low malignant potential; to low-grade papillary urothelial carci-
noma (e-Fig. 21.7A and B); to high-grade papillary urothelial carcinoma
(e-Fig. 21.8A and B; e-Fig. 21.9). The full spectrum of invasive urothelial
carcinoma and its variants as found in the urinary bladder can also be
found in the upper tract. Of note, unusual histomorphological variants
seem to be more common in the upper tract (Mod Pathol. 2006;19:494 ),
including carcinomas with micropapillary, lymphoepithelioma-like, sar-
comatoid, squamous, clear cell, glandular, rhabdoid, signet-ring, and
plasmacytoid features or areas.
Primary resections performed for renal pelvic urothelial carcinoma
show high-grade carcinoma in > 70% of cases, with deep invasion
(pT2 or greater) in 45% of cases and with lymph node metastases in
376 I SECTION V: URINARY TRACT
lis! ,.,.,.. - ! I I
Nit Rl!PNIIymJ*I mc!M ..,I'd be .........S
NO ~ AOP>nlll)ml*o noclo - •
Nl .........,.,. • olnlllo tJrnl>h IIOdo :s2 ctn In .,._<lmemlor!
N2 .........,aloin • olniilo tJrnl>h IIOdo 2-5 ctn In.,._<lmemlor!
N:9 MM•·"+In•¥nphi'!Dd•>5ant'l~d~
-"·'. .~~-·"'''(Ill
MX Dtlllnt mlltl.lltub:<:~m.X be aallilliiNHt
~ ~d~m-•
hql_,...
Ml
SlqoO.
~mm~•
T1 NO ~
51qo0111 n. NO MO
5lqo I T1 NO MO
$QIII T2 NO MO
sq,m 'D NO MO
Slqa IV T4 NO MO
An,T Nl-3 MO
An,T M,rN Ml
378 I SECTION V: URINARY TRACT
Ta T1 T2
Epithelium
Subepithelial
connective tissue
Muscularis
propria
T= pT
T3
Figure 21.1 Depiction of pT stages pTa, pTl, pT2, and pT3. (Modified
from: Greene FL, Compton CC, Fritz AG, et al., eds. A]CC Cancer Staging
Atlas. New York, NY: Springer; 2006.)
I. I NORMAL MICROSCOPIC ANATOMY. The wall of the urinary bladder is formed by four
layers (e-Fig. 22.1)."' The thickness of the innermost layer, the urothelium, depends
on the degree of bladder distension, and the shape of its constituent urothelial cells
ranges from smaller cuboidal cells at the base to larger polyhedral cells toward
the surface. Umbrella cells, the most superficial cells, have abundant eosinophilic
cytoplasm and are often binucleated. The underlying lamina propria is separated
from the urothelium by a thin basement membrane and is composed of loose con-
nective tissue with blood vessels, nerves, adipose tissue, and a variable number of
smooth muscle fibers forming a discontinuous muscularis mucosae. Aggregates
of urothelium termed von Brunn nests (e-Fig. 22.2) are often seen as invagina-
tions or separate clusters in the lamina propria. The term cystitis cystica is used
when these nests become prominent and undergo cystic change (e-Fig. 22.3). Cys-
titis glandularis is similar to cystitis cystica except that the cells lining the cysts
are mucin-secreting cuboidal or columnar cells, or true goblet cells, in which case
the term cystitis glandularis with intestinal metaplasia is used (e-Fig. 22.4). These
proliferative lesions, although sometimes seen associated with local inflammation,
represent variants of normal histology. Their main importance lies in the fact that
they can occasionally cause visible lesions simulating a bladder neoplasm. The third
layer, the muscularis propria or detrusor muscle, is composed of large bundles of
muscle fibers and is covered by the outermost adventitial layer, including perivesical
adipose tissue. It is important to note that adipose tissue can also be found in the
lamina propria and wall (e-Fig. 22.5), so identification of fat does not equate to a
specific layer of the bladder wall.
II. GROSS EXAMINATION AND TISSUE SAMPLING OF THE BLADDER. The most common
samples submitted for surgical pathology examination include small biopsies, larger
transurethral resection specimens, and partial and radical (complete) cystectomies.
A. Biopsy specimens. These are usually obtained without cautery ("cold-cup") and
should be immediately immersed in formalin. If multiple biopsies are submitted
separately, as in mapping procedures, they should be processed separately. After
gross examination, bladder biopsies should be marked with ink or hematoxylin,
then placed in a cassette after being put in a fine mesh envelope, wrapped in lens
paper, or sandwiched between sponge pads. After processing, three hematoxylin
and eosin (H&E)-stained slides should be prepared, each with a strip of three
to four levels.
B. Transurethral resection of bladder specimens. These specimens are usually
obtained with the aid of thermal cautery, often for the transurethral resection of
bladder tumors (TURBT). Because of the significant prognostic and therapeu-
tic implications for the presence of muscularis propria invasion by the bladder
neoplasms, it is often necessary to process all of the submitted tissue to ensure
that such foci of invasion are not overlooked.
C. Partial cystectomy specimens. Partial or segmental cystectomy is indicated in
only a minority of bladder cancer patients, typically those who suffer a first
time tumor recurrence with a solitary tumor, and tumor location that allows for
a 1- to 2-cm margin of resection, such as at the dome. Urachal carcinomas at the
"'All e-figures are available online via the Solution Site Image Bank.
379
380 I SECTION V: URINARY TRACT
dome and above, with extension toward the umbilicus, may also be treated by
partial cystectomy, as can carcinoma in a bladder diverticulum. Carcinoma in
situ elsewhere in the bladder (or multifocal tumors) is an absolute contraindica-
tion. The specimens usually consist of a sheet-like portion of tissue that should
be pinned down and fixed overnight. In addition to describing and sampling any
gross tumor(s) as described for cystectomy specimens, the status of the margins
is very important; these can be shaved off or sampled by perpendicular sections,
depending on their relationship and proximity to the tumor(s). Frozen section
of the mucosal margin may be requested.
D. Total cystectomy and cystoprostatectomy specimens. Radical cystoprostatectomy
in men and anterior exenteration in women, along with pelvic lymphadenec-
tomy, are standard surgical approaches for muscle wall-invasive bladder car-
cinoma in the absence of metastatic disease. Cystectomy may be performed in
some cases for nonmuscle wall-invasive bladder carcinoma, if the bladder is non-
functional, or for high-grade pT1 carcinoma that is not responsive to intravesical
therapy. If not sampled separately, a request to perform frozen sections on the
ureteric and urethral margins may be received (Arch Pathol Lab Med. 2005;129:
1585-1601). After orientation and inking, one of two methods can be used to
fix the specimen. The first entails filling the bladder with formalin (through the
urethra, or by using a large bore needle through the dome) and fixing overnight;
the second entails opening the bladder (usually through the urethra extending
upward on the anterior surface) and pinning it flat, then fixing overnight. After
opening the bladder (in the fresh or fixed state), the mucosa is examined for
tumors(s) and, if present, the size, location, pattern of growth (exophytic, endo-
phytic, and/or ulcerated), and depth of invasion are recorded. The mucosa of the
adjacent bladder should also be examined for areas of hemorrhage and discol-
oration that may represent areas of carcinoma in situ. In addition to sampling of
any tumor(s) (three to four sections of each), representative sections need to be
submitted from the different areas of the bladder including the trigone; poste-
rior, lateral, and anterior walls; and the dome (Fig. 22.1). If ureteric and urethral
shave margins were not submitted for frozen section examination, they should
be sampled for permanent sections, as should any possible lymph nodes iden-
tified in the perivesical fat. In cystoprostatectomy specimens, additional blocks
from the prostate and seminal vesicles should be submitted, the extent of which
depends on whether a preoperative diagnosis or suspicion of prostatic carci-
noma exists (see Chap. 29). When the bladder (with or without the prostate) is
removed as part of larger pelvic exenteration specimens (that may include por-
tions of the rectum and/or the gynecological tract in females), it then becomes
imperative to document the presence or absence of involvement of these addi-
tional organs by preferentially sampling suspicious areas, as well as by sampling
the resection margins of these organs.
Ill. DIAGNOSTIC FEATURES OF COMMON DISEASES OF THE BLADDER
A. Congenital malformations
1. Urachal abnormalities. The urachus is a vestigial structure that connects the
dome of the bladder to the umbilicus; it normally closes by the fourth month
of fetal life. Persistence or malformations of the urachus can present in child-
hood and occasionally in adulthood; they include urachal remnants (e-Fig.
22.6), patent urachus, urachal cysts, and urachal sinuses, all of which can
result in secondary infection or development of secondary tumors, most fre-
quently adenocarcinoma (e-Fig. 22.7).
2. Exstrophy. This is a rare congenital anomaly characterized by failure of devel-
opment of the anterior wall of the bladder and abdominal wall, usually
resulting in severe secondary infection if left untreated.
B. Inflammatory conditions. Cystitis most frequently has an infectious etiology.
There are, however, specific variants of cystitis that produce somewhat
Chapter 22 • The Urinary Bladder I 3 81
Dissection method
Distal
./ureter
)
~Distal
urethra
Bladder
wall and
lesion
Ureteric
orifice
near lesion
B
Figure 22.1 Gross dissection of radical cystectomy specimens by whole organ bivalve
method (A) and by opening along urethra and anrerior wall, with demonstration of sec-
tions to be taken (B). lbree to four sections, or complete embedding, of masses should be
performed. (Modified from Schmidt WA. Principles and Techniques of Surgical Pathology.
Menlo Park, CA: Addison-Wesley Publishing Co.; 1983:525.)
............
Ul!llllollal po~1111
- · -pcpllllma
ti:qU&.n'l)l.ll 011111 01!1/d"'oma
v.tru~:t~W:c.IU\oma
P~ma
..- ......a ...
C.l<t!o/1\11 of Slo>no, CUWpof, and Wre
pnclt
Me!MIIc tu....,.and111nore OX!Nns
..........,,....
Squamouo Cldl poplbna from mtw "'11'N
-
AdOIIOCO-l
Mudn...,.
Chapter 22 • The Urinary Bladder I 38 5
Small Cell
-.-- ---.--
Figure 22.2 Patterns of urinary bladder carcinoma in situ. (From McKenney JK, et al. Am] Surg
Pathol. 2001;25:356, with permission.)
386 I SECTION V: URINARY TRACT
nature of the neoplastic cells that often leads to denudation and a "cling-
ing" pattern of growth in which only scarce malignant cells remain
attached to the bladder wall (e-Fig. 22.25). Surface carcinoma in situ cells
can also extend into and "cancerize" von Brunn nests (e-Fig. 22.26}. In
uncommon cases, glandular differentiation, in the form of more colum-
nar cells with apical cytoplasm, can be noted in urothelial carcinoma
in situ (Am] Surg Pathol. 2009;33:1241). In difficult cases where the
differential diagnosis includes reactive urothelial atypia, immunostains
for cytokeratin 20, p53, CD44, and K.i-67 can be useful {Semin Diagn
Pathol. 2005;22:69). Urothelial carcinoma in situ is often associated with
invasive urothelial carcinoma and carries a significant risk of death from
bladder carcinoma (Cancer. 1999;85:2469}.
2. Papillary urathelial neoplasms
a. Urathelial papilloma. This uncommon benign neoplasm is composed of
delicate papillary urothelial fronds with no or minimal branching or
fusion. The constituent cells are identical to normal urothelial cells, and
no mitoses are present (e-Fig. 22.27). The classic cystoscopic finding is a
solitary lesion in a younger patient with hematuria. Papillomas may recur
(in up to 80% of cases) or progress to higher grade disease (in 2% of cases).
b. Inverted papilloma. Another uncommon neoplasm, an inverted papilloma
has a polypoid or sessile appearance cystoscopically. It is composed of
anastomosing islands and cords of bland urothelial cells that invaginate
and grow downward in the lamina propria with peripheral palisading, no
to rare mitoses, and no to minimal cytological atypia (e-Fig. 22.28) (Am
] Surg Pathol. 2004;28:1615, Cancer. 2006;107:2622). These latter two
features help distinguish this lesion from other papillary neoplasms that
may also occasionally have an inverted growth pattern. Inverted papillo-
mas rarely recur.
c. Papillary urathelial neoplasm of law malignant potential (PUNLMP). This
neoplasm shares the clinical and endoscopic features of papilloma but is
characterized histologically by occasionally fused papillae and ordered, yet
larger, nuclei than are seen in papillomas {e-Fig. 22.29A and B). Mitotic
figures are rare and basal. Compared with papillomas, PUNLMP has
higher recurrence (25% to 35%} and progression rates (up to 4%), and
thus close follow-up is warranted.
d. Noninvasive papillary urathelial carcinoma, law grade. In contrast to
PUNLMP, this urothelial neoplasm shows frequent branching and fusion
of papillae and variations in nuclear size, shape, and contour {e-Fig.
22.30). Mitoses are occasional and may be found at any level. These
tumors tend to be larger than papillomas and PUNLMPs and are more
likely to be multiple. They are also more likely to recur (64% to 71 %)
and progress (2% to 10%).
e. Noninvasive papillary urathelial carcinoma, high grade. These are uncom-
mon noninvasive papillary neoplasms; more frequently there is associated
invasion. They are characterized by frequent branching and fusion with
moderate to marked cytoarchitectural disorder and nuclear pleomorphism
(e-Fig. 22.31). Mitoses are frequent. Similar to low-grade tumors, these
tumors frequently recur (56%) and progress to invasive carcinoma (18%).
3. Invasive urathelial neoplasms. Invasive urothelial carcinomas can have papil-
lary, polypoid, nodular, or ulcerative configurations. Most are cytologically
high-grade tumors. Determination of anatomic depth of invasion by carci-
noma is vital, because pathological stage is the single most important prog-
nostic feature.
Recognition of diagnostic patterns of lamina propria invasion can facili-
tate pT1 stage assignment (Table 22.2). There are several different patterns,
C111ptw 22 • l~t-t Uri11wy B!•erliel I 117
IIJ.I
r«
,.,...._!II RBP>noiiJmph- Clllnol bo .. -
NO ~ ......,, tl!nllll nod& - •
Nl S*l.l.to ltmlil node m-•ln die truo~>et.t> ~ obtunmr,
_.,.,!lee,.. p-noiiJmph l'oOI!Il)
~ Polt.lii1Jio •••olb'mph l'oOdt .,......... In tile title peMt ~
ollll.rllor, - . , , lie 11 praocnllb'mph ncdo)
ro LJmP. ,_ mac.lll..la" ll>o c:ommon ,... !~'mph """"'
Mllln•n•••••
MO ~dlllol!t .,'t:t'1k (no polhoJoD> PolO, ....,dntol Ill tr> """P'*
'*'llJ!o 1!7"'JP)
Ml otdlnt mtUst••
.Cpolllil(lo . . . . . . .
sq,oa Tt NO MO
SQ~Oh 'II> NO MO
Slqa I T1 NO MO
Slqa II T2e NO MO
T2b NO MO
5lqo Ill T3a NO MO
T3b NO MO
T4a NO MO
SQIIV T4b NO MO
AllyT Nl MO
AllyT N2 MO
AllyT N3 MO
AllyT AllyN loll
388 I SECTION V: URINARY TRACT
Figure 22.3 Patrerns of invasion from papillary carcinoma. A: Microinvasive carcinoma at base.
B: Stalk invasion. C: Lamina propria invasion up to muscularis mucosae. D: Lamina propria
invasion beyond muscularis mucosae. (From Amin MB, et al. Am J Surg Pathol. 1997;21:1057,
with permission.)
A B c
:·:::::m.:::
...
.,
D E
Figure 22.4 Patrerns of lamina propria invasion. A and B: From carcinoma in situ. C: Nested
variant. D: Endophytic growth and invasion. E: Inverted growth and invasion. (From Amin MB,
et al. Am J Surg Pathol. 1997;21:1057, with permission.)
Chapter 22 • The Urinary Bladder I 389
only focally present and is often found in the deeper aspects of the prolif-
eration. Although the nested variant of urothelial carcinoma has a higher
proliferation index than florid von Brunn nests by MIB-1 immunostaining
(8.8% vs. 2.8%), and a higher p53 immunopositivity (4.2% vs. 1.5%),
the degree of overlap precludes use of these markers as diagnostic tools
(Am I Surg Pathol. 2003;27:1243).
f. Microcystic variant. This variant is also deceptively bland and somewhat
similar to the nested variant, except for characteristic prominent cystic
change (e-Fig. 22.41). It is uncommon, accounting for only about 1% of
bladder carcinomas. Microscopically, there are variable-sized cysts rang-
ing up to 1 to 2 mm in diamete.t The cysts are round to oval and may
contain necrotic material or pink secretions. The layer of lining cells may
be flattened or denuded. The differential diagnosis includes cystitis cys-
tica, cystitis glandularis, and nephrogenic adenoma. Correct diagnosis is
achieved by the detection of an association with usual urothelial carci-
noma, haphazard and infiltrative growth, and variability in cyst size and
shape. In the largest series, 25% of cases had invasion of muscularis pro-
pria and 11 of 12 were high grade (] Urol. 1997;74:722).
g. Micropapillary variant. This rare pattern of urothelial carcinoma resembles
papillary serous carcinoma of the ovary, an important differential diag-
nosis in women. It is characterized by the presence of small nests of cells
and filiform papillae that have retracted from the surrounding stroma
(e-Fig. 22.42). Lymphovascular invasion is common. Admixed invasive
usual urothelial carcinoma is detected in a majority of cases. Because the
proportion of the tumor that is micropapillary seems to be of prognos-
tic significance, the percentage of the invasive tumor that is micropap-
illary should be reported (Adv Anat Pathol. 2010;17:182). This variant
is characteristically aggressive. While muscle wall invasion is commonly
detected, some cases, especially those with a low percentage (<10%) of
micropapillary component and surface micropapillary growth, can be low
stage (pTa or pT1). It has been argued that early cystectomy should be
offered to patients with such low-stage, nonmuscle-invasive micropapil-
lary urothelial carcinoma (Cancer. 2007;110:62). Intravesical therapy is
ineffective (Urol Oncol. 2009;27:3). The 10-year overall survival is 24%.
h. Lymphoepithelioma-like variant. This is characterized by sheets and nests of
poorly differentiated malignant cells that grow in a syncytial pattern with
an admixed dense lymphoplasmacytic infiltrate (e-Fig. 22.43) (Am I Surg
Pathol. 2011 ;35:4 74 ). It may be pure or mixed with usual urothelial carci-
noma. There is a tendency for patients to present with muscularis propria-
invasive disease. The differential diagnosis centers on large cell lymphoma
and severe chronic cystitis, including follicular cystitis; immunostains for
pan-cytokeratin and CD45 are confirmatory, and Epstein-Barr virus is
not present. These are aggressive carcinomas with a 26% mortality at 3
years. These tumors should be treated as other bladder carcinomas-that
is, based on stage-although they do also appear to be chemoresponsive.
Histologic variants of urothelial carcinoma with unusual cytologic fea-
tures:
i. Lymphoma-like and plasmacytoid variants. These variants are exceedingly
rare and are usually admixed with conventional urothelial carcinoma.
However, the diagnosis of urothelial carcinoma in small biopsies com-
posed solely of such variants (e-Fig. 22.44) may be achieved only with
the help of immunohistochemistry (positive reactivity to cytokeratin with
negative reactivity to CD45 and other lymphoid markers).
j. Giant cell variant. This high-grade variant is characterized by numerous
pleomorphic and bizarre tumor giant cells (e-Fig. 22.45), and needs to be
392 I SECTION V: URINARY TRACT
and this accounts for the poor outcome for most patients. No molecular
genetic abnormalities are currently used for diagnosis or prognosis. Treat-
ment is radical cystectomy, with or without radiation therapy and chemother-
apy.
3. Verrucous squamous cell carcinoma. This variant of squamous cell carcinoma
is seen almost exclusively in patients with schistosomiasis and appears as an
exophytic "warty" mass composed of thickened papillary squamous epithe-
lium with minimal cytoarchitectural atypia and a rounded pushing border.
This tumor is considered to be clinically indolent.
G. Glandular neoplasms
1. Villous adenoma. An uncommon exophytic papillary neoplasm histologically
resembling its colonic counterpart, villous adenoma is usually located in the
trigone and, unless associated with an invasive component, does not recur
following excision.
2. Adenocarcinoma. Primary adenocarcinomas are rare, representing 2% of
malignant bladder neoplasms, and may be of urachal or nonurachal origin.
The latter are more common and usually arise in patients with a nonfunc-
tional bladder or with exstrophy. Urachal adenocarcinomas usually arise
from the dome or anterior wall of the bladder but may also involve urachal
remnants in the anterior abdominal wall. Characteristics of urachal adeno-
carcinomas that are helpful in differentiating them from nonurachal adeno-
carcinomas are that their bulk is in the wall rather than the lumen of the
bladder; they lack an associated in situ component or cystitis glandularis;
and they are sharply demarcated from surface urothelium. Identification
of urachal tumors is important because, unlike nonurachal tumors, surgi-
cal management of urachal adenocarcinomas usually includes excision of
the median umbilical ligament and umbilicus. Bladder adenocarcinomas (of
urachal or nonurachal type) may have different morphologic appearances
including enteric (e-Fig. 22.48A and B), mucinous (e-Fig. 22.7), signet-ring
cell, dear cell, and mixed. The main differential diagnosis for most of these
patterns is the more common metastasis or secondary extension from another
primary tumor site, most notably the prostate and the colon. Immunohisto-
chemistry may be helpful in this situation, especially when the clinical findings
are not helpful or available. Immunoreactivity with PSA and prostatic acid
phosphatase, ,8-catenin (nuclear), or thrombomodulin supports a diagno-
sis of prostate, colon, or bladder adenocarcinoma, respectively; cytokeratin
7 and 20 immunostains are not very useful in this context because of sig-
nificant overlap (Semin Diagn Pathol. 2005;22:69). Adenocarcinoma of the
bladder has a generally poor prognosis with 5-year survival rates ranging
from 18% to 47%.
H. Neuroendocrine neoplasms
1. Paraganglioma. Derived from bladder paraganglia, paragangliomas are typ-
ically found in the muscularis propria and are not infrequently associated
with hypertension and/or headaches, palpitations, and sweating that may be
precipitated by micturition. The tumor is composed of cells with abundant
amphophilic, clear, or eosinophilic cytoplasm arranged in a diffuse or nested
(Zellballen) pattern of growth with an associated thin capillary network
(e-Fig. 22.49). Nuclear atypia can occasionally be prominent. The tumor
is frequently immunopositive for neuroendocrine markers and negative for
cytokeratin (useful in distinguishing the tumor from nested urothelial car-
cinoma), whereas the spindle cells surrounding tumor nests (sustentacular
cells) are positive for 5100 protein. The majority (85% to 90%) of bladder
paragangliomas are benign and do not recur following surgical excision.
2. Small cell carcinoma. This is a rare neoplasm, which is diagnosed even
when mixed with other bladder carcinomas (urothelial, squamous, or
394 I SECTION V: URINARY TRACT
Subepithelial
connective --+~~~
tissue
-+-- Prostate
-+-------.(__ Urethra
Cytopathology of the
Urinary Bladder
Rosa M. D~vila
I. TYPES OF SPECIMENS
A. Voided urine normally has a mixture of benign urothelial cells and squamous
cells. Although the squamous cells may be a vaginal or skin contaminant, they
can also be derived from areas of squamous metaplasia that often develop in the
bladder trigone. When urothelial cells are present in papillary-like clusters, the
possibility of a low-grade urothelial neoplasm should be raised. Similar dusters
may also be due to recent instrumentation or lithiasis.
B. Catheterized urine normally has papillary-like dusters of urothelial cells resulting
from the mechanical disruption of the urothelial mucosa. They should not be
confused with low-grade urothelial carcinoma.
C. Bladder washings are obtained by irrigating the bladder with saline instilled
via a catheter, or during cystoscopic evaluation. The cytologic findings in this
specimen type are similar to those seen in catheterized urine.
D. Neobladder or ileal conduit samples have abundant degenerated cells, some of
which are arranged in dusters and have vacuolated cytoplasm. Well-preserved
intestinal-type epithelium is rarely present. A variable number of inflammatory
cells, macrophages, and bacteria are seen (e-Fig. 22.55).
Chapter 22 • The Urinary Bladder I 3 97
B. Squamous cell carcinoma can occur as a primary bladder tumor or can arise
in adjacent organs, such as the uterine cervix, and extend into the bladder.
A more common scenario is the presence of a squamous component within a
conventional urothelial carcinoma. Cytologic features that indicate the presence
of squamous differentiation include neoplastic cells with a variable amount
of dense and eosinophilic cytoplasm, pyknotic nuclei, and bizarre cell shapes
(e-Fig. 22.59). Parakeratotic and/or anucleated cells are often seen in the back-
ground. Poorly differentiated squamous cell carcinoma that is nonkeratinizing
may be confused with high-grade urothelial carcinoma.
C. Adenocarcinoma of the bladder can be primary or metastatic. Although most
(87%) of adenocarcinomas ofthe bladder can be identified as malignant by urine
cytology, only 67% will be additionally classified as adenocarcinoma (Cancer
Cytopathol. 1998;84:335). Columnar or cuboidal cell shapes, with cytoplas-
mic vacuoles within the neoplastic cells, are cytologic features that support the
diagnosis of adenocarcinoma.
Urethra
Souzan Sanati and Peter A. Humphrey
I. NORMAL ANATOMY. The male urethra is divided into three anatomic regions: pro-
static (bladder neck to apex of the prostate), bulbomembranous (apex of the
prostate to inferior surface of urogenital diaphragm), and penile (inferior sur-
face of urogenital diaphragm to the urethral meatus). The prostatic portion is
lined by urothelium, the bulbomembranous portion is lined by pseudostratified or
stratified columnar epithelium, and the penile portion shows a transition from the
stratified columnar epithelium at its origin to squamous epithelium at the meatus.
The female urethra is lined by urothelium in the proximal one-third and squamous
epithelium in the distal two-thirds.
The urethra has associated periurethral glands. Skene glands are present in
females and are concentrated distally. Bulbourethral (Cowper glands) and glands
of Littre, located in the bulbomembranous portion and along the penile urethra,
respectively, are present in males.
II. GROSS EXAMINATION AND TISSUE SAMPLING
A. Urethroscopic biopsy tissue samples should be entirely submitted for histologic
examination, and three levels should be examined.
B. Surgical excision of urethral carcinoma. For men, the type of surgery is depen-
dent on tumor location and extent and includes transurethral resection (TUR),
local segmental excision, partial or radical penectomy, and cystoprostatectomy.
TUR chips should be submitted in their entirety. Segmental excision specimens
should be sampled to include sections of the proximal and distal margins and
area of deepest growth. Urethrectomy (primary or secondary) involves stripping
of all or part of the urethra with preservation of the penis and is performed for
patients with primary urethral carcinoma or secondary involvement by bladder
carcinoma; sampling should include sections of the proximal and distal margins
and area of deepest growth. Gross processing of penectomy and cystoprostate-
ctomy specimens is covered in Chapters 30 and 29, respectively.
For women, local excision of the distal urethra and adjacent vaginal wall
is often sufficient surgical therapy for carcinoma of the urethra; sections of the
mass, and urothelial, radial soft tissue, and vaginal mucosal margins should be
submitted. For proximal urethral cancer in women, cystourethrectomy (ante-
rior exenteration, with excision of part or all of the vagina) is often necessary;
sections of the mass demonstrating relationships with adjacent structures and
depth of invasion, grossly uninvolved urethra and urinary bladder, and ureteral
and radial soft tissue margins should be submitted.
Ill. DIAGNOSTIC FEATURES OF BENIGN DISEASES
A. Congenital anomalies
1. Urethral valves are mucosal folds lined by normal urothelium that project
into the urethral lumen causing obstruction, hematuria, or inflammation.
Posterior urethral valves are usually seen in men and are associated with
bladder neck hypertrophy.
2. Diverticula are invaginations of urethral mucosa usually seen in women as
a result of infection, trauma, or obstruction. They are lined by urothelium
that may undergo squamous or glandular metaplasia (e-Fig. 23.1).*
*All e-figures are available online via the Solution Site Image Bank.
399
400 I SECTION V: URINARY TRACT
.......
MO
biiiiOO
mutiplo lflnph nodM
Nodlltllnt-
......... 01111trt-·
Ml
~
fllq&Oo Ta NO MO
SlqtOia 11s NO MO
llsi)Y NO MO
llspd NO MO
SQII n NO MO
fllq& II T2 NO MO
fllq& Ill n, Nl MO
T2
l3 NO,NI MO
SIQ>IV T4 NO,Nl MO
N\YT N2 MO
N\YT anrN Ml
F.,,. £<W& SB, e,rd DR, Com...... OC:, .. tl,- AJCC Clw8r511,11>y-.ol: 7111 ed. N'"' - ·
NV,~-- 2010.llood- 1"""'-l.
Chapter 23 • Urethra I 4 03
Thyroid
Changqing Ma, James S. Lewis Jr.,
and Rebecca D. Chernock
I. NORMAL ANATOMY. The thyroid gland is a bilobed organ in the lower neck sur-
rounding, and in intimate contact with, the trachea. It consists of right and left
lobes connected by a small isthmus in the midline (Fig. 24.1). Two functioning
cell types, follicular cells and C (calcitonin) cells, comprise the thyroid follicle. The
follicular component develops from invaginating tissue from the tongue base (fora-
men cecum) at 5 to 6 weeks gestation. Through differential embryonic growth and
migration, the bilobed gland assumes its definitive location in the neck, and the thy-
roglossal duct, the structure along the path of its downward migration, undergoes
atrophy. The C-cells, which comprise 0.1% or less of the total thyroid cell mass,
originate from the ultimobranchial body that develops from the fourth branchial
pouches; they are distributed in a density gradient, being most prominent in the
upper lobes.
The normal gland has a relatively firm consistency, is light brown, and lacks any
obvious nodules. Microscopically, it consists of follicles lined by low cuboidal bland
epithelial cells. This follicular epithelium surrounds a central core of eosinophilic
colloid. In normal glands, the follicles are relatively consistent and regular in size
(e-Fig. 24.1)."' The C-cells lie within the follicular epithelium and are invested by
the basement membrane. They are inconspicuous in normal thyroid.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION
A. Biopsy. Tissue needle biopsies of the thyroid gland are only very rarely performed
because fine needle aspiration is technically easy to perform, has low morbidity,
and is effective for triaging lesions for further management (see the section on
Cytopathology).
B. Resection. Hemithyroidectomy and total thyroidectomy are common proce-
dures for management of thyroid disease. The specimens from both types of
procedure are handled in a similar manner. The gland is oriented, if possible, on
the basis of the anatomy alone or on markings provided by the surgeon. It is then
measured and weighed. The surface is inspected for any disruptions or attached
soft tissue. Small nodules of tissue situated in the adjacent tissues may repre-
sent lymph nodes, parathyroid glands, or sequestered thyroid in cases of nodu-
lar hyperplasia. The gland should be inked, sectioned in an axial plane (bread
loafed) at intervals of 3 to 5 mm, and described, clearly indicating masses and
their size(s), color, and consistency. For diffuse or inflammatory lesions, three
• All e-figures are available online via the Solution Site Image Bank.
404
Chapter 24 • Thyroid I 405
.... ,
rI ' \
\ \
I I
[J
I I
Tumor I
I
I
I
I I
I I
I I
I \
II
I
p' \
I
I
I
I
Capsule
Figure 24.1 Thyroid anatomy/grossing (R, right lobe; ~ left lobe; P, pyramidal
lobe).
or four sections from each lobe should be submitted for microscopic exami-
nation, along with one of the isthmus. For a solitary, encapsulated mass, the
entire circumference of the capsule should be sampled, including the surround-
ing thyroid tissue because the distinction of adenoma from carcinoma relies on
features seen in the capsule. For a solitary, nonencapsulated or not completely
encapsulated mass, at least one section per centimeter should be submitted,
and some sections should demonstrate the closest inked margin (Fig. 24.1).
For multinodular glands (nodular hyperplasia), sections of each major nodule
should be submitted, including the edge and/or adjacent soft tissue margin. Any
attached perithyroidal soft tissue should be removed and sampled. Any lymph
nodes or parathyroid glands should be removed and sampled as well, although
oftentimes, the parathyroid gland(s) are first detected by microscopic examina-
tion (the number of parathyroid glands should be recorded in one of the final
diagnostic lines to highlight their presence).
Prophylactic thyroidectomy is currently recommended in infancy, early
childhood, adolescence, or early adulthood for patients with a germline RET
gene mutation. The gross specimen often consists of the thyroid gland with no
grossly identifiable lesions. Each lobe should be entirely submitted sequentially
from superior to inferior to allow thorough examination for C-cell hyperplasia
or medullary microcarcinoma.
Ill. DIAGNOSTIC FEATURES OF COMMON DISEASES
A. Developmental. Residual thyroid tissue can be present anywhere along the path
of thyroid migration during development. Carcinomas can rarely develop out-
side of the thyroid gland in these residual tissue sites.
1. Pyramidal lobe. The most common development remnant is a pyramidal
lobe, a linear projection of thyroid tissue from the isthmus pointing cranially
(Fig. 24.1).
2. Thyraalassal duct cyst Failure of closure of the thyroglossal duct commonly
results in a midline cyst known as a thyroglossal duct cyst (TDC). Excision
specimens generally consist of several nondescript fragments of tissue, and a
visible cyst is not always appreciated. Because remnants of the midline tract
pass through the hyoid bone, the latter should be identified. Failure to resect
the bone often results in persistence of the TDC. If a cyst is identified grossly, it
should be sampled in a few sections to document the type of epithelial lining,
which is often obscured by acute inflammation. Because IDCs develop from
a hollow tube, not all of them have thyroid tissue in their walls (only ""5%
406 I SECTION VI: ENDOCRINE SYSTEM
has a "scalloped" appearance from dearing at the interface with the fol-
licular epithelium (e-Figs. 24.5 and 24.6). Treated cases have a variable
morphology and often have cellular nodules mimicking adenomas. After
radioactive iodine treatment, there can be marked nuclear atypia. The dif-
ferential diagnosis includes papillary carcinoma when the stellate outlines
of follicles resemble papillae. The lack of cytologic changes of papillary
carcinoma and the diffuse gland involvement are keys to the correct diag-
nosis of Graves disease.
d. Riedel thyroiditis. A peculiar form of fibrosing disease, Riedel thyroiditis
is a chronic thyroiditis of unknown etiology which is more common in
women, occurs most commonly in the fifth decade, and is commonly
associated with fibrosing disease at other sites such as the mediastinum,
retroperitoneum, and lung. Patients present with firm thyroid enlargement
and local symptoms such as dysphagia, stridor, or dyspnea. Recurrent
laryngeal nerve or sympathetic trunk involvement can lead to hoarseness
or Horner syndrome; compression of the large vessels can lead to superior
vena cava syndrome. Most patients are euthyroid at presentation, but
many subsequently develop hypothyroidism.
Grossly, the thyroid gland is usually received in irregular pieces because
the fibrosis makes it difficult to remove surgically. It is tan to white, firm,
and may have attached muscle. Microscopically, the characteristic find-
ing is dense and hypocellular eosinophilic fibrous tissue with scattered
and patchy aggregates of lymphocytes, plasma cells, neutrophils, and
eosinophils, without germinal centers or granulomas. Rare entrapped and
atrophic thyroid follicles are seen without Hiirthle cell change. A charac-
teristic finding is small veins with infiltrating lymphocytes and myxoid inti-
mal thickening (features of occlusive vasculitis). Marked fibrosis extends
into and involves the surrounding soft tissue as well.
The differential diagnosis includes hypocellular anaplastic thyroid car-
cinoma and fibrous or sclerosing Hashimoto thyroiditis. The lack of necro-
sis or markedly atypical cells rules out anaplastic carcinoma. The lack of
Hiirthle cell change and germinal centers, and the profound degree of
perithyroidal fibrosis, rules out fibrous Hashimoto disease.
C. Nodular hyperplasia. Nodular hyperplasia (clinically termed multinodular goiter)
is an extremely common disorder of the thyroid gland. It is the enlargement of
the gland with varying degrees of nodularity. The pathogenesis is complex but
has been classically related to iodine deficiency with impaired thyroid hormone
synthesis and subsequent TSH stimulation. With supplementation of iodine in
the diet, nodular hyperplasia has also been related either to excess iodine intake
with impaired organification or to genetic factors. It is much more common in
women than men and typically presents in middle age as asymptomatic enlarge-
ment. Large goiters, however, can cause dysphagia, hoarseness, or stridor and
can extend into the upper mediastinum. A small percentage of patients present
with hyperthyroidism (toxic multinodular goiter).
Grossly, the thyroid gland is diffusely but irregularly enlarged. Some glands
can attain a weight of several hundred grams. Parasitic nodules that have only a
tenuous connection to the gland can develop. On sectioning, the nodules may be
semitranslucent, glistening, fleshy, red-brown, tan, and solid or, more commonly,
show varying degrees of degeneration with cystic change, hemorrhage, fibrosis,
and calcification. Heterogeneity of the nodules is typical (e-Fig. 24.7).
Microscopically, nodular hyperplasia is also heterogeneous. The common
appearance is nodules composed of variably sized follicles (e-Fig. 24.8). The
nodules may be quite cellular with tightly packed follicular epithelium and lit-
tle colloid, resembling follicular adenomas or carcinomas. Around cystic areas
there is often fibrosis with variably sized foci of dystrophic calcification and
Cllojbr :u • n.,..;, I •••
''I.I II Jtlbl WltO HlttlliolfC~I Cultlcllloft o1 ~nacn
n.,no~•-···
Plplllry c:an:lnoma
Fokuilr Clllf'dnome
~dl'lllllll'ldotod ...........
Undiii'•!M~Dd ~"'plullol .,.,_.,,
Squ""""" ool.....,l...,.
M._llllnnolll aorct>orn1
Scloo"'*'Cm~Xmplllonnald corct>orn• \0!111 ""'IMpNIII
Muc:fnOI.IS<::e~a
Meduii!Y .,.,.,.,..,.
Mlcod m~ulory Uld ~lwlu Clltl1101110
Splndloooltl.mOr\Oiih 111ymLO...k.e dl'lll,.,tlat!Gn
c.mom.c-na111ym.,.....,d_•
"""' ...............til. .
Fokuilr ad1n0me
ltldnlzlll!l11'1becUar\lrnor
O..•fr,jnllll-...
T-
Pit!wy lympllomallld plaouoaqloma
6doi>lc tlrjmoma
An&tooo"""'•
Sll\00111 muael& 111rnore
"-'>honl· - -.,.,....
~......1om.
SobJy ftlln>uslllmor
-ulor 0Dnd1111c cdllllmor
~ .....11~14
SeoorldiJYliiMOIO
410 I SECTION Vlt EHDOCAIHE 5YS1EM
I' 1,1,! I Jt!J1H~ FMturtt DIP!'wen411u111 111e adltiiJ 1nnv ,._ 111e
'
...,.. - Mal&l'lllll Foil leW•......, .........
Oomplalla tMA dalloola <:~poult Omoo,<talm-lflb,..
E'M!OPDGdor--od~IM. 'lhlnocoi>OIIIor 'lllwilroomlnl"ln""""'
<OPOUio
J-poood .. prolopood thj<old- ,.., Adlorono&of~tlillu•tolll-m
..-,butlOI!IIIntad..-olum 101111_,.,1111d 11\rombua
Chapter 24 • Thyroid I 41 1
,~...,•-m
lX Pltnlrt tl.mof' Ql mat be eaa:MOd
TO t1o eWier>:e d lltlmOJY 111m«
T1 1\lm« :S2 em-. l)oo1Dot ,..........,, llmbd 101118~
Tie lUMOf ~ l c:m, llrrit6d t) the~
Tlb Tumor> 1 an but not >2 an II\ lfllllbll dlrniNion, Urnttad mH1t l:h)'rdd
T2 Tum« >2an but not ><4 an,IImbd !Dille~
l3 TumOf :.4an In 1 1 - dlmONion llmllod loll>o lh;j!1>1c1C>' .., _ _
mtllmolall'llt~ ..S.n•lon <•«·•- 11>....,~ m-a«
DOI1IIill'ol:l ooft 1ilouoo)
T4o 1\lmOf d•nyU.. -ndll1! beftond 1M lh;j!1>1d """"'\&1 0 -
tubel!Wti!IIOUttdttlsaua&J il.rynx. tr'*d~M~ 6!liti~J Of MC:UI16rTt
-
~rynfiOI)noNO
T41> TumOf t1¥ad1111 --~-Of """""l""trAI:I niY Of modloollno!l
AJIIIII/IIIItlc ~ <1e~ T4 ftHno/lo
T4o ~,.,.,t>iu11c:«<elnomo
T41> All11>1ude oueln""" wllll-..
-mlll!li!!l~lft'"'*!ai-
IIIINII.,.flll- (II)
r« R...,.llpnph .-...,nctbo.,_
NO t1o .....,I tl!nllil node""""""'*
Nla , _ . 1 0 "-1 Yl (-heal, pe!WAcllool, 01\d p~\/Deli>Nan
tl!nllil nodes)
Nib fliMM"+t) unllftelll!l\ ~ orOGI'II:n!ll.atl!illl!ll~l (~~wah I, 11, 11, ~
orVl« r411rojNrynJIIII a 111peoior modlaiiNIIJmflll noclos ()owl VII)
......,1... 00
MX Ololantm-loconnctboo-.d
MQ tlodlllll!!l_ ...
Ml Oiolant m-.olt
lll,p........
Soportdo....., . . .~ ""' 111C0flltn01101odl0< popllluy or l'dl1::11\er (dllo...-.s), """*'filly,
tnd """'Ia* (uhdl'lereMioledl CITdn..,.,,
~"' F'<llbi.llrCllt!nn11a11ld), <45 l'OOI'8
fllqol Allyl AllyN MO
Slqtii AnyT AllyN Ml
~ ar Folllt:dt (<tffnnllllod), 45 I"'"' and <*Jtf
Slqj:ll T1 NO MO
Sla.elll '12 NO MO
Sla.ellll l3 NO MO
fllqoiii T1 Nl4 MO
fllqo III '12 Nl4 MO
SIQt III T3 Nit MO
Slqj:l filA T4o NO MO
Slqj:l filA T4o Nl.l MO
Sla.el ~ T1 Nib MO
Sla.el ~ '12 Nib MO
fllqo filA T3 Nib MO
SIQt filA T4o Nib MO
Slqj:l fiiB T41> Any N MO
Slqj:l fiiC Any T Any N MI
Cllojbr :u • n.,..;, I •n
I' I.I II ftlt$1 l'IIIMI.!We.IWI.ttalilll (TNIll).._ kllllme hw T - o1111e
' - lll!rold (t:MII...,)
~CM:Inomt (d ""'l/fOUPII)
51qol n NO MO
5lqo II T2 NO MO
$QIII 13 NO MO
Slqa III n Nl4 MO
Slqa III T2 Nl4 MO
Slqo III l3 Nt. MO
Slqo IVA T4a NO MO
5lqo IVA T4a Nt. MO
sq, Ml n Nib MO
sq, Ml T2 Nib MO
Slqa IVA 13 Nib MO
Slqo IVA T4a Nib MO
Slqo 1VB T4b Any N MO
5lqo IVC AnyT AllyN Ml
AMIJ/IIotlc~
AlloniC>Iudc Clleln<tno..re CIOIIOllerecl S1aal> ti
Slqa IVA T4a Any N MO
Slqo 1VB T4b Any N MO
Slqo IVC AnyT AllyN Ml
416 I SECTION VI: ENDOCRINE SYSTEM
*All e-figures are available online via the Solution Site Image Bank.
422
Pmwy ~lorn ~ '""'"" llllmu!La
s-nduy i~Jl*P<IIIIIY\'ddllm Kn-.llllmu!La tueh .. cl~ronk: reno1 fdurt,
m~.«YIImln l>m<U- abnormalllf
TIM1ioly '-"'11\)!'oldlom Aftorlon-dinsleiiOifai~n>\01111 dolroloc>l!\fllt ~
............. I>Oiell\)!'old lt/ll011'111dSoh
Pai'Uh)!okt Cln:homl
Plrllllyrold odonoma
Sooonclll)'tu,....
-
F!Otne Oel.al!a AA_ Ul7j!t RV,HelxP.elal.,eda. ~IWII:n~~otT~
- - - - r"""""'"',_~ '-r....... ww.._ 2004. Uoodw!th
....
Chapter 25 • Parathyroid Glands I 4 25
I. NORMAL ANATOMY AND HISTOLOGY. The definitive or adult adrenal glands are
located anterior to the upper poles of the kidneys. The glands are pyramidal on
the right and crescentic on the left. In adults the normal combined weight should
not exceed 6 g. Each adrenal gland is divided into head (most medial), body
(middle), and tail (most lateral).
The adrenal gland is composed of an outer cortex and an inner medulla, and
as a compound gland is unique to mammals (Pharmacol Rev. 1971;23:1971).
The gland is derived from two embryologic progenitors: the coelomic epithelium
between the urogenital ridge and dorsal root mesentery as the primordial cor-
tex, and migratory neural crest cells as the future medulla as well as the ganglia
and paraganglia. Migratory neural crest cells are identified as small dark cells
as they pass through the cortex into the center of the gland as individual cells
or small aggregates of neuroblasts (Endocr Pathol. 2009;20:92; Folia Histochem
Cytochem. 2010;48:491); however, a definitive medulla does not become appar-
ent until 1 to 11/2 years of age.
Microscopically, the cortex consists of three zones: the outer zona glomeru-
losa (secreting aldosterone), middle zona fasciculata (secreting mainly cortisol
and minor amounts of sex steroids), and inner zona reticularis (secreting mainly
sex steroids) (e-Fig. 26.1).* The zona glomerulosa is composed of a thin, usually
discontinuous layer of cells with ball-like formations; these cells have less cyto-
plasm than those in other two cortical zones. The zona fasciculata consists of
radial cords or columns of cells with abundant lipid-rich cytoplasm. The cells of
the zona reticularis have compact, finely eosinophilic cytoplasm with or without
lipofuscin pigment. Normally the medulla accounts for 10% of the adrenal vol-
ume and grossly has a gray-white color. The predominant cells in the medulla are
the mature chromaffin cells, the pheochromocytes, organized in nests and cords
(e-Fig. 26.2). The cytoplasm of the chromaffin cells is usually basophilic but may
be amphophilic or even eosinophilic. These cells have indistinct cell borders and
usually a single nucleus which may show variation in size and hyperchromasia.
The chromaffin cells are peripherally surrounded by the sustentacular cells; rare
ganglion cells may be identified in the adrenal unlike their prominence in ganglia
(e-Fig. 26.3). The major function of adrenal medulla is the synthesis and secretion
of catecholamines (epinephrine and norepinephrine).
II. GROSS EXAMINATION OF ADRENAL GLAND SPECIMEN. The adrenal gland is removed
either as part of a radical nephrectomy or for excision of an adrenal tumor. Biop-
sies of the adrenal are generally fine needle aspirations or thin needle biopsies to
evaluate for the presence of metastatic tumor.
A. Needle biopsy. The entire tissue should be submitted for histologic examination.
B. Adrenal gland removed as part of radical nephrectomy. After gross examination
of the kidney, the gland should be measured and weighed. Then the gland
should be serially sectioned at 2- to 3-mm intervals perpendicular to its long
axis; the thickness of the cortex and medulla should be noted. Infrequently,
*All e-figures are available online via the Solution Site Image Bank.
428
Chapter 26 • The Adrenal Gland and Paraganglia I 429
the adrenal may be invaded directly by renal cell carcinoma or be the site of
discontinuous metastases.
C. Adrenectomy for a primary pathologic process. The first step is to orient the spec-
imen and examine the contour of the adrenal gland. If it is apparent that the
gland has been largely replaced by a mass, the periphery should be inked since
the margins may be important (assuming that the mass has not already invaded
surrounding structures like the liver). If the cortex and medulla maintain their
normal relationship to each other, the thickness of each should be recorded.
Serial sectioning should be done at intervals appropriate for the pathology.
If the disease process is apparently diffuse hyperplasia, the periadrenal soft
tissue should be removed and the gland should be measured and weighed. If
the adrenal contains a solitary mass or multiple nodules, three dimensional
measurements should be obtained. The cut surface of the lesion and its rela-
tionship to any identifiable normal tissue should be described, including color,
consistency, presence of hemorrhage or necrosis, degree of circumscription, and
degree of encapsulation. If any portions of adjacent organs such as liver, kidney,
spleen, or abdominal wall are attached (usually for tumors), their appearance
and relationship to the gland should be noted. A large, en bloc resection will
require numerous sections from the peripheral margins. For diffuse and/or
nodular hyperplasia, representative sections are sufficient.
For a neoplasm, the following sections should be taken: tumor, (including
sections demonstrating the relationship of the tumor to the associated soft tis-
sues and adjacent organs, and relationship of the tumor to uninvolved adrenal
gland); the tumor capsule, if present; margins; periadrenal fatty tissue overlying
a bulging mass; a representative section from uninvolved adrenal, if any; and
regional lymph nodes. The gross description should clearly document the site
of the sections. For large specimens, a gross photograph is extremely helpful
in that it depicts the specimen at a time when landmarks are still maintained
with some anatomic orientation.
D. Neuroblastoma specimens. Neuroblastic tumors present some special issues
regarding acquisition of neoplastic tissue for a variety of special studies to
biologically profile the tumor for children enrolled in a Children's Oncology
Group (COG) protocol. If the specimen is an adrenal-based neuroblastoma
(NB), then generally the amount of available tumor is sufficient for patho-
logic diagnosis as well as for all other ancillary studies. The difficulties arise
in those cases of NB when only biopsies are obtained prior to chemotherapy
in cases of nonresectable tumors. Accuracy of tumor grading is based upon
a thorough microscopic examination which may be limited by the amount of
well-preserved tumor in the specimen.
A resected primary NB of the adrenal, or extraadrenal retroperitoneal mass,
should be examined fresh if at all possible as discussed in section C above. The
COG reference laboratory requests at least 1 g of snap frozen tumor tissue but
will accept any frozen sample of tumor; the snap freezing should occur as soon
as the specimen becomes available following resection. Tumor samples should
be labeled "primary" or "metastatic"; involved bone marrow is required as
well. Storage at -70°C is preferable to -20°C.
Fresh tissue should also be collected for local institutional studies including
conventional cytogenetic studies. Snap-frozen fresh tissue should be saved for
possible molecular studies. Tumor for fluorescent in situ hybridization can be
recovered from formalin-fixed, paraffin-embedded tissue without compromis-
ing the quality of results.
Ill. ADRENAL CORTICAL LESIONS
A. Congenital abnormalities. The most common congenital anomaly of the adrenal
is an incidental finding of heterotopia consisting of microscopic foci of cortical
tissue or nodule(s) along the path of descent of the gonads, although rare cases
430 I SECTION VI: ENDOCRINE SYSTEM
anencephalic type (without CNS or pituitary defects but the cortex is atten-
uated and the fetal cortex is inconspicuous); and a miniature type with a
definitive cortex and an attenuated fetal cortex.
2. Secondary and tertiary adrenal cortical insufficiencies. These are due to the
failure of the pituitary gland to secrete ACTH (secondary) or of the hypotha-
lamus to secrete CRH (tertiary). The gland size is decreased. Histologically,
the zona fasciculata is atrophic whereas the zona glomerulosa and medulla
are usually relatively normal.
D. Adrenal cortical hyperplasia can be divided into congenital and acquired types.
1. Congenital adrenal hyperplasia is an autosomal recessive disorder caused
by one of five enzymatic defects that result in a failure in cortisol synthe-
sis; 21-hydroxylase deficiency accounts for 90% to 95% of cases (Lancet.
2005;365:2125; Endocr Dev. 2011;20:80). Marked diffuse hyperplasia
of the zona fasciculata results from ACTH stimulation, whereas cells of
the zona fasciculata are lipid-depleted due to their conversion into zona
reticularis-type cells with compact eosinophilic cytoplasm. Persistent ACTH
stimulation may also give rise to adrenal cortical neoplasms. Nodules resem-
bling hyperplastic adrenal cortical tissue in testis can develop into the so-
called testicular tumors of the adrenogenital syndrome (Am] Surg Pathol.
1988;12:503).
2. Acquired adrenal cortical hyperplasia is a nonneoplastic bilateral process
characterized by a gland weighing in excess of 6 g. The gland has either a
diffuse, nodular, or combined appearance.
a. Diffuse hyperplasia is usually ACTH dependent and caused by a
hyperfunctional pituitary (most often pituitary adenoma, less often
corticotropin-releasing hormone (CRH) from the hypothalamus) or an
ectopic ACTH/CRH-producing tumor. The latter neoplasms include
small cell carcinoma (usually from the lung), low grade neuroendocrine
carcinoma or carcinoid (usually from the lung or thymus), medullary
thyroid carcinoma, pancreatic endocrine neoplasms, and pheochromo-
cytoma (PHEO). Both glands are symmetrically enlarged. The zonae fas-
ciculata and reticularis are expanded with their relative proportions vary-
ing from case to case. The zona fasciculata is lipid depleted but markedly
expanded by a population of cortical cells with abundant eosinophilic
cytoplasm (e-Fig. 26.5).
b. Nodular cortical hyperplasia is ACTH-independent in most cases. The
glands are markedly enlarged, in excess of 15 to 20 gin some cases. The
cortical nodules may constitute a transformation from diffuse hyperpla-
sia in its late stage; these nodules are yellowish and vary from 0.2 to over
4.0 em. Fasciculata-type clear cells, reticularis-type cells, or a mixture of
these two cell types characterize the nodules. Nodular adrenal corti-
cal disease with discrete nodules of hyperplastic zona glomerulosa with
intervening cortical atrophy is seen in children with McCune-Albright
syndrome (Am] Surg Pathol. 2011;35:1311).
c. Primary pigmented nodular adrenocortical disease (PPNAD), an uncommon
but specific form of ACTH-independent nodular hyperplasia, is generally
diagnosed in the second decade of life with or without the other stigmata
of Carney complex (Orphanet] Rare Dis. 2006;1:21; Best Pract Res
Clin Endocrinol Metab. 2010;24:389). A germline heterozygous inacti-
vating mutation of PRKR1A is found in the Carney complex (65% to
70% of cases) and may also have a role in primary PPNAD (Pituitary.
2006;9:211). The glands are of normal size, but the cut surface has scat-
tered pigmented micronodules, or less often macronodules measuring 1
to 4 mm in size. Uniform compact cells with eosinophilic cytoplasm and
some balloon cells are histologic features of the nodules. Pigmentation is
4X2 I SECTION Vlt ENDOCRINE 5YS1EM
Mnllc.lll:•l- lalluhMIPII...MII'•
Adr••ll- od&noms JUJl)Jatyrnpo•le
Adr•l'ltl eorta1 t~~~n::!nan.a VqtJ
Mnii-IIIJ- urynpll
Bal-..~~· OrUtoii'IOIOp/lol)l...l
Ma..,_.llt phooclu~- C.n>ltl boctt
Oomi>OIIb Dlleocltom0<$:rooa ~ul-
lilllll&bYIIc
S..Cilf' d--
AIJenomlliDkf b.lmor
--
M'111olbolllll S\IIIOI!of pof16011ic
IIIM>r~
U~•IIY blod<llr
Goln.llfono II'Om.t
Anp"""""
ulom)'Oma
Mno tnnor fll:olnor&noll
t1:11 a ,..-.rtte~
- -- ' lloteii.. RA. U<¥1 RY,._P, .. al,ed!l. --~-!lm<lf
1i.lmol.na. Pl~.mJ~ TuMOU12td~ "'JM.t, I.Jm, fl'lnoet IARC P,_; 2004.
l.Mied""" permllllllltln.
Chapter 26 • The Adrenal Gland and Paraganglia I 4 33
..,., ,,.,..,...on
T4 Tumor<>fanylllla-ln- of ad)lconl "'11IN"
I'1,1,! J t-!II
I Compm.tiM bu101t1111 c..,ltt!J.r "*-I Cof11c111'111Ma.,..
Olllef Sllolll• ~ .. Eldnadrellal Neojlllat~.t
JI:f ICI' In tt!CIILo...UIL MIST Uloll' !II!SO
~M +· + + +
Ct( rMJ./U:Sl :!: :!: + +
~ +
CIO
01(20
34,!El2
EIAA
+ -·
:!:
:!:
+
:!:
HEP.f'AR +
COlO ±' :t:• +
CD34 +
CDII7 +
HMB~ +'
MEL-A103
INH
+
+
-·
:!:'
+
eN. +
RCC +
SIAA + :!: +
WTI +
a.._,26•Th&Adl'&ll'lllt01Mdatle1Pa,.....ia I 4J7
I' I.I II ft.l$1 ~omu 1M I'll.,_.lima, •l'llt ~ lllmhl TulftOf
.,.,_
MEN2
..~-~
IM•l•
R£1710q11.2
-IoCr'
Phlll:d\I'OI'ni:IC)tlma
Nouroll- tJpe 1
Ph...:ii'""""Y'>molabOornNI
-IJjkma
Ph""""rom--rq;arWiom• PhlltV!t\~mtJ hM.d, MC:k,
~mol and abdomnl -natoma
Ph"""""""~tqt'lfloma SV/Ialq21-:!3 Heed and nock poltill\IIIOml
~mo3
Ph""""rom--'""rWiomo Ph...:ii'""""Y'>miAiood,I\Od(,
~mo4 111oredc,llld •bdotrlnol
-IJjkma
438 I SECTION VI: ENDOCRINE SYSTEM
have slightly eosinophilic and finely granular cytoplasm, but they may be
amphophilic to basophilic or even oncocytic (e-Fig. 26.14). A finely vacuo-
lated cytoplasm is present in some cases due to lipid accumulation. Intracy-
toplasmic periodic acid-Schiff positive, diastase-resistant hyaline globules are
yet another finding (e-Fig. 26.15). Melanin-containing PHEOs are rare but
well documented (Hum Pathol. 1993;24:420). The polygonal cells can vary in
size and nuclear detail, with marked nuclear enlargement and hyperchromasia,
although less impressive cytologic atypia is often present. Prominent nucleoli
and nuclear pseudoindusions are additional but inconsistent findings (e-Fig.
26.16). Mitotic figures, if seen, are not an indication of malignancy per se.
The stroma may exhibit extensive hyalinization, fibrosis, or rarely amyloid
deposition, and the vasculature may be prominent. The periadrenal adipose
tissue often has a hibernomatous appearance regardless of the age of the
patient (e-Fig. 26.17). The chromaffin cells are positive for chromogranin
A, synaptophysin, and cytokeratin in 25% of cases (Arch Pathol Lab Med.
1990;114:506); thetumorcellsdonotexpress EMA, melanA, orinhibin. S-100
labels sustentacular cells that are usually located at the periphery of the nests.
Prognostic assessment of an individual PHEO which is confined to the gland
and without metastasis is an exercise in uncertainty. A number of morphologic
features of the tumor have been assessed from the architectural pattern (nested
vs. diffuse), presence or absence of confluent necrosis, mitotic rate, Ki-67 index,
as well as several others including weight (less than or greater than 10 g), but
the conclusion that there are "no absolute histologic criteria for predicting
malignant potential" still seems to be true (Histopathology. 2011;58:155).
Overall, 10% to 25% of PHEOs prove to be malignant as demonstrated by
metastasis to bone, lymph nodes, lungs, and liver (e-Fig. 26.18).
Composite or compound PHEO, a rare variant, is defined in most cases as a
PHEO with a ganglioneuromatous (80%), ganglioneuroblastomatous (20%),
neuroblastomatous (< 1% ), malignant peripheral nerve sheath tumor, or neu-
roendocrine carcinoma (extremely rare) component (e-Fig. 26.19) (Arch Pathol
Lab Med. 1999;123:1274; Am] Clin Pathol. 2009;132:69; ] Clin Pathol.
2009;62:659). This variant accounts for 1% to 3% of all PHEOs, and these
tumors are reported in the clinical settings of neurofibromatosis I and MEN
2A (Am] Surg Pathol. 1993;17:837; Am] Surg Pathol. 1997;21:102). There
is also a case with a cortical adenoma (Ann Diagn Pathol. 2011;15:185).
C. Neuroblastic tumors (also known as peripheral neuroblastic tumors) are embry-
onal tumors arising from the sympathoadrenal neuroendocrine system and are
the most common extracranial solid malignancy of childhood (Nat Rev Can-
cer. 2003;3:203; N Engl] Med. 2010;362:2202). About 600 new cases are
diagnosed each year in the United States. Approximately 98% of neuroblas-
tomas are diagnosed by 10 years of age, and 85% to 90% are detected before
5 years of age. The abdomen and/or retroperitoneum is the site of clinical pre-
sentation in 65% of cases; 50% of NBs arise in the adrenal medulla. Though
most neuroblastomas have a number of overlapping histopathologic features,
they are clinically, pathologically, and molecularly divisible into indolent or
aggressive subtypes (Pediatr Clin North Am. 2008;55:97; Curr Probl Cancer.
2009;33:333; Oncogene. 2010;29:1566).
1. Neuroblastoma (NB} is best characterized as having a variegated appearance
since the gross features are dependent on its morphologic composition and
on secondary changes such as hemorrhage, yellowish foci of necrosis and
calcification, cystic degeneration, and fibrosis; some or all of these find-
ings may be found in any one tumor (e-Fig. 26.20). It is often difficult to
judge whether some of the gross changes are spontaneous since resection
may be preceded by adjuvant therapy. A well-circumscribed, soft, gray-tan
tumm; measuring 2 to 10 em in dimension, with or without hemorrhage and
calcifications, is the common appearance of a poorly differentiated NB
SIIJl
I
2A
Ddlltf•
1-INdtu"""\0!111 .,..complol&-,10111
· - ·tum0f10111
Unlolonll lnoomplalo
nopt!wo ""'"' -
Of- mlt:r"""''''< mldLIII
in patients over a wide age range as a solitary, polypoid mass that projects
into the lumen of the intestine and measures up to 7 em in diameter. It is an
infiltrative neoplasm composed of three cell types in variable proportions,
namely spindle cells, epithelioid cells, and ganglion cells (e-Fig. 26.36). The
spindle cells are elongated and have wavy nuclei resembling Schwanni cells,
with S-100 and neurofilament immunopositivity; these cells can envelope
the ganglion and epithelioid cells, analogous to sustentacular cells. The
larger epithelioid cells are arranged in solid nests, ribbons, or pseudoglan-
dular or papillary structures and have neuroendocrine features including
granular eosinophilic to amphophilic cytoplasm with uniform oval nuclei
and finely stippled chromatin (e-Fig. 26.37); these cells express cytoker-
atin, chromogranin, and synaptophysin. The ganglion cells have atypical
features, and there may be a morphologic continuum with the epithelioid
cell population. Recurrences are rare, but metastatic behavior is restricted
to regional lymph nodes or liver U Gastrointest Surg. 2007;11:1351; Ann
Diagn Pathol. 2011;15:467).
VI. OTHER ADRENAL PARENCHYMAL LESIONS
A. Myelolipoma accounts for <5% of primary adrenal tumors and is usually
detected incidentally on imaging studies of the abdomen, although hemorrhage
with pain is a rare presentation (Asian] Surg. 2009;32:172). Several cases have
been discovered in individuals with congenital adrenal hyperplasia and Cush-
ing syndrome (Exp Clin Endocrinol Diabetes. 2009;117:440; Endocr Pract.
2011;17:441). Whilethetumorcan belargerthan20cm and2 kg, it measures 3
to 5 em in most cases. The tumor has a soft yellow to red surface, depending on
the fat content and presence of hemorrhage. The usual microscopic appearance
includes varying proportions of mature adipose tissue admixed with normal
trilineage hematopoiesis (e-Fig. 26.38). Clonality has been demonstrated (Am
] Surg Pathol. 2006;30:838). Lipoma, angiomyolipoma, and liposarcoma are
in the differential diagnosis.
B. Adenomatoid tumor, like myelolipoma, is usually detected as an incidentaloma
(Am J Surg Pathol. 2003;27:969). The tumor is a well circumscribed, solid
or solid and cystic and measures from 0.5 to 9 em. Microscopically, the
tumor is composed of tubules, cysts, papillary structures, and occasional solid
sheets of low cuboidal cells (e-Fig. 26.39). The tumor cells may have cytoplas-
mic vacuoles with signet ring features (e-Fig. 26.40) but are mucicarmine-
negative for mucin. The tumor cells have the same immunophenotype as
mesothelial cells with reactivity for vimentin, CK7, calretinin, and WT-1
(Pathol Res Int. 2010;2010:702472). Adenomatoid tumor is believed to arise
from mesothelial inclusions within the adrenal gland. The main differen-
tial diagnoses include lymphangioma, metastatic carcinoma, and vascular
tumors, especially epithelioid angiosarcoma (Arch Pathol Lab Med. 2011;135:
268).
C. Mesenchymal tumors include lipoma, schwannoma, hemangioma, leiomyoma,
leiomyosarcoma, angiosarcoma, perivascular epithelioid cell tumor, Ewing
sarcoma-primitive neuroectodermal tumor, solitary fibrous tumor, heman-
gioblastoma, and papillary endothelial hyperplasia.
D. Lymphoma is most commonly seen in previously diagnosed cases of non-
Hodgkin lymphomas (NHL) and occurs in as many as 25% of cases at some
point in the clinical course; bilateral involvement is present in 75% to 80% of
cases, and adrenal insufficiency is a known complication. However, rare exam-
ples of NHL with a primary presentation in the adrenal have been reported
(Exp Clin Endocrinol Diabetes. 2011; 119:20 8). Diffuse large B-celllymphoma
is the most common pathologic type (Mod Pathol. 2009;22:1210), although
Burkitt lymphoma also occurs (Tumori. 2007;93:625). Hodgkin lymphoma of
the adrenal is rare (Br J Haematol. 2010;148:341).
444 I SECTION VI: ENDOCRINE SYSTEM
VII. ADRENAL CYSTS are relatively uncommon and are divided into four cate-
gories: parasitic cysts (7%), epithelial cysts (9%), pseudocysts (39%), and
endothelial cysts (45%) (Arch Surg. 1966;92:131; Endocr Pathol. 2008;19:
274).
A. Pseudocysts are discovered as another incidentoma on CT imaging. The usual
appearance is a well-defined cyst with water-like density and a median size of 6
to 10 em in diameter. Pseudocysts are unilocular, are filled with yellow-brown
to bloody amorphous semi-liquid material, and have a wall thickness of 1 to
5 mm. The densely hyalinized connective tissue of the wall may contain focal
calcifications or even metaplastic bone formation, and entrapped cortical tissue
may also be present; the smooth muscle in the wall of the cyst is continuous
with the smooth muscle of the adrenal vein. An identifiable lining is absent. The
differential diagnosis includes a cystic NB or PHEO (Cancer. 2004;101:1537).
B. Vascular cysts are the most common type of adrenal cyst in some series (Arch
Pathol Lab Med. 2006;130:1722). Endothelial cysts are usually well circum-
scribed and multiloculated (e-Fig. 26.41). The endothelial cells are immunopos-
itive for CD31, and if derived from lymphatic endothelium, then also positive
for D2-40 (e-Fig. 26.42A and B).
C. Epithelial cysts are divided into true glandular cysts and embryonal cysts (World
I Surg. 2006;30: 1817), although in some classification schemes cystic adrenal
tumors are also considered in the category of epithelial cysts. Mesothelial cysts
also occur (Endocr Pathol. 2008;19:203).
D. Parasitic cysts are a manifestation of echinococcal infection in the adrenal and
retroperitoneum (Bull Soc Pathol Exot. 201 0; 103:313; Int Surg. 201 0;95:189).
The wall of the cyst is often calcified.
VIII. METASTATIC NEOPLASMS to the adrenal gland are common. Metastatic carcinoma
is present in the adrenals in 25% to 30% of carcinoma-related deaths at autopsy
(Cancer. 1950;3:74). The lung is the most common primary site (adrenal metas-
tases are found in 30% to 35% of cases) (e-Fig. 26.43) followed by the breast;
other malignancies that frequently metastasize to the adrenals include adenocar-
cinomas of the kidney, stomach, and colon (e-Fig. 26.44); melanoma (10% to
15% or greater of cases) (e-Fig. 26.45); hepatocellular carcinoma; and urothe-
lial carcinoma (Clin Endocrinol. 2002;56:95; Am I Surg. 2008;195:363; Semin
Oncol. 2008;35:172). Renal cell carcinoma (RCC) involves the adrenal by either
direct extension from an upper pole tumor or metastasis in 5% to 10% of cases;
metastatic RCC should always be differentiated from a primary adrenal cortical
carcinoma (Am I Surg Pathol. 2011;35 :678; Eur Urol. 2011;60:458) (Table 26.3 ).
Other types of neoplasms arising in the retroperitoneum or as a metastasis to the
adrenal can be differentiated in most cases by immunohistochemistry (see Table
26.3).
IX. ADRENAL GLAND CYTOLOGY
A. Fine needle aspiration (FNA). FNA is frequently used to evaluate adrenal
gland mass lesions and is performed under percutaneous CT and ultrasound
guidance, or endoscopic ultrasound guidance for left adrenal lesions (Diagn
Cytopathol. 2005;33:26). FNA diagnosis of adrenal lesions has an accuracy of
98% and specificity of 100% (Diagn Cytopathol. 1999;21:92). FNA biopsy
of pheochromocytoma is regarded as a relative contradiction due to possible
induction of hypertensive crisis (Radiology. 1986;159:733).
B. Specific Neoplasms
1. Myelolipoma. The aspirate shows a mixture of mature adipose tissue and
hematopoietic elements, including nucleated red blood cells, granulocytes
and precursors, and megakaryocytes (Acta Cytol. 1991;35:353).
2. Adrenal cortical neoplasms. The distinction between adrenal cortical hyper-
plasia, adrenal cortical adenoma, adrenal cortical carcinoma, and normal
Chapter 26 • The Adrenal Gland and Paraganglia I 445
*All e-figures are available online via the Solution Site Image Bank.
446
Chapter 27 • Pituitary Gland I 44 7
result from compression of the infundibular stalk, which blocks the transport
of dopamine (formerly, PRL inhibitory factor) from the hypothalamus. Some
macroadenomas may compress the normal pituitary tissue and cause panhy-
popituitarism. In many cases, macroadenomas are detected only after they
compress the optic chiasm, producing bitemporal hemianopsia or other vision
disturbance. Approximately half of nonfunctioning adenomas are referred to as
null-cell adenomas. These chromophobic (or less commonly, oncocytic) tumors
have a negative hormonal immunoprofile despite showing reactivity for synap-
tophysin and exhibit few if any secretory granules on ultrastructural analysis
(oncocytic adenomas show abundant mitochondria). Although some null-cell
adenomas appear to express the alpha subunit of the glycoprotein hormones
(LH, FSH, TSH) and thus might be considered "silent" gonadotrophic or thy-
rotrophic adenomas, expression of the alpha subunit has also been documented
in somatotroph adenomas (Neurol Res. 1999;21:247), so this feature may have
limited specificity.
Microscopically, adenomas lack the normal reticulin-rich acinar structure
of the adenohypophysis. Instead, they may appear patternless, pseudorosette-
rich, papillary, endocrine/organoid, or paraganglioma-like and may focally form
glands. In ambiguous cases, a reticulin stain may be useful for accentuating
loss of acinar architecture. Adenoma tumor cells are typically larger than their
nonneoplastic counterparts and have round-to-oval nuclei, delicate stippled
("salt and pepper") chromatin, and small or inconspicuous nucleoli. Occasion-
ally, adenomas exhibit moderate to marked cytologic atypia, but this feature is
not associated with aggressive behavior unless accompanied by elevated mitotic
and/or proliferation indices (see section on "atypical pituitary adenoma"). Cyto-
plasmic quality varies within and among different adenoma (hormonal) sub-
types and may even vary among cells within an individual tumor, particularly in
some tumors that secrete more than one hormone. Cytologic, histochemical, and
ultrastructural features can be used to subtype adenomas. Although immuno-
histochemical profiles of hormone expression and cell proliferation are now in
greater use for this purpose, familiarity with morphologic and histochemical
features remains worthwhile.
Indirect histologic dues toward adenoma biology (hormonal secretion pat-
terns) include the presence of calcium and amyloid bodies (rare) in prolacti-
noma. In prolactinoma treated with dopamine agonists, fibrosis and smaller cells
with high nuclear to cytoplasmic (N/C) ratios are common. Perivascular pseu-
dorosettes usually indicate a gonadotrophic or null-cell adenoma (e-Fig. 27.5).
Deposits of Crooke's hyaline (ringlike cytoplasmic cytokeratin inclusions) most
commonly accumulate in nonneoplastic corticotrophic cells in patients with
hypercortisolism of any cause, including, but not limited to, Cushing disease
(see section on corticotroph adenoma). Strong PAS staining suggests a corti-
cotroph adenoma, whereas weak PAS positivity is seen in glycoprotein (FSH,
LH, TSH expressing) adenomas. Round, weakly eosinophilic, cytokeratin pos-
itive, paranuclear "fibrous bodies" in a relatively chromophobic tumor most
often indicate a GH-producing adenoma (e-Fig. 27.6).
B. Prolactinoma. Also called lactotrophic adenoma, prolactinoma comes to clinical
attention most commonly in women of reproductive age with amenorrhea and
galactorrhea. Men with prolactinoma are usually asymptomatic or present with
decreased libido. Patients with prolactinomas are often treated medically with
a dopamine agonist (e.g., bromocriptine) to impair adenoma cell growth and
inhibit PRL production, but such agents are not cytotoxic. Tumors resected post-
therapy usually show interstitial fibrosis, reduced cell size, and high N/C ratio
and may be reminiscent of small-cell carcinoma (e-Fig. 27.7). However, they are
usually at least focally positive for PRL and display a low mitotidproliferative
index.
Chapter 27 • Pituitary Gland I 44 9
are formed by closely apposed polygonal cells with small nuclei and generous
granular eosinophilic cytoplasm. Spindled/fascicular areas may also be present.
Lymphocytic infiltrates are common and may be robust. Staining with PAS is
resistant to diastase digestion. Immunoreactivity is strong for S100 and TfF-1,
and variable for the lysosomal marker CD68; reactivity for pituitary hor-
mones, synaptophysin, cytokeratins, neurofilament, and chromogranin A is not
observed. Ultrastructurally, these cells show abundant phagolysosomes and no
evidence of neurosecretory granules.
H. Craniopharyngiomas (discussed in Chap. 41).
V. LOW-GRADE NEOPLASMS
A. Atypical pituitary adenomas are histologically similar to benign pituitary ade-
nomas except for elevated mitotic and proliferative indices. For this diagnosis,
current World Health Organization criteria require nuclear immunoreactivity
for the proliferation marker Ki-67/MIB-1 in >3% of tumor cells and extensive
nuclear immunoreactivity for p53; others suggest that Ki-67 labeling > 10%,
regardless of p53 status, correlates with more aggressive behavior and war-
rants an "atypical" designation (Neuroendocrinology. 2006;83:179). Relative
to benign adenomas, these tumors are more likely to invade adjacent structures
and to recur.
VI. HIGH-GRADE NEOPLASMS
A. Pituitary carcinoma is a very rare neoplasm that cannot be diagnosed by its innate
appearance or any known markers; instead, it is diagnosed by the presence
of craniospinal or extracranial metastases. The Ki-67/MIB-1 and p53 labeling
indices are typically high, but cytologic and histologic features are often benign.
These tumors are usually functional, secreting PRL or ACTH. Its designation in
this chapter as a high-grade neoplasm may be inappropriate in the truest sense,
but the term is applied here to reflect its more aggressive clinical behavior.
SECTION VII
Reproductive Tract
I. NORMAL ANATOMY. The normal adult testis is an ovoid paired organ, measuring
4.5 x 2.5 x 3 cm3 , and weighing approximately 20 g. The testes are suspended
within scrotal sacs by spermatic cords. The testis is covered by a capsule composed
of an outer tunica vaginalis lined by mesothelium, the collagenous tunica albuginea,
and the inner tunica vasculosa. The tunica vaginalis forms a sac filled with serous
fluid. The posterior portion of the testis not covered by a capsule is called the
mediastinum and contains blood vessels, nerves, lymphatics, and the extratesticular
rete testis.
The testicular parenchyma is subdivided into lobules containing seminiferous
tubules separated by fibrous septae. The terminal portions of the seminiferous
tubules drain into the tubuli recti that connect to the tubules of the rete testis at the
mediastinum. The tubules of the rete testis anastomose with the ductuli efferentes,
which form the head of the epididymis and empty into the vas deferens, which
traverses the inguinal canal as a component of the spermatic cord. The testicular
artery arises from the aorta and is the major source of vascular supply to the testes.
The venous drainage occurs through numerous small veins that form a convoluted
mass known as the pampiniform plexus that surrounds the testicular artery. These
small veins anastomose to form the right testicular vein, which drains into the
inferior vena cava, and two left testicular veins, which drain into the left renal
vein.
Histologically, prepubertal and postpubertal seminiferous tubules are quite dif-
ferent. Prepubertal tubules are small, with few or no lumina, and contain mostly
Sertoli cells with a few primordial germ cells (e-Fig. 28.1).* Postpubertal seminif-
erous tubules are larger and harbor Sertoli cells and germ cells at varying stages
of maturation (e-Fig. 28.2). The Sertoli cells abut the basement membrane and
are aligned perpendicular to the membrane; their nuclei are round to oval with
prominent nucleoli, and the cytoplasm has Charcot-Bottcher crystals, which can
occasionally be seen by light microscopy. Within the seminiferous tubules, the least
mature germ cells-spermatogonia-are present along the basement membrane,
with the most mature cells--elongate spermatids-found at the luminal border.
Primary and secondary spermatocytes are found in an adluminal position.
The interstitial tissue between the seminiferous tubules contains Leydig cells,
vessels, and connective tissue. Leydig cells are arranged singly and in clusters
*All e-figures are available online via the Solution Site Image Bank.
451
452 I SECTION VII: REPRODUCTIVE TRACT
(e-Fig. 28.3), and can be associated with nerves. They are large and irregularly
spherical to polyhedral, with small spherical nuclei and abundant acidophilic cyto-
plasm. The cytoplasm can exhibit lipofuscin and rod-shaped Reinke crystals.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION. Tis-
sue samples of the testes received for surgical pathologic examination include testic-
ular biopsies, and unilateral and bilateral orchiectomy specimens. Retroperitoneal
lymph node dissection can be performed as part of a staging maneuver for testicular
cancer.
A. Fine needle aspiration biopsy of the testis in infertile patients (with sperm aspi-
ration and cytopathologic examination) is occasionally performed. Cytologic
touch imprints or wet preparations may be made at the time of open testicular
biopsy in infertile patients to rapidly identify the presence of sperm. The role of
cytology in the evaluation of testicular tumors is limited to diagnosis of lymph
node metastases by fine needle aspiration. Although seminomas can usually be
differentiated from nonseminomatous tumors, subtyping of nonseminomatous
tumors cannot be reliably performed by cytology.
B. Testicular biopsies, which can be open or percutaneous, are typically performed
for evaluation of infertility. They are usually contraindicated in the evaluation of
solid testicular masses, with the possible exception of epidermoid cysts, which
can be removed by excisional biopsy. Testicular biopsy specimens are usually
received in Bouin's fixative. An accurate documentation of the number and
size of the biopsy fragments and exact site(s) of the biopsy for each container
should be made during gross dictation. The biopsy fragments should be inked
with hematoxylin to facilitate identification during embedding, wrapped in lens
papet; placed between sponges, and processed entirely. Three hematoxylin and
eosin (H&E)-stained slides, each with three to four serial sections, should be
prepared from each block.
C. Unilateral simple orchiectomy is performed in cases of testicular torsion. Gross
examination of the testis similar to that for tumor cases should be performed.
One section of the testicular parenchyma in relation to the capsule, and a section
each of the epididymis and spermatic cord, should be submitted with description
and sampling of focal lesions.
D. Radical orchiectomy is performed for testicular tumors. The specimen consists of
the testis and paratesticular organs (surrounding tunica vaginalis, epididymis,
soft tissue, and a segment of spermatic cord). In cases of tumor resection, the
specimen should ideally be sent fresh and intact to the surgical pathology labora-
tory for immediate gross examination. Alternatively, when delay is anticipated,
the specimen is placed in 10% buffered formalin and sent intact. In such cases,
tumor morphology is often suboptimal due to poor fixation. The surgeon may
occasionally bisect the specimen to aid fixation; this approach is not recom-
mended as it prevents evaluation of involvement of the tunica by the tumor as
well as procurement of fresh tissue for ancillary studies.
The specimen is weighed, and measurements are recorded in three dimen-
sions. The length and diameter of the resected segment of spermatic cord are
noted separately. The external surfaces of the testis and spermatic cord are
examined for involvement by tumor. The proximal shave resection margin of
the spermatic cord is submitted in a separate cassette, and then the cord is
serially sectioned and inspected for tumor involvement; representative sections
are submitted proximal to distal. The tunica vaginalis is opened anteriorly to
show the tunica albuginea; presence of fluid, if any, within the sac is noted.
The testis is then bisected anteroposteriorly through the epididymis, and serial
sections are made parallel to this plane. Each slice is examined, and the tumor
is described in relation to the epididymis and the tunica albuginea. The size,
colot; and consistency of the tumor should be noted. Foci of hemorrhage, necro-
sis, and variegation, as well as multifocality, if present, should be described.
Chapter 28 • Testis and Paratestis I 4 53
Photographs or digital images should be taken and tissue procured for tumor
bank and ancillary studies such as flow cytometry and karyotyping, if necessary.
The specimen should then be fixed overnight in an adequate amount of 10%
buffered formalin prior to submission of one section per centimeter of tumor.
Representative sections should include heterogenous areas and sections of tumor
in relation to uninvolved parenchyma, epididymis, and tunica albuginea. One
section of grossly normal-appearing parenchyma should be included. If correla-
tion of identified histologic tumor type with serum markers (alpha-fetoprotein
[AFP] and human chorionic gonadotropin [hCG]) is not achieved, additional
sections should be submitted (note that such correlation will not always be per-
fect because metastatic deposits may harbor different elements than the primary
tumor).
E. Retroperitoneal lymph node dissection is performed as a separate procedure. The
specimen is received in 10% buffered formalin. During gross examination, the
tissue fragments should be measured in aggregate and carefully dissected to
harvest as many lymph nodes as possible, and the size of the largest and the
smallest putative nodes should be noted. Possible foci of tumor encountered
during dissection should be measured and sampled. An effort should be made
to sample any area(s) suspicious for viable tumor.
F. Bilateral orchiectomy specimens may be submitted as part of treatment of car-
cinoma of the prostate. Gross examination and sectioning are similar to that
for unilateral simple orchiectomy. Prostate cancer is rarely encountered within
these specimens.
Ill. DIAGNOSTIC FEATURES OF BENIGN DISEASES OF THE TESTIS
A. Congenital abnormalities
1. Cryptorchidism is maldescent of the testis in which the testis is found, after 1
year of age, to be located high in the scrotum, within the inguinal canal,
or in an intra-abdominal location. Grossly, the prepubertal undescended
testis differs little from normal, but after puberty the undescended testis
is smaller. Histologically, there is a progressive loss of germ cells with age,
along with decreased size of seminiferous tubules and increased thickness of
tubular tunica propria. Sertoli cell nodules (e-Fig. 28.4), which are foci of
tubules containing immature Sertoli cells and laminated calcific deposits, are
often seen in cryptorchid testes. These are likely hyperplastic foci, although
they have also been termed tubular adenoma of Pick. Rarely, Sertoli cell
nodules can present as a mass (Am ] Surg Pathol. 2010;23:1874). The
major complications of cryptorchidism are infertility and germ cell neo-
plasia, ranging from intratubular germ cell neoplasia (IGCN) to invasive
germ cell tumors, particularly seminoma, embryonal carcinoma, and embry-
onal carcinoma/teratoma. For patients > 1 year of age, placental-like alkaline
phosphatase (PLAP) and CD117 immunostains can be useful in highlighting
intratubular germ cell neoplastic cells.
2. Anorchism and polyorchidism are absence of testes and more than two testes,
respective!y.
3. In testicular-splenic fusion, encapsulated splenic tissue is found adjacent to
the left testis, which can show germ cell aplasia in the seminiferous tubules.
4. Adrenal conical rests are usually incidental, millimeter-sized nodules of
adrenal cortical tissue that are detected along the pathway of descent of
the testis, including along the spermatic cord and testis.
B. lnfenility. The causes of infertility may be pretesticular, which include endocrine
disorders involving the pituitary and adrenal glands; testicular, including genetic
disorders; or posttesticular, which are mainly obstructive and include varicocele
and cystic fibrosis. The evaluation of the patient includes a detailed clinical
history, physical examination, semen analysis, tests of endocrine function, anal-
ysis of sperm function, and serology for antisperm antibody. Testicular biopsy
..... I SECTION VII· REPRODUCTIVE TRACl
,,/,t!i Jl !\I
- -.""
Repo111111 lni..UittyliojMIM
.. ,_,_,penn
b.
c.
..
..... __
1. -odololiiOI"..J:,.mplo
On:lllooto,.,.-
~
2. P-..e.'tbooncooltoo11eulllrllloue
a. s.m-.l'oloii411Jbuleo
a. Numbtlr
b. Tunica proprll, Nonnlllllllckonod
c. ,._, numbor of ~b&M {oount 20 lull&lloo)
d. _ _ .,..<ll..,.nmalop'"*
•• SoriD!I- - - -
... lrl18ndllm
•. '-"11111:- p_,tfobiiaillli)""plulle
-·--manit
b. MY&rmrllnbnllllallnftom-
c. Pr111nca d mta'tlphaaaand mast celt
!. 1. [jlldll)nft PlliOI!ntllbiii!nt
b. V..d.W..,., Pt.ntloboort
c. lmmunoh....,h"'*"lll!>ee~l.iilllna
f.H~dlo.!;laM
1. Tho mot!.,.,_ h~ pt11.wn b'
b. Th& pn><bnlni111 hliillq)o pallom 110
Chapter 28 • Testis and Paratestis I 4 55
........--,-
Chotloca-llnd touwlomll4!mb(\'!Nic:ormoma
0111101
Purt forma
L"fdi.Joell tunv
Ma..,_.llt l<!tdllcoi11Jmor
s.n.t<dltum«
- Ml .,..,,llpld-lt:ll v.actont
_...,._coil tumor
~C'JOOIIk:alcl(j1~s..tol col lumot
Ma..,_.ntSo<IDII<dt.mor
Gllnulcoo ... !limo<
hlul~D&Il11lUiooo <dl tu,...-
Jwonl~ IVIJIUiaaa oellblmor
1\JmOfl of!M !Moo<MIItbn>rna 1!701JP
Thloom•
ft'"""-
S..~IIamettnnor
llcoml>loloiY dlll'ell1111otod
s.. conl/aollodll otromt~llllrno,., mlltldfMno
""""'rt- oold/g<Ndol-lltumora
Tummoonblll~ both pm Mland,.. Clll'dJiii>Nidllstrom.ot...,_
GoNdablum.
GofmoeiiiOl~olflromolt.mor, ""*"''''od
a..~ .2:8 • Tee:t:ie tnd Plltateetia I ... ,
I' f.t:] I f ~: fj I WHO Hlttololfc Cllul1'kl:tbl•t 1-.. of ttte T..Ua 1ncl ,..teltf.t
' - (CIIIIIIuilt
•-••..,11-tftlo-
c..t!oldtl.mor
Tumoroof-nepllld~l~
_ _, cf bordeltfto molalii/ICY
SelaJa: C:.rdftOII"'''Il
W.ll-dll'll,_ en<lo- .,.,..,..,,.
Mudnow:QJ~:tld«<cn,..
Mudn,..Ofl-..:on:h>ma
Brer~nar tumw
,.,Dhrol>iaCimo
Pn.lalllllorno
11n1111 1 u~
n-tfco~..._n*•UI'IIIt
AdOIIOINI
c..t!omo
n-tl'l*llltiCIIIf-
Ad.,.mmldtumor
Molpnl moso-••
BMIIJn-olomo
W.ll-<!lla- IJillllby m - . . .
C)'*.-olome
Ad ......n:m.... cfllio ·~tt.'rnl>
OJIDdon""'•
......
PoplloiJ
.....
O.•IOIIIMII<..,.II round colllunu
_
458 I SECTION VII: REPRODUCTIVE TRACT
Emb!)o>nll + + + + + v + +
an:h>m.t
Yolk ... tunv
Sodolll.e)olla
+
- + +
-tv -
v
lid
+ +
lid -tv lid
- v- -111+
<dltUil'l«
v - -
~""-
lid lid +
460 I SECTION VII: REPRODUCTIVE TRACT
and/or glandular pattern (e-Fig. 28.21). Nuclei are polygonal and vesic-
ular with coarse chromatin. Mitotic activity and necrosis are extensive.
IGCNU can be present at the periphery and frequently displays intratubu-
lar necrosis. Vascular invasion can be seen (e-Fig. 28.22) and should be
differentiated from retraction artifact and artificial implantation during
sampling. Pan-cytokeratin, SALL4, OCT4, and CD30 positivity is seen
in tumor cells; PLAP and AFP are only focally positive in pure tumors.
Epithelial membrane antigen (EMA) is negative, which is important in
the differential diagnosis with somatic carcinomas (Table 28.3). Evalua-
tion of H&E-stained slides is usually sufficient to establish the diagnosis;
in occasional cases, immunohistochemistry is needed to help differentiate
embryonal carcinoma from yolk sac tumor, seminoma, anaplastic large-
cell lymphoma, and/or choriocarcinoma.
e. Yolk sac tumor (endodermal sinus tumor) shows differentiation reminiscent
of embryonic yolk sac, allantois, and extraembryonic mesenchyme. In
infants and young children, it tends to occur in pure form, whereas in
adults it is found as a component of malignant mixed germ cell tumors.
The morphologic appearance is varied and includes reticular (most com-
mon), microcystic, endodermal sinus-like (with Schiller-Duval bodies),
papillary, solid, glandular, alveolar, enteric, polyvesicular vitelline, and
hepatoid patterns (e-Fig. 28.23). Rarely, a neoplastic spindle cell compo-
nent has been observed in association with the myxomatous and reticu-
lar variants. The immunoprofile reveals AFP, glypican 3, SALL4 (e-Fig.
28.24), PLAP, and low-molecular-weight cytokeratin positivity. IGCNU
is commonly seen in adult yolk sac tumors but not as frequently in child-
hood yolk sac tumors. Embryonal carcinoma may, in certain foci, be dif-
ficult to distinguish from yolk sac tumor, and indeed the two tumor types
can appear to merge; however, embryonal carcinoma nuclei are usually
more pleomorphic, and immunostains can help in difficult cases (Table
28.3). Follicle-like areas of granulosa cell tumors in infants may resemble
the solid and cystic pattern of yolk sac tumor, and the enteric pattern may
appear similar to glandular areas in teratoma; usually, the former problem
can be resolved with immunohistochemical stains for AFP and inhibin.
f. Trophoblastic tumors are almost always choriocarcinoma. Pure chorio-
carcinoma is extremely rare; instead, choriocarcinoma most commonly
occurs as a component of mixed germ cell tumors. Grossly, necrotic
and hemorrhagic nodules can be observed. Microscopically, the more
viable peripheral areas show randomly admixed syncytiotrophoblasts,
cytotrophoblasts, and intermediate trophoblasts, although one compo-
nent may predominate giving rise to a monophasic tumor (e-Fig. 28.25);
the tumor has a propensity for vascular invasion (e-Fig. 28.25). Very rare
tumors composed of intermediate trophoblastic cells resembling placen-
tal site trophoblastic tumor have also been reported. The syncytiotro-
phoblasts are positive for hCG, the alpha subunit of inhibin, and EMA,
and the intermediate trophoblasts are reactive for human placental lac-
togen (HPL); all cell types are positive for cytokeratin. The differential
diagnosis includes syncytiotrophoblast-rich seminoma, and isolated syn-
cytiotrophoblasts found in nonseminomatous germ cell tumors.
g. Teratomas include mature and immature teratoma, dermoid cyst, mon-
odermal teratoma, and teratoma with somatic-type malignancies. The
age distribution is bimodal. Teratomas occurring in children are benign,
whereas those in young adults have significant rates of metastases despite
their histologic appearance. Grossly, there are cystic and solid areas,
and cartilage (e-Fig. 28.26) and bone may be evident. Microscopi-
cally, teratomas may show well-differentiated elements derived from one
Chapter 28 • Testis and Paratestis I 4 61
SOX2 only labels embryonal carcinoma and teratoma (Am J Surg Pathol.
2007;31: 836; f Pathol. 2008;215: 21) and SOX17 labels IGCNU, classic
seminoma, and yolk sac tumor ( J Pathol. 2008;215: 21; Am J Clin Pathol.
2009;131:131). Among these markers, SALL4 demonstrates the highest
sensitivity and showed particular utility for yolk sac tumors. For yolk sac
tumors, SALL4 is much more sensitive than PLAP, AFP, and glypican-
3 (Am J Surg Pathol. 2009;33:1065). SALL4 is also particularly useful
in differentiating metastatic germ cell tumors from metastatic carcinoma
of nontesticular origin, a setting in which it has been found to be more
sensitive than OCT3/4 (Cancer. 2009; 115:2640).
More recently, an RNA-binding protein UN28 has been discovered as
a novel sensitive and general germ cell tumor marker, which shows sensi-
tivity similar to SALL4 (Hum Pathol. 2010;42:710). A potential diagnos-
tic pitfall that must be kept in mind is that all new markers described above
are also expressed in early fetal germ cells. SALL4 is also expressed in sper-
matogonia in both children and adults (Am JSurg Pathol. 2009;33:1065).
k. Molecular genetics is not currently used in diagnosis. The only exception
is detection of isochromosome 12p by fluorescence in situ hybridization
to identify metastatic germ cell neoplasms, since gain of material in 12p
is the most common structural chromosomal alteration in invasive germ
cell tumors (Mod Pathol. 2004;17:1309).
B. Sex cordlgonadal stromal tumors comprise 4% to 6% of all testicular tumors in
adults and include Leydig cell tumor, Sertoli cell tumor, granulosa cell tumor,
thecoma, and fibroma.
1. Leydig cell tumors are the most common and are known to occur in patients
with gynecomastia, Klinefelter syndrome, and cryptorchidism. Grossly, the
tumor cut surface is homogeneous, solid, and brown to yellow. The tumor
cells are large and polygonal, and have abundant eosinophilic, lipid-laden
cytoplasm (e-Fig. 28.32). The characteristic Reinke crystals are found in only
30% of cases. The tumor cells are positive for inhibin, calretinin, and Melan-
A by immunohistochemistry. The main differential diagnoses are Leydig cell
hyperplasia and syndromic adrenogenital tumors. Leydig cell tumors form a
nodule without seminiferous tubules, whereas in Leydig cell hyperplasia, the
foci are bilateral and multifocal, and wrap around and extend between the
tubules (e-Fig. 28.33); adrenogenital tumors are bilateral and dark brown,
with pigmentation and fibrous stroma. About 90% of Leydig cell tumors are
benign, but constellation of features, including size > 5 em, cytologic atypia,
increased mitotic activity, necrosis, and vascular invasion, favors malignancy.
2. Sertoli cell tumors account for only 1% of all testicular tumors. Grossly,
the mass is usually well circumscribed, with tan-yellow to white, sometimes
hemorrhagic cut surfaces (e-Fig. 28.34). The pathologic subtypes are the
lipid-rich, large cell calcifying (e-Fig. 28.35), and sclerosing variants. The
known clinical associations are with Carney syndrome, Peutz-Jeghers syn-
drome (which can be associated with bilateral tumors), and androgen insen-
sitivity syndrome. Histologically, the cytologically bland cells are arranged
in tubules, potentially with retiform, tubular-glandular, and solid nodular
areas. The tubules can be closed (solid) (e-Fig. 28.36) and are surrounded by a
basement membrane. Intratubular growth can be present. Sertoli cell tumors
should be distinguished from small incidental Sertoli cell nodules (benign
and thought to be nonneoplastic) as can be seen in cryptorchid testes. The
tumor cells are cytokeratin and sometimes inhibin and calretinin positive,
but PLAP and OCT4 negative, by immunohistochemistry. Malignant Sertoli
cell tumors are rare.
3. Granulosa cell tumors and thecoma/fibroma tumors are similar in appearance
to those in the ovary, and are rare in the testis. There are two granulosa cell
tumor variants in the testis, the adult and juvenile types, as in the ovary.
Chapter 28 • Testis and Paratestis I 4 63
C. Mixed germ cell and sex cord/gonadal stromal tumors include gonadoblastoma,
most commonly seen in the setting of mixed gonadal dysgenesis, ambiguous
genitalia, and 45X/46XY mosaicism. Microscopic examination shows two cell
populations: a germ cell component resembling seminoma and a component
resembling immature Sertoli cells (e-Fig. 28.37). Round deposits of basement
membrane-like material and coarse calcification are common features. A large
number of patients develop invasive germ cell tumors, particularly seminoma,
and so patients are treated with bilateral orchiectomy.
D. Miscellaneous tumors of the testis include carcinoid tumors, tumors of ovarian
epithelial types (serous borderline tumm; serous carcinoma, mucinous cystade-
nomas and cystadenocarcinomas, Brenner tumor, and endometrioid carcinoma),
nephroblastoma, and paraganglioma.
E. Hematolymphoid neoplasms, of which the most common is malignant lymphoma,
comprise 5% of all testicular malignancies. These are the most common bilat-
eral tumors of the testis, and their incidence is higher in elderly men. The most
common subtype is diffuse large B-celllymphoma. The growth pattern is typ-
ically intertubular (e-Fig. 28.38), and the main differential diagnosis is with
seminoma, especially the spermatocytic type. Immunostains can be helpful in
establishing the diagnosis (Table 28.3 ). The prognosis is generally poor. Young
age, low stage, and presence of sclerosis are indicators of a good prognosis.
Isolated plasmacytoma of the testis is rare.
F. Tumors of the collecting ducts and rete. Benign tumors of the rete include
adenoma, cystadenoma, and adenofibroma. Adenocarcinomas of the rete are
rare, and their diagnosis is subject to strict histologic criteria that include
tumor centered on testicular hilum, morphology distinct from any other testic-
ular/paratesticular tumot; solid growth pattern, transition between tumor and
normal tissue, and absence of histologically similar extratesticular malignancy
(especially lung and prostate). The main histologic patterns are tubular, pap-
illary, and solid. The main differential diagnoses are extratesticular adenocar-
cinomas and mesothelioma. The tumor shows extensive regional spread and
distant metastasis, and the overall prognosis is poor.
G. Tumors of the paratesticular organs.
1. Benign. The most common benign neoplasm of the testicular adnexa is adeno-
matoid tumor, representing almost 60% of all cases. These are of mesothelial
origin (Semin Diagn Pathol. 2000;17:294) and arise in the upper or lower
pole of the epididymis as solitary, round to oval nodules invariably <5 em
in size. Histologically, the tumor shows round to oval or slit-like tubules
in a fibrous, hyalinized, and/or muscular stroma (e-Fig. 28.39 and 28.40).
The lining cells are columnar or flat with vacuolated cytoplasm. Adenoma-
toid tumors show immunoreactivity for cytokeratin, EMA, and mesothelial
markers including calretinin and Wf-1. The tumor should be differentiated
from signet-ring-cell carcinoma and mesothelioma; tumors with a more dif-
fuse growth pattern simulate Sertoli or Leydig cell tumors (inhibin positivity,
lipofuscin pigment, and presence of Reinke crystals favor Leydig cell tumor).
Another benign paratesticular tumor is papillary cystadenoma of epi-
didymis (e-Fig. 28.41). About two-thirds of cases are seen in patients with
von Hippel-Lindau disease, and in this setting, the tumor tends to be
bilateral.
A much rarer entity is the retinal anlage tumor or the melanotic neuroec-
todermal tumor, which is composed of two cell populations: larger melanin-
containing cells and smaller neuroblast-like cells.
2. Malignant. Malignant mesothelioma of the testicular adnexa has a micro-
scopic appearance and immunophenotypic profile similar to those of
mesothelioma of the pleura. The main differential diagnoses are with adeno-
matoid tumot; which is better circumscribed, and with carcinoma of the rete
testis.
484 I SECTION VII: REPRODUCTIVE TRACT
pT1 pT2
= with vascular/
lymphatic invasion
pT3 pT4
Figure 28.1 Pathologic staging of germ cell tumors of the testis. (Modified from:
Greene Fl., Compton CC, Fritz AG, et al., eds. A]CC Cancer Staging Atlas. New York,
NY: Springer; 2006.)
,,, , ..... (!)
plX PrfrNuytumorcannotbaas.....ad
pTO Noaoldonoaol~lllmo< (o... ,hb!Wp:tcOr in11>o~
P'lll IGCN <co"*'«na 1'1 otul
pll '1\/...,..llmbd l.o!M-•rod Ojlidldyml& -..ut""""lu/l.ll'nphllle
lnvulon, wrna· 111111.,.,.,•
.,.,1M tuna albiJBj.,.. bill n.:tlh•tunla!
"''lllnlk
pT2 Tu...,llmbd l.otho-•rod opidldyml&- _..rll)mphltlc 1-,
OfiLmol -ln,t
11\oU\IcaVtlllnolo
Ill""'"'""'
tunica ollluill""'-""'
In~ of
p'TS '111....-lnlladoo lllo GI*TIIo.1ic COfd Mill or~-ulon'bml>hollc In~
-
plol 'lllnwln\18d""lll&"""""'- 0<-..11110&11......,111/lyTnphllle lnvul<>n
..., u,,,.._!ll
r« R"'l)onnllymph ncdOI .... nat bo 1010011011
NO No """"'I lim~ !lOde mollotaolo
Nl M-.ol&- •lim~ nodo m.... or ;::2 ern In ~UMWt < 1 - or
mu._,l& tlmph node~,"'"'" :.2em In .,...,_d-slon
M.WWI&- a lymph nod& musof >2 an but nat >5 an In ..,...0.1
<lmomm;a'mulllplolymphncdoo,anyono,._ >2an but not
>0 em In .,-..r\DIIIdl"""...,
M-1&- a lim~ node mooo ..S<tn l'lll)llii1Botdmenol<>n
1
Rop>nlllymph nod• -nat""""
No ,.....I lymph mc1t ,.,.,,.,.,.
M-lntlh a lymph nodom. .at ;::2c:mln.,-..r\DIIIdlmoo...,and
:!15 IIOdea ~. 110110 •2"" In~~- dl'nonoion
M-1&- a lim~ nodo mooool' •2"" but nat >5"" In • -
dml!ltliSb'l; t:1 >5 node::l ~.!'10M >5 an; ot M'lddno&of .,..I'IIXlal
-•!tumor
pN3 M.WWI&- a lymph nodo rnus >San In"""""' dmensl<>n
IWI1t•1t1111dt OIQ
MO No<lotortrr•-""•
Ml Dlalam..........U.
Mle Non,.,.,.I nodal or pumorwy m-.sl&
Mlb Dlllontmol&llallldllorlllen nonropwllrmph n-arolllnp
·---~~~
SX MOII<er otudloo nat....,loblo Of nat~
$0 MOII<er oludJ lewll...t!in nonnolllmb
Sl I.DH <1.5 x N' •nd hCG (lllllimU <Sil(l)orod AFP !nii/IIIU < 1000
S2 I.DH 1.5-10" Nor hCG. (lllh>'mU .500()...51),000 Of AFI' ~U
1000..10,000
S3 I.DH >10 x N Of hCG. (llllutmU >fiO,OOOor AfP (npU >10.000
.... I SECTION VII· REPRODUCTIVE TRACl
'' 1,1,! I J}(51 TN II hill Ill Sche•a foo Ceml c.! I , . _ ol111e lltta (fHIIIflll...e
~~qo .......
pllt NO so
Slqj:IO
Slqj:ll pTl-4 NO ""
"" Sl(
SQIIA PTl NO so
fllqol9 pT2
Pnl
NO
NO
""
"" so
so
pT4 NO ""
"" so
Alrf plfrx NO s1-a
SlqtiS
Slqj:lll Alrf plfrx Nl-.3 ""
"" Sl(
SQIIIA /vry Pllf'l. Nl so
/vry Pllf'l.
Nrr pT/T'I.
Nl
N2
"""" Sl
so
fllqo 119
Alrf plfrx N2 ""
"" Sl
Alrf plfrx N3 so
SlqtiiC
Alrf plfrx N3 ""
""
Ml
$1
Slqj:llll Alrf plfrx M,N Sl(
SQIIII~ /vry Pllf'l. AllYN Mia so
Nrr pT/T'I. AnyN Mia Sl
Nrr pT/T'I. Nl-3
fllqo 1119
Alrf plfrx AnyN ""
Mia
S2
S2
Alrf plfrx Nl-.3 sa
SlqtiiiC
Alrf plfrx M,N ""
Mia sa
/vry Pllf'l. AllYN Mlb Any$
I. NORMAL ANATOMY. The normal weight of the prostate is 20 g for ages 20 to 50, and
30 g for ages 60 to 80. Anatomically, the prostate gland comprises three zones: cen-
tral zone, transition zone (where benign prostatic hyperplasia [BPH] occurs), and
peripheral zone (where most carcinomas originate). (e-Fig. 29.1). *Microscopically,
the normal adult prostate is a branching duct-acinar glandular system embedded
in a dense fibromuscular stroma (e-Fig. 29.2). The epithelium has two layers: a
luminal or secretory cell layer and a basal cell layer. Central zone epithelium can
normally have architectural patterns that include cribriform and Roman bridge-like
structures (e-Fig. 29.3 ).
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION.
The most common prostatic parenchymal tissue samples examined in surgical
pathology laboratories in the United States are, in order, 18-gauge needle cores,
transurethral resection of prostate (TURP) chips, radical prostatectomy specimens,
and fine needle aspirates.
A. Needle cores. Needle core biopsy sample handling and processing begins in the
room where the procedure is performed. The needle biopsy tissue should be
immediately placed into a container with fixative, which is usually 10% neutral
buffered formalin, although a few laboratories prefer Bouin's solution, Hol-
lande's solution, or IBF fixative. Bouin and Hollande's solutions are picric acid-
based fixatives that provide superior nuclear detail, but these strong oxidizing
agents can react violently with combustible materials and reducing agents. Fixa-
tion in formalin should be for at least 6 hours. The number of cores received per
container is highly variable, from 1 to >20. If the urologist and treating physi-
cian desire site-specific diagnosis, the core(s) should be placed in separate site-
designated containers. Inking of cores to indicate site, with placement of cores
marked with different colors into the same container, should not be performed
because fragmentation renders site assignment impossible. Gross examination
of prostate needle core tissue is not diagnostic, but is important for correlation
with amount of tissue seen in the histologic sections and so it is vital to record,
for each container, the size and number of tissue cores or fragments. It is recom-
mended that no more than two cores be submitted per cassette for processing
and embedding; some laboratories submit one core per cassette. Prostate cores
can be marked with ink, which facilitates identification during embedding and
the ability to see the cores in the paraffin blocks. Regardless, the cores should be
placed into a cassette after being put into a fine mesh envelope, wrapped in lens
paper, sandwiched between sponge pads, or double-embedded in agar-paraffin
wax. After processing, the cores should be embedded in the same plane, in the
same direction, with even spacing. From each paraffin block, three hematoxylin
and eosin (H&E)-stained slides should be prepared, with three to four serial
sections on each slide. Some laboratories cut interval, unstained sections on
coated slides in case special studies such as immunohistochemistry are needed.
Clinical requests for frozen section diagnosis of prostate needle cores are rare
and should be restricted to patients with clinical evidence of metastatic cancer
who are to undergo immediate treatment (usually orchiectomy) for pain relief.
*All e-figures are available online via the Solution Site Image Bank.
467
468 I SECTION VII: REPRODUCTIVE TRACT
inside the gland while preserving the inked periphery. Alternatively, after ink-
ing, margin sampling, and seminal vesicle amputation (see below), the whole
unfixed gland can be sectioned with a large sharp knife from apex to base at
4-mm intervals perpendicular to the prostatic urethra; areas suspicious for
carcinoma, as judged by palpation or visual inspection, may be sampled by
imprints, scrapes, core biopsy, or small wedge sections.
Fixation of the inked radical prostatectomy specimens (sectioned or
unsectioned) is accomplished by at least overnight (or 24 to 48 hour) room
temperature immersion in 10% neutral buffered formalin at 10 times the
volume of the specimen. For gross examination of unsectioned glands after
inking, the seminal vesicles are amputated (including the soft tissue and pro-
static tissue at the base of the seminal vesicles) and submitted separately as
right and left seminal vesicles. The prostate weight without the seminal vesi-
cles is recorded, and distal apical (urethral) and bladder neck margins are
taken if not already submitted separately by the surgeon. The distal apical
margin is evaluated by amputating the distal5 to 10 mm of the gland, dividing
it into the right and left sides, and submitting radial sections (as for a cervi-
cal cone biopsy). The bladder neck margin can be assessed by a thin 2-mm
shave margin or by conization; the latter is recommended. Ink on tumor cells
is indicative of a positive margin for cone sections and the peripheral margin,
whereas tumor anywhere in shave margin tissue indicates a positive margin.
Vasa deferentia stumps may be sampled using en face sections, but this is
not routine. The prostate gland is serially sectioned in a plane perpendicular
to the urethra at 3- to 5-mm intervals using a long knife. The cut surfaces
should be evaluated for gross evidence of BPH and carcinoma. Photographs
or digital images can be used to document location and gross appearance of
tissue in submitted cassettes. Diagrams (Fig. 29.1) or pictorial maps can also
be used to indicate location of sections and any gross abnormalities.
Both complete and partial embedding methods are acceptable (Mod
Pathol. 2011;24:6). Several protocols for partial submission have been pub-
lished (Scand] Urol Nephrol Suppl. 2005;216:34; Mod Pathol. 2011;24:6).
When there is grossly visible tumor, all lesions grossly suspicious for carci-
noma should be submitted, along with distal apical and bladder neck margins
and seminal vesicles. For cases with no grossly evident tum01; the posterior
aspect of each transverse slice is submitted, as well as a mid-anterior block
from each side, distal apical and bladder neck margins, and the seminal
vesicles. Sections should be submitted as quarters or halves of the prostate,
depending on gland size. Whole-amount sections are rarely made and do not
provide additional morphologic information. If no or minimal tumor is seen
in initial sections of a partially submitted gland, all remaining tissue should
be embedded (including any frozen tissue sent to a tissue bank). If still no
tumor is seen, basal cell and AMACR immunostains of atypical foci should
be performed, and deeper sections should be obtained from block(s) from
the region of the positive needle biopsy and areas of high-grade PIN. If can-
cer remains undetected, the tissue blocks should be flipped, and histologic
sections prepared from the new tissue faces (Mod Pathol. 2011;24:6).
E. Cystoprostatectomy. The prostate gland in radical cystoprostatectomies per-
formed for bladder cancer can be sampled by taking several sections of prostatic
urethra and surrounding prostate tissue, any gross lesions, one block from the
periphery of each side, and both seminal vesicles. The distal urethral shave mar-
gin is important in urothelial carcinoma cases.
Ill. DIAGNOSTIC FEATURES OF COMMON DISEASES OF THE PROSTATE
A. Inflammation and infection. Histopathologic identification of inflammatory cells
in the prostate is common, but histologic identification of specific infectious
agents is rare.
470 I SECTION VII: REPRODUCTIVE TRACT
An tenor l'linddor neck
Radical Prostatectomy case# _ _ _ _ __
Guide to Taking Sections Name _ _ _ _ __
Weight _ _ _ _ _ grams
Ink right BLACK, Left BLUE o r RED
Guide to block numbers:
Block 1 - Tumor in Methacarn Fixative
Right- odd numbers
Left - even numbers
Posterior
Anterior Anterior Anterior
Righ~
·
3
At>ex
Ovemead
view
'A 2,4
Left~
Posterior Posterior Posterior
Im
Ri:~no.
Bladder Neck
Ovemead
view
Cl' CilllO.
Figure 29.1 Diagram depicting guide to taking sections from a radical prostarectomy specimen.
(Modified from: True LD. Surgical pathology examination of the prostate gland. Practice survey
by American Society of Clinical Pathologists. Am] Clin Pathol. 1995;103:376.)
Figure 29.2 Architectural patterns of h.igh~grade PIN'. (Reproduced from: Humphrey PA. Prostate
Pathology. Chicago, ll.: ASCP Press; 2003. Used with permission.)
........
Alrolllllc
l'o<w.llollyporplde
l'oomy l.eiJ"""'"'
-~~
C.llolil O;'adenOII'oO
SV>ot~ .. ,_pll.,_a (Wimt. tunttlf)
~ Rhobdold tt.mor
i.)m~ Go/In ooll tllmm
Oll<h>lllll 101111 Ol>llldlo eel dill'~ Yo((~ b.lmor
(o.reb:taareoma. u1oon'lil!ltti:t e~momll) Seminoma
P-lnlrlllpllte/lllnooplak (PIN)
Duct.JI ad6nccadll0mt
CJI!rlonn
~
a..r.,.ll-rdn"""'
...
Elllbl'!llNI<O.-..,otre end - •
Poplllry
$old
~tllmMO
UIOIII...Ioormornl ---
.......,..-...-.
""'~-
_.,..... __
a,__,~z~mo~o
l'!lllhllll-lft
-~-
Adlf'IC*IUln'IICU.l a.rc!l"'a"'W
Squtii'IOUII ... """'""""
&.lcoillli/nfn
Cjoladoln....
_.
a..l ...l adenoma Mllpnl
a..1oo11oormorn•
-11101••-
Endoetfft• dffetel1'tllrl:bn wth!n t.dertoou(!!l'll:lma
Cln:lnoldl!.mor
.......,........
illriiJ1
L.elon'I')'OIIrooma
Smoll odl<:~rmorna Mabla-•
u_...
"-"'1om•
,.,.,.blaelomo Malllnont~broua hll1iooyl)mo
- ..
So!Wyftbnlu& tumor
rt.llllllo-lti- Hom.a ....~.
..........
SO"olnol tumot ~ uiiC<lrtoln malllnant IIOleltllol
LAiomyou-
,.,.,..
Lelontyon•
-~~-~~-
~lnan.l
..
Malo od1101<.61 tumor of p-bly Woll!lon
Rhobdomyou,...,...
Chondrou""'""'
An&Mo,_•
Mllpnt fl~u1 h~a
Malllnont l*llilentl""""' ~ ll!mot
Halllll*m•
---
oti&tl
Chondran.a
LAio~
Gflf'\ulllr eel t.rmor
HoiTII ....Piflcjbna
Soliuy~broua tum"
478 I SECTION VII: REPRODUCTIVE TRACT
bbr
PSA 94-100
(Jill-·
~ c:MtiiiA.. Vre:te'•l CMI'
G f - pulh't)
0
•••
PSAP 89-100 0
P$MA 92 0
NIQCS.I !15 I)
p- 100 6
p63 0-3 8l..Q6
llu..,bomoduln 0 69-&1
UR>jlllkin Ill 0 67
Hlatt-molo:IJior....,qt,t ~n• o-10' 6&-100
478 I SECTION VII: REPRODUCTIVE TRACT
along with serum PSA level and clinical or pathologic stage, in tables (Partin
tables) (http://urology.jhu.edu/Partin_tables/index.html) and nomograms (] Urol.
2001;165:1562; Cancer. 2009;115(Suppl):3107) to predict pathologic stage and
response to treatment and outcome.
Grading should be performed using the modified Gleason system (Fig. 29.3). All
adenocarcinomas of the prostate should be graded, except those that are posthor-
monal therapy or postradiotherapy (when radiation effect is evident). Grading is
based solely on architecture and does not incorporate cytologic atypia or mitotic
counting. At low magnification (40 to 100x), the most common pattern and the
second most common pattern are summed to yield a score (on a scale of 2 to 10).
Recent recommendations in application of the Gleason system include the follow-
ing (Am I Surg Pathol. 2005;29:1228; I Urol. 2010;183:433):
A. Do not (or rarely) assign a well-differentiated Gleason score 2 to 4 to carcinoma
in needle biopsy. This almost always represents undergrading.
B. When three grades are present, in a needle biopsy give most common grade and
worst grade. Thus, if 3 + 4 + 5 are present, the Gleason grade is 3 + 5 = score of
8. In a radical prostatectomy, give the most common and second most common
grades, but if there is a minor tertiary high-grade 4 or 5 pattern, this should be
noted in a comment.
C. If there is 95% high-grade pattern 4 or 5 and 5% or less of 2 or 3, ignore the
lower-grade component. Any high-grade pattern (4 or 5) should be incorporated
into a needle biopsy Gleason score; thus, 98% pattern 3 and 2% pattern 4 in a
needle biopsy is Gleason grade 3 + 4 =score of 7.
D. Variants of prostatic adenocarcinoma can be graded (see above).
E. Cribriform adenocarcinomas are high-grade pattern 4.
F. For needle biopsies, provide grade by clinically submitted container, even if
several cores are within the container. Alternatively, provide the Gleason grade
for the core with highest Gleason score, if different from the overall Gleason
score for cores from that container.
V. PATHOLOGIC STAGING. Staging applies only to adenocarcinomas of the prostate, and
not to sarcomas or prostatic carcinoma variants that are not adenocarcinomas.
The 2010 Tumor, Node, Metastasis (TNM) American Joint Committee on Cancer/
International Union Against Cancer (AJCGUICC) staging classification is given in
Table 29.4. Clinical staging should be distinguished from pathologic staging.
A. Needle biopsy. Pathologic staging is not performed. However, extraprostatic
spread should be diagnosed if carcinoma is seen in fat or in seminal vesicle
tissue.
B. TU RP chips and open prostatectomy. Pathologic staging is not done, but for inci-
dental carcinoma, the amount of carcinoma determined by light microscopic
inspection of tissue involved will place the patient into clinical stage Tla or Tlb.
C. Radical prostatectomy and pelvic lymphadenectomy are used for pathologic
staging (Table 29.4) (Arch Pathol Lab Med. 2009;133:1568).
1. pT2: Organ-confined prostatic carcinoma, subdivided into a, b, and c.
2. pT3: Extraprostatic extension (EPE) by carcinoma, diagnosed if carcinoma
extends into posterolateral periprostatic adipose tissue (e-Fig. 29.45), beyond
the outer boundary of normal prostatic glands at the anterior or apical
prostate, or microscopically into the bladder neck. Note that carcinoma in
skeletal muscle at apex does not always mean EPE. Site(s) and extent ofEPE
should be specified. EPE extent should be given as focal (only a few glands
outside the prostate) or nonfocal. Capsular invasion is not part of the staging
scheme. pT3b is seminal vesicle wall invasion (e-Fig. 29.46).
3. pT4: Gross bladder neck or rectal involvement by carcinoma.
4. pN: Number of involved and total number of examined lymph nodes should
be given.
5. pM: At time of radical prostatectomy, patients are clinical MO (cMO).
Cllojbr29•- I ... ,
1'1.1 llft($1 ~IWI.ttalll(TNIIO.._klllmnfdrottatfc
''' , ............. m
1X Pr11i111)'tumorCinl1<1t bo nltlrtlllid
ro
n
No -·"'of ptlmel)'tum'"
alnleol~lnii>IIO/ent111m«lltliher 11011><1>1& "'" >iliblo b:t l'nll!n1
n. 1\Jrnor lndllellllll ~~ ....,In :;5\\ oftlsou•-
nb
nc
T2
1\Jrnorlndlle!!lal~~ndln,ln >5'1'.a!lllauor-
T u " " " - byneeclle blo~sy C..,., bocau .. of
Tu""" coni!ned willIn p -
-tad
PSA)
• All e-figures are available online via the Solution Site Image Bank.
483
484 I SECTION VII: REPRODUCTIVE TRACT
,_, .....
Buo!QOj! aorct><MIIl
f'<fllofll1roec>llllelal • ...,~
-···-
Poeoldl-..
--..........
..........
MNI1Ct)1Sc ni'IUI
...,
·-""-
488 I SECTION VII: REPRODUCTIVE TRACT
Figure 30.1 Verruciform tumors ofthe penis. A: Verrucous carcinoma with regular papillae, broad
pushing base, and hyperkeratosis. B: Papillary carcinoma with irregular papillae and cores, and
ragged infiltration at base. C: Giant condyloma, with branching cores and koilocytosis. D: Warty
(condylomatous) carcinoma with irregular papillae and koilocytosis. (Modified from: Young RH,
Srigley JR, Amin MB, et al., eds. Tumors of the Prostate Gland, Seminal Vesicles, Male Urethra,
and Penis. Washington DC: Armed Forces Institute of Pathology; 2005:424.)
. ..........."'..._
1\Miblll-.....
Tho !Ulflk "m"lndl::o1os tho P""""" of mullplo pmwy to.morund lo 10001dod In
1>1181111- forOl!llnl>lo. ~T~CrnlNOI.IO.
,
fllq&O Til NO MO
Ta NO MO
Slqj:ll Tla NO MO
Slqj:lll Tlb NO MO
T2 NO MO
T3 NO MO
fllq& lila THI Nl MO
Slqtlllb Tl-3 N2 MO
SIQ>IV T4 Nf1N MO
NvT N3 MO
AIIYT AllYN Ml
F.,,. ~ SB, e,rd DR, Com...... OC:, .. 01,- AJCC Ciw8r511,111y-..: 7111 ed. N'"'"""'
~. sro~ .....,
21)10. tJood- pelll'lllolcr\.
T=pT
I I
""'
T2: Tumor invading corpus spongiosum or cavemosum
T=pT
pT4
tumor: tumor size, histologic type, histologic grade (Gl, G2, or G3), origin, depth
of invasion (mm), anatomic level of invasion, structures involved, vascular and
perineural invasion (if present), and status of the margins of resection. For regional
lymph nodes, the report should include the number identified and their location,
number involved by tumor, size of metastatic deposit (if present), and extracapsular
extension (if present). Additional pathologic findings that can be noted (if present)
are penile intraepithelial neoplasia and therapy-related changes.
SUGGESTED READING
Epstein JI, Cubilla AL, Humphrey PA. Tumors of the prostate gland, seminal vesicles, penis, and
scrotum. In: Atlas of Tumor Pathology. 4th Series. Washington, DC: American Registry of
Pathology; 2011.
The Ovary
Meredith E. Pittman, John D. Pfeifer,
and Phyllis C. Huettner
I. NORMAL GROSS AND MICROSCOPIC ANATOMY. The ovaries rest on either side of the
uterus and are anchored by the broad ligament. Age and reproductive status greatly
impact size and weight, which range from 3 to 5 em and 5 to 8 g, respectively.
The outer surface of the ovary is white-tan and smooth during early reproductive
years but becomes more bosselated with age due to repeated rupture of ovarian
follicles. The cut surface is vaguely organized into three ill-defined zones: an outer
cortex, underlying medulla, and inner hilum. The cortex and medulla contain cystic
follicles, yellow or orange corpora lutea, and gritty, white corpora albicantia.
Histologic sections of the ovary show a simple cuboidal surface epithelial layer
derived from mesothelium. Within the ovarian cortex and medulla, follicular struc-
tures composed of an inner layer of granulosa cells and an outer layer of theca cells
(e-Fig. 31.1* and e-Fig. 31.2), in varying phases of development, are surrounded
by a stroma comprised closely packed S-shaped spindled cells and collagen (e-Fig.
31.3 ). Centrally hemorrhagic corpora lutea composed of granulosa cells with abun-
dant eosinophilic cytoplasm and smaller theca cells (e-Fig. 31.4 and e-Fig. 31.5),
as well as white acellular corpora albicantia (e-Fig. 31.6), may also be seen. The
hilus, where the ovary connects to the broad ligament, is composed of abundant
blood vessels, nerves, and interspersed eosinophilic hilar cells that are histologically
similar to the Leydig cells of the testis (e-Fig. 31.7). Rete ovarii, the developmental
analogue of the rete testis that are composed of slit-like spaces lined by nonciliated
cuboidal epithelium, are also present in the hilum (e-Fig. 31.8).
II. GROSS EXAMINATION AND TISSUE SAMPLING. The most common ovarian specimens
encountered in surgical pathology are from oophorectomy (with or without hys-
terectomy) or cystectomy procedures. The weight and gross measurements of all
three dimensions are recorded. The capsule is inspected for areas of rupture, adhe-
sions, tumor involvement, or other abnormalities. In most cases, the ovary is then
bivalved along the long axis and any lesions on the cut surface are noted. Solid, cys-
tic, or papillary lesions are thoroughly sampled (one section per em). If no lesions
are identified, one section for every 2 em is adequate. If cysts are present, the color
and consistency of the cyst fluid are noted, and any areas of nodularity or papillary
excrescences are sampled. Prophylactic oophorectomy specimens, performed for a
personal history of cancer or family history of a hereditary cancer syndrome, are
cut perpendicular to the long axis and entirely submitted.
The surgical management of malignant primary ovarian tumors typically
includes a staging procedure, and so an ovary excised for a primary malignancy
will usually be accompanied by multiple abdominal-peritoneal biopsies, an omen-
tectomy specimen, and regional lymph nodes. The small peritoneal biopsies are
submitted entirely. The omentum sample must be serially sectioned. If grossly vis-
ible tumor is present, it should be measured and only one section needs to be
submitted; when no tumor is identified, from 5 to 10 sections are submitted. All
identified lymph nodes are submitted from the lymph node dissections.
Ill. DIAGNOSTIC FEATURES OF COMMON BENIGN DESEASES OF THE OVARY
A. Inflammatory Diseases
1. Infection of the ovary is almost always accompanied by infection of the
fallopian tube (e-Fig. 31.9), systemic infection, or the development of a
*All e-figures are available online via the Solution Site Image Bank.
493
494 I SECTION VII: REPRODUCTIVE TRACT
..,...,....,_,_
'' 1,1,! I J!!'I
SDroul tumofl
MIJip\lnt
Modi led WHO lllllolotle CIMtltlcaUOft., TilllltcUI lilt 0\wJ
hi.......,..,.,..
Bonl0111.,. tumor
Ben~!~>
O,.!a-don"""''"' "· -.cnbtoml, """"'-trurna
Mudn.:w: t.rmora
MIJip\lnt
hi.......,..,.,..
Boni0111.,.11Jmor
Ben~!~>
0,.-••· -.cnbtoml, """"'-trurna
Mudncuqo!Sc tumor will ~rna portloMI
Eilcl-lllrn""'lndudl111 \llrlonls\llllloqullnOUSdlllormtlltion
MIJip\lnt
hi.......,..,.,..
MIJip\lnt......., MDiellln turn" (o81d>o••,..,mal
Eilc!Oini01IIolil ldiOii\llaroorne (low~
Undlllronllltad OVIJ1an ....,rna
Bonl0111 .. lumot
Bonlin
~. odeho11bluno. 0'1C0d~trumo
Clellr ooi11Jmoro
MIJip\lnt
hlonoc.t-trum•
Bonl0111 .. lumot
l!onlln
~. odeho11bluno. 0'1C0d~trumo
nanolllonolceil tum""
MIJip\lnt
T11nsnlonll col ,...,.rna Cn<>n·BIIIIrw typo)
MIJip\lnt Brem.-l!.mot
Bonloo1lne
l!onlln
Brenherl!l....-
ti:qu&.n'l)ua 011111 tuMor&
S;qu.-noua 011111 Ollf'C:ftoma
MUd <ljl)lhllll tumm (lpecl) corn-nil)
MIJip\lnt
Bonloo1lne
Benlln
Ulldl'l&1111111r1ed and uhCIMollled11J,....
Undl'l&llnlllted cormorn1
...,......,,..,.,.,.. nrt alti-"'*lliad
llrcrMIIHifiRII . . .
Gnnu-lcoll tuh'l01"8
Glonulello collllmor 11"01/P
hlutgno"'""' Ml "''""
liMnII& gnonuloo4 oelll!Jird"
(~
~a1• ne.ovay I ... ,
I' I.I II f:£1'1 Modi liM! WHO llllloiOI!e CIIAltlcat!Oft .t'l'll- of tilt Ol'lr.J
' - (CIIIIIIuilt
n-t\a-1\b~o III'OIIP
n-n.. notOI!IoM!oo oDOdllod
~
lu111-
FII...,.,.
Cell"'' flllroll\l
Fbt'OiiiiM'OOMI
~bJmor~ mln~k'I:Xcon:i•leml!lf'ltt
Scloloostw-ol tumor
~rtl,t-tumllf
U n - (llblllltiOCilflll)
- ! c o l i Ill....
s..toi,IAjdiJc<llllllmor III'OIIP
Wolld!tfnnt1111d
Ollramw!lo1ad!ll'n-
V.!WrtMII IIA>!Oiocouo...,.,..,,...,_to !Gilo<lfJ1YPOl
-
~dl!'ell1111otod lelloo""'>ldl
Vlrlilrt Mil l>alo,.qow,oloo,..,b ("""'!ly typo)
.......,.._
MIIIpnt
Moo-••
C)Oilc
f<lltlonm ''"'''"" (homunwll;)
tn1tma and solftltlo.lype Ill moll
Thyroid Ill"""....p
SlruiNI <IYIJ11
--lid will dotmold c:ysb
811111!;n
Mal...nt (OI>OCifY~pe)
.... I SECTION VII· REPRODUCTIVE TRACl
I' 1,1,! I J!!'I ....... WHO llllloiOI!e CIMtltlcat!Oft., 'l'iiiMa ., lilt OI'IIJ
' _ (CNatN
01-....p
Notnectcdlm'lill tumw pup
o.,_•.,..p
""""'.........._
~IJOUD
M&llnoeylfe....,.
San:un.t '""'p
(lpoo:l!y IJlMI)
......___._
s.btctoua tumor woup
Pilll~ 11/mor.,..P
RtOial•,._ tumor II'OIIP
--
Olhe!l
_........
Myoo>mt
Soft_tu.,.,. nd specllt to 1116 aitl'/
Ollie!>
LiiiDorno <111>1..,0/ICY
SOUnol~
SOUnol ilypelplula
Flbnmltmls
- - t l l n o d ....
Ollie!>
...............1111 .....
Ma..,_.ftt limPharno '-'i'JIYPOl
U.ulillrolo <oi>OCti 1l'liOl
Plumoeylorna
till.,. .....
Chapter 31 • The Ovary I 499
and shows fusion of the tips of papillae with the formation of cribri-
form structures (e-Fig. 31.25). Both of these patterns are associated with
a poorer prognosis due to their association with invasive implants. Some
groups classify tumors with these patterns as a form of low-grade serous
carcinoma and use the term micropapillary or micro-cribriform serous
carcmoma.
c. Serous borderline tumors with microinvasion. Occasional serous borderline
tumors exhibit microinvasion where individual eosinophilic cells or small
nests of cells, often surrounded by a clefted space, invade the stroma (e-
Fig. 31.26). These microinvasive areas should involve < 3 mm in greatest
linear extent or <10 mm2 in area. The prognostic significance of microin-
vasion is not completely understood, but it may be a risk factor for disease
progression (Am] Surg Pathol. 2006;30:1209).
d. Serous adenocarcinoma most commonly presents as bilateral masses with
widespread peritoneal metastases. Grossly, the tumors are large and fri-
able with multiloculated cysts and polypoid growths. Microscopically, the
tumors may show a wide variety of architectural patterns including papil-
lary, solid, and nested with slit-like spaces. Nuclear atypia is variable but
often marked. By definition, serous carcinomas demonstrate destructive
stromal invasion (e-Fig. 31.27). Psammoma bodies may be present.
Serous carcinomas should be graded into high grade and low grade.
Low-grade tumors (< 12 mitoses/1 0 high power fields and low-grade cyto-
logic atypia) (e-Fig. 31.28) are frequently resistant to chemotherapy and
are more appropriately treated surgically.
A rare form of serous adenocarcinoma known as psammocarcinoma
is characterized by invasive stromal growth, abundant psammoma bodies
in at least 75% of the papillae, and no areas of solid growth > 15 cells
across (Gynecol Pathol. 1990;9:110). This tumor generally has a good
prognosis.
2. Mucinous tumors are the second most common type of surface epithelial
tumor. The neoplastic epithelium resembles intestinal (goblet cells with intra-
cytoplasmic mucin), gastric (foveolar), or endocervical (apical mucin with
no cilia) epithelium. Eighty percent of these tumors are benign, 10% are
borderline, and 10% are malignant. Because mucinous neoplasms are quite
heterogeneous and a malignant component may be very focal, it is imperative
that all mucinous neoplasms be well sampled (one to two sections per em).
a. Mucinous cystadenomas are usually unilateral, multiloculated, and large
(up to 50 em). They have a smooth external surface and are filled with thick
viscous secretions. Microscopically, the cysts are lined by a single layer of
tall columnar cells with bland basal nuclei, most often of intestinal type
(e-Fig. 31.29). If there is a prominent stromal component, the lesion is
termed a mucinous cystadenofibroma.
b. Borderline mucinous tumors (mucinous tumors of low malignant potential)
also present as unilateral, multiloculated masses. They are most common
in perimenopausal women. Grossly, these tumors have thick cyst walls
sometimes with papillary excrescences. Microscopically, mucinous bor-
derline tumors have a stratified, tufted intestinal-type epithelial lining with
mild to moderate nuclear atypia (e-Fig. 31.30). By definition, borderline
mucinous tumors do not demonstrate destructive stromal invasion.
Borderline tumors with endocervical-type epithelium (15% of muci-
nous borderline tumors) have the hierarchical branching pattern of
serous borderline tumors but the papilla are lined by mucinous epithe-
lium; they are often associated with acute inflammation and frequently
with endometriosis and bilaterality (e-Fig. 31.31). Some borderline
tumors show a mixture of endocervical-type mucinous epithelium, serous
Chapter 31 • The Ovary I 50 1
epithelium, and even endometrioid epithelium; these tumors also have the
hierarchical branching architecture of serous borderline tumor and are
also associated with endometriosis and bilaterality.
c. Mucinous borderline tumors with intraepithelial carcinoma resemble border-
line tumors but contain focal areas that demonstrate increased cytologic
atypia with marked pleomorphism and prominent nucleoli (e-Fig. 31.32).
No destructive stromal invasion is present, and the intraepithelial carci-
noma does not appear to portend a worse prognosis (Ann Surg Oncol.
2010;18:40).
d. Mucinous borderline tumors with microinvasion. These are mucinous bor-
derline tumors with small foci of invasion either as small nests or as indi-
vidual cells, often with more eosinophilic cytoplasm. These foci should not
exceed 3 mm in greatest linear extent or 10 mm2 in area (e-Fig. 31.33).
e. Mucinous adenocarcinoma also presents in perimenopausal women as uni-
lateral, multiloculated cystic masses. Frank stromal invasion is present
microscopically (e-Fig. 31.34), but mucinous carcinomas frequently
demonstrate areas of benign and borderline mucinous epithelium as well.
Two patterns of invasion are recognized. The expansile pattern shows
crowded glands, little stroma, and, sometimes a cribriform architecture.
The destructive pattern shows single glands or individual cells invading
>3 mm in two linear dimensions or >10 mm2 in area.
Approximately 5% of women with a mucinous ovarian tumor will
present with pseudomyxoma peritonei, a condition in which pools of
mucin, with or without associated neoplastic epithelium, fill the peri-
toneal cavity. Although controversial in the past, there is now general
consensus that most mucinous tumors in the ovary associated with pseu-
domyxoma peritonei are metastatic in origin. The appendix is the most
common primary site (Am ] Surg Pathol. 1995;19:1390). Microscop-
ically, the ovary shows pools of mucin dissecting through the stroma
(pseudomyxoma ovarii) with or without associated mucinous epithelium
(e-Fig. 31.35).
When trying to distinguish a primary from a metastatic ovarian muci-
nous adenocarcinoma, both histologic and immunohistochemical features
are helpful. Primary tumors exhibit benign, borderline, and malignant
epithelium whereas metastatic tumors are more uniformly malignant.
Features seen more commonly in metastatic disease include bilateral-
ity, concomitant extraovarian disease, involvement of the external sur-
face of the ovary, pseudomyxoma ovarii, and extensive lymphovascu-
lar space invasion. Metastatic adenocarcinoma from the colorectum will
show abundant luminal karyorrhectic debris (dirty necrosis), a garland
pattern of glands lining cystic spaces, an abrupt transition between viable
and necrotic epithelium, and immunopositivity for C:I<20 and CEA but
immunonegativity for CK7 and CA125 (e-Fig. 31.36).
3. Endometrioid tumors. The majority of endometrioid tumors of the ovary are
carcinomas. From 10% to 20% are associated with endometriosis, and about
15% have a concomitant endometrioid tumor of the endometrium. Squa-
mous differentiation may be seen in all types of endometrioid tumors.
a. Benign endometrioid tumors are adenofibromas/cystadenofibromas, with
organized endometrial glands arranged in a fibrous stroma. These are quite
rare and can be distinguished from endometriosis by a lack of associated
endometrial-type stroma and hemosiderin-laden macrophages.
b. Borderline endometrioid tumors. Endometrioid tumors that show cytologic
atypia and areas of confluent epithelial proliferation without stromal sup-
port up to 5 mm in maximal dimension are borderline endometrioid
tumors, although there is some controversy as to diagnostic criteria and
IDf I SECTION VII· REPRODUCTIVE TRACl
mitotic rate than adult granulosa cell tumors. The nuclei do not have
grooves (e-Fig. 31.53).
JGCTs are usually confined to the ovary and high stage is a poor
prognostic factor. Unlike their adult counterparts, patients with JGCT
who recur usually do so within 2 years of diagnosis. Cytologic atypia is
not a poor prognostic factor in JGCTs.
2. Fibroma-thecoma
a. Fibromas represent the most common of the sex cord-stromal tumors.
They typically occur in perimenopausal women and are not hormonally
active. Grossly, fibromas are unilateral, solid, and lobulated with a firm,
white-gray cut surface. Cystic degeneration sometimes occurs. Histolog-
ically, they are characterized by interlacing bundles and storiform areas
of spindle cells that show no atypia and few mitoses (e-Fig. 31.54). The
tumor cells stain diffusely positive for vimentin and are usually negative
for inhibin.
Fibromas may be associated with two syndromes. Meig syndrome
(fibroma, ascites, and pleural effusion) and Gorlin syndrome (basal cell
nevus syndrome). In Gorlin syndrome, fibromas occur in younger women
or even children, are often multiple or bilateral, and are calcified.
b. Thecomas are unilateral solid tumors found most often in postmenopausal
women. Hyperestrogenic symptoms are present in 50% to 80% of cases.
Grossly, these tumors have a lobulated, yellow-tan cut surface. Histo-
logically, thecomas comprise round to oval, lipid-laden theca cells in a
fibromatous stroma with little atypia or mitotic activity (e-Fig. 31.55).
Immunohistochemically, the tumor is positive for inhibin. Oil Red 0 fat
stains (which require fresh tissue) highlight the intracellular lipid. When
clusters of lutein cells (eosinophilic cells with large round nuclei) are
present, the tumor is termed a luteinized thecoma (e-Fig. 31.56); this vari-
ant is more common in younger women.
3. Sertoli and Sertoli-Leydig Cell Tumors
a. Sertoli cell tumors are rare, low-grade, nonfunctioning tumors that occur in
women of child-bearing age. Grossly, they are yellow-tan, solid, lobulated
tumors. Microscopically, they are composed of closely packed tubules
separated by fibrous stroma. The tubules are lined by cuboidal to colum-
nar cells with abundant pale eosinophilic cytoplasm with little atypia or
mitotic activity.
b. Sertoli-Leydig cell tumors (SLCT) are rare, unilateral neoplasms that occur
in young women and, in 30% of cases, secrete androgenic hormones.
Grossly, these tumors average 10 em in diameter, are yellow-orange to
red-brown, and frequently have a nodular appearance with a central scar
(e-Fig. 31.57). Well-differentiated SLCTs contain tubules (similar to those
seen in Sertoli cell tumors) and interspersed clusters of Leydig cells that
have abundant eosinophilic cytoplasm (e-Fig. 31.58). SLCT of intermedi-
ate differentiation contain solid cords of Sertoli cells (e-Fig. 31.59), while
Leydig cells may be more difficult to find but are typically located at the
periphery of cellular nodules. The Leydig cells often show lipidization
with foamy rather than eosinophilic cytoplasm (e-Fig. 31.60). Poorly dif-
ferentiated SLCT show densely packed atypical spindled cells resembling
a sarcoma (e-Fig. 31.61).
Two morphologic variants of SLCT occur, usually in association with
tumors of intermediate differentiation or poorly differentiated tumors.
About 20% of SLCf harbor heterologous elements that take the form
of intestinal type mucinous glands (e-Fig. 31.62) or carcinoid tumor,
(which have a good prognosis) or rhabdomyoblastic or cartilaginous dif-
ferentiation (which have a poor prognosis). About 15% of SLCT contain
Chapter 31 • The Ovary I 50 5
any age. Grossly, they are usually cystic with a single solid nodule that may
contain fat, teeth, bone, and many other tissue types. The cysts usually con-
tain hair, soft yellow sebaceous debris. Microscopically, mature tissue from
all three germ layers (ectoderm-skin or central nervous system elements;
mesoderm-smooth muscle, teeth, bone; endoderm-respiratory epithe-
lium, GI epithelium, thyroid) may be present (e-Fig. 31.68). The term
dermoid cyst is commonly used to refer to mature cystic teratomas lined
by squamous epithelium that containing skin appendages (e-Fig. 31.69).
Mature cystic teratomas should be thoroughly sampled (one section per
em) in order to exclude an immature component, and to exclude malignant
transformation of one of the mature components (a rare occurrence).
b. Immature teratomas are rapidly growing malignant tumors that occur in
children and young adults. They are unilateral and typically have solid
and cystic components. The solid areas are generally more extensive than
in a mature teratoma. Microscopically, they contain immature or primi-
tive tissue (derived from any or all three germ cell layers) that is usually
mixed with areas of mature tissue. The most common immature element is
neuroectodermal and consists of rosettes, masses, or tubules of primitive
neural cells (e-Fig. 31.70).
Immature teratomas are graded based on the relative amount of imma-
ture tissue present (lnt] Gynecol Pathol. 1994; 13:283 ). Tumors with more
than one low-power field of immature neuroepithelium on any given slide
are considered high grade and require adjuvant chemotherapy.
c. Monodermal teratomas are teratomas in which all the tissue is derived from
one germ cell layer. Struma ovarii is the most common monodermal ter-
atoma and consists of mature thyroid tissue, including follicles and colloid
(e-Fig. 31.71). Secondary changes such as hyperplasia, adenoma, and even
carcinoma may be seen.
2. Carcinoid tumors of the ovary usually arise in a mature cystic teratoma. They
most commonly have an insular or trabecular pattern identical to that seen
in the gastrointestinal tract. The cells are small and uniform with round
nuclei with stippled chromatin. Goblet cell carcinoids are very uncommon.
Metastasis from a primary carcinoid tumor outside the ovary must always be
excluded; metastatic carcinoid tumors are more likely to be bilateral, larger,
unassociated with a teratoma, and more commonly associated with carcinoid
syndrome.
3. Dysgerminoma is the most common malignant germ cell tumor. It occurs as
pure dysgerminoma or as a component of mixed germ cell tumor. Dysger-
minoma develops most commonly in adolescents and young women and is
frequent in patients with ovarian dysgenesis. Patients with dysgerminomas
frequently have elevated LDH levels and occasionally mildly elevated hCG
levels. Pure dysgerminomas have an excellent prognosis when treated by
current therapeutic regimens.
Grossly, dysgerminomas are large and solid with a smooth external sur-
face and a lobulated gray-tan cut surface. The tumor should be thoroughly
sampled (one section per em) for microscopic examination to exclude other
germ cell tumor types, and special attention should be directed to hemor-
rhagic and cystic areas.
Dysgerminomas are analogous to testicular seminomas and have an iden-
tical histologic appearance. They are composed of nests and sheets of uni-
form large round cells with abundant clear cytoplasm, large nuclei, and
prominent nucleoli. Tumor cells are separated by a lymphocyte-rich fibrous
stroma (e-Fig. 31.72). Often a histiocytic or granulomatous infiltrate is
present, and multinucleated syncytiotrophoblastic cells may also be identi-
fied (sometimes the former component can obscure the dysgerminoma cells).
Chapter 31 • The Ovary I 50 7
..., ,,.,..._{I,
Nl(
dlmeMioll n'of I'IIIISonol tJrnl>h nod&.-.....
Mllln•n•••••
MO ~ dll1ant m-olo
Ml PI llldart m-la <-b!M ~~ .,..,..,...,
-~~ nl ....,...,_ .,.,.
5lqo I T1 NO MO
SQIIA Tlo NO MO
SQIIB Tlb NO MO
Slqa IC Tie NO MO
Slqa II T2 NO MO
Slqo IIA T2.n NO MO
51qr:!IIB m NO MO
5lqo IIC T2r: NO MO
SQIIII l3 NO MO
SQIIIIA '1\lo NO MO
Slqa Ill 8 l3b NO MO
Slqo IIIC TaciAny T NOINI MO
5lqo PI AnyT A.nJN Ml
510 I SECTION VII: REPRODUCTIVE TRACT
I. NORMAL ANATOMY. The fallopian tubes are formed from the Miillerian
(paramesonephric duct) system and lie within the broad ligament between the ovary
and the uterus. They conduct eggs from the surface of the ovary to the uterine cavity
and are the usual site of fertilization. Each fallopian tube is shaped like an elon-
gated funnel and is divided into four parts from lateral to medial: infundibulum,
ampulla, isthmus, interstitium; the infundibulum contains the finger-like fimbriae
distally.
The fallopian tube mucosa is branched and folded into plicae. The mucosa is
lined by a nonstratified epithelium composed of three cell types: ciliated, secretory,
and intercalated cells. The most common are the ciliated cells, followed by secretory
cells, which together comprise over 90% of the cell population. The intercalated
cells are seen as elongated nuclei sporadically present between the ciliated cells. The
wall of the fallopian tube contains smooth muscle to aid in moving the fertilized
egg into the uterus. The serosa contains abundant blood vessels and is continuous
with the broad ligament.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION. Fal-
lopian tubes are usually received as a portion of a total abdominal hysterectomy-
bilateral salpingo-oophorectomy specimen. Short cross sections of fallopian tube
are received after tubal ligation procedures. Ectopic pregnancy specimens also usu-
ally contain a portion of fallopian tube. Rarely, specimens are received for primary
fallopian tube tumors.
At the grossing station, the length and diameter of the fallopian tube should be
documented, as well as the presence or absence of a fimbriated end, or evidence
of prior ligation. The serosal surface should also be assessed for the presence of
adhesions, cysts, exudates, rupture, or metastatic tumor. The tube is then serially
sectioned.
A. Benign specimens. Three sections are submitted from a normal fallopian tube,
specifically from the fimbriated end, ampulla, and isthmus end. Additional sec-
tions of any gross lesions are also submitted. Complete cross sections must be
identified from a tubal ligation specimen.
When examining an ectopic pregnancy specimen, an embryo and/or pla-
cental villi will often be grossly evident. Hemorrhagic areas, including blood
dot, along with obvious villous or embryonic tissue, should be submitted for
histologic examination.
B. Neoplastic specimens. Primary tubal carcinoma specimens will show a dilated
lumen filled with a papillary or solid tumor. An ovarian tumor secondarily
involving the fallopian tube is more common than a primary fallopian tube
tumm; and careful sectioning can help distinguish the two. Grossly, papillary and
solid areas should be sampled thoroughly (at least one section per centimeter of
tumor), along with uninvolved areas. If possible, a section showing the tumor's
relationship to the ovary should be submitted.
C. Prophylactic excision specimens. Fallopian tube specimens received as part of
a prophylactic hysterectomy-oophorectomy from patients with hereditary can-
cer syndromes (e.g., BRCA1 syndrome) should be, together with the ovaries,
entirely submitted for histologic examination. For the fallopian tube, the goal
is to ensure sectioning and extensive examination of the fimbria (so-called SEE-
FIM protocol), since the majority of early serous tumors occur in this area.
51 1
512 I SECTION VII: REPRODUCTIVE TRACT
Figure 32.1 Approach to sectioning fallopian tube specimens received as part of a prophylactic
hysterectomy-oophorectomy from patients with a hereditary cancer syndrome. Each tube and
ovary should be entirely submitted for histologic examination; note that the fimbriated end of
the tube should be transected, opened longitudinally, serially sectioned longitudinally, and then
entirely submitted for microscopic examination.
The protocol (Am] Surg Pathol. 2006;30:230) specifies that the entire tube is
fixed for at least 4 hours to minimize loss of epithelium during manipulation,
the distal2 em of the fimbriated end is transected, the fimbria! mucosa is secw
tioned longitudinally into four pieces, the remainder of the tube is sectioned
transversely every 2 to 3 mm, and that all the sections are submitted in toto
(Fig. 32.1).
Ill. DIAGNOSTIC FEATURES OF COMMON DISEASES
A. lnftammabJry and nonneoplastic lesions of tha fallopian tuba
1. Cystic lesions. Embryologic remnants may be found in the fallopian tube wall,
usually as an incidental finding (e-Fig. 32.1).• Paratubal cysts of Miillerian
origin (ciliated lining) or Wolffian origin (stratified transitional lining) may
be encountered; if the lining is atrophic secondary to compression, the two
can be difficult to differentiate microscopically. Pedunculated paratubal cysts
with a Miillerian epithelium present along the fimbriae are termed hydatid
of Morgagni.
2. lnftammatory lesions
a. Acute salpina;itis usually presents in youngw to middlewaged women, is
generally an ascending infection initiated by Chlamydia or Neisseria
*All ewfigures are available online via the Solution Site Image Bank.
Chapter 32 • Fallopian Tube I 5 13
-·-~-
Mudnow:.tdlnOQIJ'dnorna
EndMMI'llold ader.or:adl iOmt
Ckwlr e.a a.diii'IOOI!ft:!l"'l:::''''::l
'llonl~lonolool ..-""'
&~uomoua ceil..-1!111
Ulldl'l&""'lloled
Ollie!>
..,....,.m•
8ordoo1lnetllmo< (of botrn~ll!lllnt ,.anti~!)
Sotau1 bonS1ufnotu,...
Mudn~Ubo-tllmor
<liMIS
o.~ • .,..., (Bj>edfy'YP')
klllntum.,..
~CeDIICf1~
Adiii'IOI!brorna
Ojtt.IOono!IIM'oml
Enillmlllrlolil ~
Paplloml (>podfy typo)
-ploll!o P<!llloly1>Jmor
<liMIS
1\nnor-lko e!>tilellalloolono
1\Jbolel>tiloilal ~
Sll~lsth,__
Enll•lplnpu
.............n•ISa.ti-
MIJip\lnt l1'i>IDd M~lariBn tumllf (1:1-...r.t}
Ad_,..,mt
Mdllll•-
~"""-'-
~lomjomt
...............
<liMIll
-ell-1ft
Adonomidi:IU tumor
Tetnrno
MI1>Jre
mmtt~~re
<liMIll
'fte!NII lie • .._
Chllflo<a~a
Pilcortal 111t lropllo- tum ..
.,.•.
HyilltSitt'orm mala
Pltoilrotll 111t nodule
<liMIS
""""'rt I.--~~~~·
lAU-
lymph.....
a.: a,..._
TABL£12.
I)
I
lA
18
IC
T2 II
n. lUI.
T2b liB
1'2< IIC
Ill
lilA
IIIB
lacanclloi'NI IIIC
Ml
Dtllwrtrrttt+
'Ill NO MO
na NO MO
nb NO MO
nc NO MO
nc NO MO
T2b NO MO
T2c NO MO
'l3o NO MO
l3b NO MO
'l3e NO MO
M:tT Nl MO
Slqo IV M:tT MIN loll
'"""' £qpl SB,I!ynl 00,
NV,~2010.1M!-
eo...,.,.
CC, .. al, a:I!I.IIJCC Cilncof 5 ¥ Y - 7th ... -
,....,-.
Valle,
516 I SECTION VII: REPRODUCTIVE TRACT
I. NORMAL ANATOMY. The uterus is a pear-shaped hollow organ with a normal weight
of between 40 and 80 gin adults. It is divided into the corpus, the lower uterine
segment, and the cervix. The uterine cavity is triangular, measuring on average
6 em in length. It is composed of the inner endometrial lining and the myometrium
or muscular wall, with a serosal covering which extends to the peritoneal reflec-
tion. The peritoneal reflection is shorter anteriorly than posteriorly and so can be
used for orienting hysterectomy specimens.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION
A. Endometrial biopsy and curettage specimens. The most common endometrial
tissue samplings examined in surgical pathology are endometrial biopsy and
curettage specimens, obtained from cervical dilation and curettage procedures.
Endometrial biopsy samples are obtained from a relatively limited office sam-
pling procedure in which no cervical dilation is required. The dimension (size
range of the largest tissue fragments, or the dimensions of the tissue in aggre-
gate) and/or volume of the specimen should be documented. The entire speci-
men should be submitted for microscopic examination and three H&E stained
levels prepared for microscopic examination.
B. Products of conception specimens are usually obtained by curettage (although
the tissue is often spontaneously passed). The dimension (size range of the
largest tissue fragments, or the dimensions of the tissue in aggregate) and/or
volume of the specimen should be documented. At least three cassettes should
be submitted, focused on any villous tissue that is grossly present, to optimize
microscopic identification chorionic villi both for confirmation of the presence
of an intrauterine pregnancy and to rule out a molar gestation. If the initial
three blocks do not contain villi, the remainder of the specimen should be
submitted; if villi are still not identified, the possibility of an ectopic pregnancy
exists, a result that should be immediately communicated to the clinician.
C. Hysterectomy specimens. The type of hysterectomy (abdominal or vaginal, with
or without salpingo-oophorectomy) should be determined and the size, weight,
and shape of the uterus recorded (the processing of radical hysterectomy speci-
mens, which differs substantially, is discussed in Chap. 34). The uterine serosa
should be carefully examined for any abnormalities, which should be sampled.
The uterus is next bivalved in the coronal plane to show the endometrial cav-
ity and endocervical canal, which are examined and measured. The maximum
thickness of the endometrium and myometrium should also be noted. Both
halves of the uterus are then serially sectioned parallel to the long axis of the
uterus.
1. For specimens excised for benign disease, sections of the anterior
cervix, posterior cervix, anterior endomyometrium, and posterior endomy-
ometrium are submitted. Additional sections of any identified lesions must
also be submitted.
2. For specimens excised for malignancies, contiguous sections of both ante-
rior and posterior endomyometrium, lower uterine segment, and cervix
should be submitted to assess for tumor involvement of the lower uter-
ine segment and cervix. In addition, at least one full thickness section of
the uterine wall containing tumor from both anterior and posterior uterus
(including the serosa from the deepest area of myometrial invasion by the
517
518 I SECTION VII: REPRODUCTIVE TRACT
*All e-figures are available online via the Solution Site Image Bank.
Chapter 33 • Uterus (Corpus) I 5 19
that some of these lesions harbor PTEN tumor suppressor gene inactivation,
mutations of k-ras. and microsatellite instability. EIN is considered to be a
monoclonal proliferation composed of cells in early stages of carcinogenesis.
Diagnostic criteria for EIN (http://www.endometrium.org) are based on
size of the lesion, nuclear cytology, and glandular architecture (including glan-
dular crowding with an increased glands to stroma ratio, cytologic differences
between the neoplastic glands and the adjacent normal glands, and an area of
glandular crowding that is > 1 mm in greatest dimension). EIN can have squa-
mous, mucinous, or clear cell differentiation. The treatment of these lesions is
similar to the clinical management of atypical endometrial hyperplasia.
C. Endometrial intraepithelial carcinoma {EIC) is thought to represent the precursor
lesion to serous carcinoma. Microscopically, EIC is composed of glands lined
by cells with the same cytologic abnormalities as serous carcinoma, but without
evidence of myometrial stromal invasion (e-Fig. 33.32). The development of
EIC is independent of prior unopposed estrogenic stimulation and typically
arises in a setting of atrophic endometrium.
VI. EPITHELIAL MALIGNANCIES. Epithelial malignancies are the most common gyne-
cological malignancy in women in developed countries. Endometrial cancer can
be divided into two broad categories which have differences in their clinical and
pathologic features, as well as their underlying genetic abnormalities.
A. Type I tumors consist of endometrioid adenocarcinoma and its variants. They
account for over 80% of endometrial tumors and usually develop in post-
menopausal women in their fifth and sixth decades in the background of long-
term estrogen stimulation. Type I tumors are strongly associated with diabetes
and obesity and have a relatively good prognosis. The endometrial glands and
stroma in Type I tumors are strongly positive for estrogen and progesterone
receptors. In addition, the stromal cells show diffuse strong immunopositivity
for CD10 (CALLA) antigen. Genetically, they show microsatellite instability,
and mutations in the PTEN tumor suppressor gene, k-ras, and CTNNB 1. but
assessment for these genetic abnormalities is not necessary for diagnosis.
B. Type II tumors, for example serous papillary carcinoma, usually occur in women
in their sixth and seventh decades and are not associated with estrogen stimula-
tion, and therefore do not occur in a background of complex atypical hyperpla-
sia but rather in atrophic epithelium. They are more likely than Type I tumors
to be at an advanced stage at the time of presentation and so have a relatively
poor prognosis. Genetically they are characterized by TP53 mutations.
The WHO classification of uterine corpus malignancies is presented in
Table 33.1, and the pathologic staging of uterine corpus carcinomas is shown
in Table 33.2 and Figure 33.1.
1. Endometrioid adenocarcinoma usually arises in the uterine corpus and grossly
usually consists of a raised to exophytic, pink tan, hemorrhagic mass that
projects into the endometrial cavity.
Microscopically, the tumor consists of irregula.t;, confluent, complex
glandular or villoglandular structures lined by pleomorphic stratified
columnar cells with pleomorphic nuclei. The presence of areas with defini-
tive cribriform architecture is a microscopic feature that can be used to
distinguish well-differentiated endometrioid adenocarcinoma from com-
plex hyperplasia with cytologic atypia. Foci of squamous differentiation,
which should not be mistaken as solid component of the tumor, are often
encountered.
Myometrial invasion is recognized by the presence of an irregular
endometrial-myometrial border or by an associated desmoplastic and
inflammatory stromal response (e-Fig. 33.33). The depth of myometrial
invasion compared with the full thickness of the myometrium and the pres-
ence or absence of lymphovascular space invasion should be noted.
c - 33 • umr.o (Celp.al I au
''I.I IIJU\ I WltO Hlttlllolfc Cllulllcaltluf r - of 1M UIIIIM C«p~~t
flltillllllltl-•ro-lal.. -l"'l!.wn -~~
Eildomdllll <11<hlma Ollfuoo lolofn:lom-
Eildomdllolcllllon_..l...,. oe.-,lolofn)o>ma
V•ulolrt..tl! O<lllOmOU4dll!'erell1lalb'! IMIIIWI\OIIO lelotr(,oom'*>olo
~n<hJor1111~nt -etqlolorn)<l"""*
-.y,.llollt Mlloolh,_,, """'""'hymal1>1mo111
Cllldod col "'lllnl Mllod ...somoiJIII- and sm-
Mudnow:.tdlnOCIIIUtoma ml.ad& hti'TIOA
Sotwt edanocarcfnoma Pa-~ropill- ... llmo<
c::loor <oil &donOCIIcinM>O Adonomml:l tllmo<
Mblod QO!Iodiflocor.-.M>O Other meiiN!i ~~ tumore
Squt.n'd.i& eel t~~~n::!l"'l:::''''::l Otllor benl!l> """"""hymal1>1mo111
TIINI!Ionllcol c.clrana -ll!ltillllalnl•-•••.,_
Smll coli aol<hlm1 C._,..,me l)n&Ja11ont mblal mLI-
Undll!'--me tlimo<l
otllcn AdOIIOOl!CI)mll
E'lldomdlllll\';l>ell>lool; 0.-bra/M
NonaypUIIM>eiJll&aM Adenollbn>ma
stnplo Ad..,..,..,.
C<m!Mx Alyplcal polyJdd ..... nt
~ hyperplula
stnplo - ..... ,111 tllllt 1 ' -
'J)ot>hobloollc ·~laomo
C<mliox Chod.,..l<hlma
r.,.,.. .._
E'lldomdllll DOtr'l> _ Pl.e<>lrtll sill trat>ho- tlimOf
Ejlllholold trat>hoblullc lumor
.... . ,. . . . .Ill
Molu p""""nclos
Eildomdllll.....,.land "'IIIDd ll.mora IWalldlfllnn mol&
-P-
E'lldomdllll-1 nodulo Oomplelo
E'lldomdllll-1 ..,_,.,..., arade Pal!lal
Ul'ld~~I:I!I/'QOI'I'\1
Stnot:th muatl& turr.:R
Lelomyaun:oma ~~·~. nonmolutrat>ho-losbui
Ejlllll--1 tllo no<lulo and lllaquo
hil:p<>lcl¥1rlont
Sm- muoelo 111m ore of u""""lh
'*"lrtll
"-!lllod olio
- ,....,..,.,.
. . . . . e-n
malpllt -1181
Ser. c:.orll·lllio Iii'"""'
Lelomyano, notdllellolaa apodlled
Nol.rcoctQdon'nl tumn
HIIIDI:!P: ¥1111n11
Mllotlcallf - ..tbrt
...
1\J 11101$ of...,., QlllllYI>O
CdiiAir w.rllnt
OhiO
HomorrhOIIc colkllu \lllllolrt
E'dlll- """"
Jotpolclv.rtlnt ~····---~~~~·--
""fiN nllflilpll"'""
~ ..lllnl
Upololom)'M'II "'lllnl
,......,...,....
uou-
11.ani .. D _ , .
,...,._ m
n•
c.t~p~•
1X Pttndryt.rmor 011nnot be !l!nMMd
TO ~-""'of ptlmll)' 11/mor
1lo Cln:tlom.t In 1111 (pnoln- camom.t)
T1 I TUITIOf con!lnodto""""' Witl
n. lA TuiTIOf llm!lod """cloi1\0ii'llm « lnwdoolim tllln -half
of111e II'(I'Oinolllum
Tlb
T2
Tao
19
II
III A
TUm«ln'OILIM.....,. but-
TUm«ln'OILIM ~Wor """"of ttl& ~urn
not-.! bo)l>nd wru1
TuMOt-MIODOIId/ofa.d.- (dlrecl-nllon«
-)
T.lb 1118 Vqholl-(dl'ect--.,« mldllltalll)
T~ riA TUm«.........,.. bloddor mu .... 01\d/of b<Mol m1100011
Ml Ml Ollourt ...-~o (a<ludlnJI:,_*" ~. pel>lo
...,
r«
.... .,.,.-(II) Mr'tll:t, ,. td l"'leXX)
Ml Ollourt-llt(eo<ludo&m-.oi&W> penl.llrielymph
node<,~. peMe ....... t l etlMl!!)
lis NO MO
Tlo NO MO
Tlb NO MO
T2 NO MO
T3a NO MO
T3b NO MO
T1-T3 Nl MO
Tl-'D N2 MO
T4 AllYN MO
An/T AnyN Ml
Chapter 33 • Uterus (Corpus} ! 2!
I'1,1,! If!.,
I ~an: FIGO tta;,.., ,.,.__,.IJid Enclomttltll . _ . .
11......D _ , .
,....,._ m
n•
Collj:io1•
,.
....
1X Prtn:tuy tl.l"n« ctntu:t btl • stened
TO No-...:adplt!wyi!Jmor
T1 I Tumllfllmbd to tho ulaM
n. tA Tumor ~5 em In Jff18b:t1t d!mstl.lb'l
Tlb 18 111m« >5""
T2 II 111m« -do lloYond 11\o tnonlo. v.tlilo111o110tlt>
T2o IIA Tumor lnwtte& l!ldl'lllrltf.
T2b liB Tumor I n -oiNt 110M<-
T3
Tao
Tab
...,...1.,.,.-
T~
Ill
lilA
1118
IVA
(II)
-·
Tumor 11'11'11:rtla abdomNI tlaul!lll
tu,pll'tlllllfqfot•N
fllqo I Tl
fet.,..u••••
NO
••••tutef
MO
cr.eJ.., ••
fllqo lA Tie NO MO
SlqtiB Tlb NO MO
Slqj:lll T2 NO MO
Slqj:IIIIA T3e NO MO
Sla.e>IIIB Tab NO MO
fllqo IIIC TI-T3 Nl MO
fllqo IVA T-' AllyN MO
SIQt Ml Any T AllyN 1.41
Chapter 33 • Uterus (Corpus) I 52 9
..,..""'"
Dope of'***
1 •" ' "nmoto
aiJpla: ' - - mill {i...,lfiQUII) or modaafi>lo....,llt (oi,VIIIclnll
p,...,...,• - lliCTCN
----·-
M~llld<ll
I~ mllollclndtill. bitt_..,..,
Mil!: 20 Plf hpf, no CTCN, nolfW!eor no""'"' thon mill ~laill::lfW!eololo,..,... wtlll
lmbd
Ml < 10 por hpf, no~ but\01111- m - " ....,. ~ "'YJlle -<typloal
loa>m)orna \01tl Nrtok d """'rr..,..
Ml;. 10 por 10 hpl, no CTCN bulwtlll dll'lllt modln !<>.....,.~ J~Jl!ia
~
Ml < 20 por lOhpf, no CTCN but wtlllfoalll modaofi>lo _,~ <IJplo- otypcoi
lelorr\IO>rno. but~ lmlllld
Ally Ml,- d - mold-'>,_.. ""Yf* and wt111 ~- . . , . , _ , . ,
Ml?; lOper !Ohpt, nofi> rnlde1yJlla blt- ~----•
Ml < 10. noli> mild olyJ:il b<h!lh CTCN --111 rn.-lllmoroftow mdjp\1111 pollntlll,
but..,..,,.,..,lmllod.
Chapter 33 • Uterus (Corpus) I 53 1
lltlln
11.ani .. D _ , .
,...,._ m
n•
c.t~p~•
1X Prtn:tuy tl.l"n« ctntu:t btl • stened
TO No-...:adplt!wyi!Jmor
T1 I Tumor a>nflnod to ..,rpu& ut.w1
n. tA Tumllfllmbd to ondomollhmlancloooM<
Tlb 18 Tumor fn'IZidn 11m than OfiOohiltf ~ m~m
Ttc lC 'lllm«lnloodoe on&llohr m010 d ~m
T2 II 1\imor .....,do beyond 11\e """"'· ..tllln 11\e poMI!.
T2o IIA Tumor fnwt.te& adl"'lrrtf.
T2b liB Tumor I n -oiNt poMI:-
T3
Tao
T3b
...,...1.,.,.-
T4
Ill
lilA
1118
IVA
(II)
-·
Tumor fn'Jdws abdomhlf U.va:t
(e-Fig. 33.67). The cells are immunopositive for cytokeratin, calretinin, and
vimentin expression, but immunonegative for factor VIII-related antigen and
CD31 expression-a profile that can be used to distinguish the tumor from
metastatic carcinoma and vascular tumors. Adenomatoid tumor is clinically
asymptomatic and usually found incidentally.
XII. OTHER MISCELLANEOUS NEOPLASMS
A. Lymphoid neoplasms involving the uterine corpus (of which the most frequent
is large B cell lymphoma) most commonly represent a manifestation of dissem-
inated disease. Primary disease of the uterus is extremely rare.
B. Metastatic tumors only rarely involve the uterus. The most common primary
tumors that spread to the uterus are those of breast, lung, stomach, gallblad-
der, thyroid, and melanoma. In most instances, uterine involvement by primary
tumors of the ovary, cervix, bladder, and rectum/colon represents direct exten-
sion rather than hematogenous spread.
SUGGESTED READINGS
Mazur MT, Kurman RJ. Diagnosis of Endometrial Biopsies and Curettings. A Practical Approach.
2nd ed. New York: Springer; 2005.
Tavassoli FA, Devilee P, Eds. Tumours of the Breast and Female Genital Organs. 1st ed. Lyon:
International Agency for Research on Cancer; 2003.
Uterine Cervix
Michael E. Hull
• All e-figures are available online via the Solution Site Image Bank.
535
536 I SECTION VII: REPRODUCTIVE TRACT
8 7 6 5 4
Figure 34.1 Gross processing of conization specimens. After the endocervical and stromal mar-
gins are inked, the specimen is radially sectioned so that each tissue slice includes the endocervical
margin, the mucosal surface of the endocervical canal, and the ectocervix.
epithelium (e-Fig. 34.1); with full maturation, it may appear very similar to
native squamous epithelium. Metaplastic squamous cells in cytologic smears
occur as either singly dispersed cells or small sheets of cells, and they show
cyanophilic cytoplasm and nuclear sizes and N/C ratios between those of
normal intermediate and basal cells.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION. Cer~
vical specimens for screening and diagnosis are obtained in several ways.
A. Exfoliative cytolao (Pap test). See the section below on cytology of the uterine
cervix for a discussion of the Pap test.
B. Biopsy. Colposcopic cervical biopsy specimens are small pieces of mucosa and
superficial stroma that are taken, most often, from acetowhite areas identified
visually. Documentation of the number and size of tissue fragments is impor-
tant to ensure that the biopsy tissue fragments are adequately represented on
the slides. If a tissue fragment exceeds 4 mm in maximal dimension, it should
be bisected prior to histologic processing. In general, the biopsy tissue should be
wrapped in lens paper or placed between sponges to avoid loss during process~
ing, and the tissue should be embedded such that the microscopic sections are
perpendicular to the mucosal surface. Three H&E~stained levels are prepared
for microscopic examination.
C. Curettaae. Curettage specimens consist of numerous and often miniscule tissue
fragments in mucus, so it is imperative to both filter the contents of the container
and collect any tissue that may be adherent to the pad or paper submitted
within the specimen container. It is necessary to wrap curettings in lens paper to
avoid loss during processing. The specimens obtained from curettage procedures
should be submitted in their entirety. Three H&E~stained levels are prepared
for microscopic examination.
D. Conization. Ideal cold knife cone biopsy specimens consist of a single tube of
ectocervix and cervical canal surrounded by stroma. Marking sutures attached
by the surgeon enable the sections to be designated using the hours of the clock;
by convention, the mid~anterior location is the 12 o'clock position. The endo~
cervical margin must be identified and inked differentially from the ectocervical
and stromal margins. After fixation, the specimen should be radially sectioned,
with each section encompassing the endocervical margin, the mucosal surface of
the endocervical canal with the transformation zone, and ectocervical margin,
as shown in Fig. 34.1.
E. Loop electrosuraical excision procedure (LEEP). The key to correct processing
of these specimens is identification of the endocervical margin (which may be
Chapter 34 • Uterine Cervix I 53 7
infected cells at the edge of the ulcer (e-Fig. 34.3). Cytomegalovirus is dis-
tinctive for its nuclear and cytoplasmic inclusions. Adenovirus is notable
for its smudged nuclear inclusions. The poxvirus Molluscum contagia-
sum generates large, round, intensely eosinophilic cytoplasmic inclusions,
as it does in its cutaneous sites. Human papillomavirus (HPV) infection is
closely tied to cervical neoplasia; its features are discussed in the sections
on preinvasive and invasive squamous neoplasia.
c. Granulomatous cervicitis. Infectious causes of granulomatous cervicitis
include Mycobacterium tuberculosis and Treponema pallidum infections.
As noted above, noninfectious etiologies are also in the differential diag-
nosis of granulomatous inflammation.
d. Fungal. Candida spp. are commonly encountered in smears and are not
necessarily always pathogenic. They cannot be speciated reliably on their
morphology, in either tissue sections or cervical smears.
e. Parasitic. Trichomonas vagina/is is one of the most common causative
agent of sexually transmitted infections in women. Many infections are
asymptomatic. In Papanicolaou-stained smears or liquid-based prepara-
tions, an ovoid organism with an eccentric nucleus is observed; in liquid-
based preparations, squamous cells may be coated with the organism.
3. Vasculitis. Most cases of vasculitis involving the gynecologic tract are inci-
dental, and the vasculitis is often confined to the cervix. However, some cases
are associated with known collagen vascular disease, and rare cases repre-
sent the first manifestation of a collagen vascular disorder (e-Fig. 34.4) (Int
f Gynecol Pathol. 2000;19:258).
B. Atrophy. Epithelial atrophy is seen in the postmenopausal state, when estro-
gen levels are decreased. As noted in the discussion of normal histology, high
estrogen states are associated with large numbers of superficial squamous cells;
however, when the epithelium is thinned, the histologic picture is dominated by
small cells with increased N/C ratios and nuclei at least as large or larger than
those of normal intermediate cells. The overall appearance of a well-organized
epithelial architecture and a lack of nuclear atypia distinguish atrophy from a
severe squamous dysplasia.
C. Metaplasia. Tubal metaplasia occurs most frequently in the upper endocervix.
Transitional-cell metaplasia is rare and recapitulates urothelium; it is not asso-
ciated with a specific insult and must not be confused with neoplasia. Intestinal
metaplasia is another uncommon metaplasia; it features columnar epithelium
with goblet and Paneth cells.
D. Hyperplasia
1. Squamous hyperplasia consists of thickening of the epithelium with normal
maturation. It occurs in situations of prolapse and chronic irritation.
2. Squamous papilloma is a benign squamous proliferation that covers fibrovas-
cular cores. Squamous papilloma may be associated with HPVinfection but
it does not show classic koilocytic atypia.
3. Microglandular hyperplasia is an increase in glandular elements in the cervical
stroma. Seen in histologic sections, the exuberant proliferation sometimes has
a cribriform architecture that can raise the question of neoplasia. However,
microglandular hyperplasia shows no cytologic atypia, does not infiltrate the
stroma, and is not associated with a desmoplastic reaction.
4. Lobular endocervical glandular hyperplasia is a benign proliferation of bland
glands usually surrounding a central dilated gland and forming a well-
circumscribed lobule.
5. Diffuse laminar endocervical glandular hyperplasia. In this entity, the prolifer-
ation is primarily in the very superficial aspects of the stroma and extends in
a band-like fashion with an intermingled lymphocytic infiltrate that may be
dense.
Chapter 34 • Uterine Cervix I 53 9
-·lztl!
Kerdllltl.r
a-Ioiii
""""""""
WI~
Plpllaty
~PhOOI'IIhelome-llte
SqullnOlnlno!ISohol
EaJti lnYuW. (m~ tqu....,... ... .....,lnoma
$qUimOUlfnt7a.oplhlllll noq>lula
C.Abll-pllllllllnooplalo (aNi!) sq...,.,....oollcormomaln t1u
Ballln""""mOUI<dloama
Cond)bna awrrlnlllln
$qUIInOUO Dllfilkimo
Flbtooj>thollol """"'
GM\dulltb.lrnon.l!l"d PfdC:UrtOta
Adonoarmom•
Mudnou.a tdlnoa.rdnorna
EndOCIIit::ef
~..I
~ ......
Mlnlmai-
WicPndulor
Endorntl'llokf ad«icc:adiiOmt
Clolr cd lllla,_.lnorna
Selouo &denocoreiiiOitlO
~lie-~-
E&Jti lnYut.o.odontJCIIJ'dn"""'
Adonoarmom•lntlu
Gllnclullr~
l!onlln pondulor - .
Mllllotlon popa,me
EndocoM:o!PQtlp
Olllor tptilolll tuii\OI'l
AIJ~I.IM'I'IOUl t~~~n::!l'd'M
Glau)'CIIIIcarmom• ¥1Nnt
Adonotl <)011;..rdnoma
Adonotl - ....11'1011'11
Nel.rconllocth wmoro
Corct.otl
~I.,.J<ftld
SmlllooiiCirmoml
l.a1Jt01!lnaui'CIIftodoa1na arclnana
Undllalln- <11<h>ma
~lltumoroeNIIU....,....CUidhlollo
U.'"-""
El>d......nolll - · ..""""., ""'1Vfdle
Undlll..,llot.od tn6oo1Abl arama
(~
~34· UtariMCer.u I 141
I' f.t:] I J:~l\1 WHO Hlttololfc Clltltflr:a'tbl•t 1-.. of tttt Ulllrfnt Ctr\lllc
' - (CIIIIIIuilt
s.,.... l><lllldd•
A -!lOft Pill......,.
Anp,_•
Moll&nont l*llilnl""""' ~tum«
Lolom)'lmt
Genllll l!lobdornyana
--llvotptldlt ... . -..
Mklld tpi:NIIJ t.nd n~l tumn
C.-rcoma (mollnlllt rri>ald MO!Iollan tum..-)
Ad-
WII'no'tnnor
Adonoftbltma
Adonomyama
Mollnct)1Sc tumrn
Moll&nont
Bb>nowa
m-•
Mlloooila1100114111no~t
1\JII\0/I "'pnn ooii'YPO
Yalk uc tumt:~
o.mold.,.
Moture 0)'/ltio to1111ome
Ll"'pi>DU ..,d t>omldojlolallc tum""'
Moll&nont IJmlilorno COI>Odfi'IYI>Ol
l.o&ll:otlllo ~II'J !JPOl
S&condt.ry tuMift
542 I SECTION VII: REPRODUCTIVE TRACT
This suggests that MEC may actually be an entity distinct from adenosqua-
mous carcinoma in the cervix.
5. Adenoid basal carcinoma. This tumor occurs in a population similar to that
of adenoid cystic carcinoma. It also shows a nested cribriform architecture.
The epithelium, howevet; is composed of more uniform, round to oval,
basophilic cells, often with squamous differentiation, without significant
atypia or increased mitotic activity. There is often associated CIN. Correct
identification of this lesion is important because it is low grade and does
not have aggressive behavior; in fact, some have suggested that the tumor
is more appropriately termed adenoid basal epithelioma (Am] Surg Pathol.
1998;22:965).
E. Neuroendocrine neoplasms. A variety of neuroendocrine neoplasms may occur
in the cervix, although rarely. The classification of these tumors is similar to that
of neuroendocrine tumors of the lung.
1. Carcinoid. These benign tumors are organoid in their architecture and are
composed of small, oval to spindle cells with granular cytoplasm. Mitoses
are rare in typical carcinoid tumors. Immunoreactivity with neuroendocrine
markers synaptophysin, chromogranin A, and neuron-specific enolase is the
rule.
2. Atypical carcinoid. Moderate cytologic atypia and the presence of 5 to 10
mitotic figures/10 high-power fields (HPFs) are sufficient to classify a carci-
noid as atypical; at least small foci of necrosis are often present (Arch Pathol
Lab Med. 1997;121:34). These tumors generally retain the organoid archi-
tecture of typical carcinoids. Their biologic behavior is difficult to assess sys-
tematically due to the subjectivity involved in separating atypical carcinoids
from typical carcinoids and large cell neuroendocrine carcinomas.
3. Large cell neuroendocrine carcinoma. These tumors show frequent vascular
invasion, have higher mitotic activity than atypical carcinoids (> 10 mitotic
figures/10 HPFs s), and show loss of the organoid architecture seen in less
aggressive neuroendocrine tumors. There may be focal adenocarcinoma-like
areas with abundant cytoplasm and large nucleoli. Necrosis is frequent. The
prognosis is poot; similar to that of small cell carcinoma.
4. Small cell carcinoma. Histologically identical to its counterpart in the lung,
small cell carcinoma (also known as high-grade neuroendocrine carcinoma)
is a tumor of variably sized, round to oval to spindle cells with scanty cyto-
plasm, high mitotic activity, nuclear molding, and frequent crush artifact
(e-Fig. 34.18). Necrosis may be extensive. Clinical series are small due to the
rarity of the tumot; but the prognosis is uniformly poor.
F. Mesenchymal neoplasms. A wide variety of sarcomas may be primary to the
cervix, including leiomyosarcoma, embryonal rhabdomyosarcoma, endometri-
oid stromal sarcoma, alveolar soft part sarcoma, and angiosarcoma (e-Fig.
34.19).
G. Mixed epithelial and mesenchymal neoplasms
1. Adenosarcomas are polypoid lesions that microscopically consist of large
papillae of malignant stroma covered with benign endocervical epithelium.
The stroma can have many different appearances; it can consist of plump,
mitotically active spindle cells or more undifferentiated round cells similar to
those of small cell carcinoma. Heterologous sarcomatous elements may be
present, with skeletal muscle, cartilage, adipose, or osseous differentiation.
Prognosis after excision is apparently good, although only small numbers of
cases have been reported.
2. Malignant mixed Mullerian tumor {MMMT), or carcinosarcoma, also presents
as a polypoid mass. In contrast to its more common counterpart in the uter-
ine corpus, the malignant epithelial component is more often squamous or
basaloid, as opposed to glandular. The sarcomatous component is usually
••• I SECTION VII· REPRODUCTIVE TRACl
,....,a_m
n•alnslt ••
_,.,.,
'11111
1)(
TO
......
,.
be·-
Pltnoilytnnor oaiii>Ot
No cwtSanc:a<lf ptlma.ry tumw
n. I) C.n:b>rna In aii!J (pnoln- - I M )
T1 I C.,_....t!Oma..,.INdtl>&aNa(--.tl><l>fllu•
-ld bedkt.-)
Tla lA lnw.oM> cordn..,.,. d~ onlY b\'MicrooooDY
Tlal IA1 Slromllln- no >3.0 mm In d'!>illlnd 7.0 mm or loa In
holtlolalollteod
Tl.:! lA2 Slromllln- >3.0 mm and not >6.0 mm will! a ho<lio>nl£1
llji'I!Od 7.0 mm or losa
Tlb
Tlbl
IB
1~1
- _,.r
Cllrbllf >fllblo- oonflnod tD lllo ""Mx « mlc._ll:
ll\ln TIAQ/IA2
Cllrblli>fllllio....,., 4.0 em orlooo In..,._ dlmOi'IOb'l
T1b2 182 Cllrbllf >fllblo- >4 om In IP"'I88Idmonalon
T2 II 1\lm«ln\OI.doo bo!r'Md Ub!N• bill notm poMe ...u...., .._.
llllrd oflllo "'!!hh
T2a IlA Wlllt<Mpo~-
T2al 11111 CIIrblli>fllllio....,., 4.0 em <1t looo In..,._ dlmOi'IOb'l
T2a2 IIA2 Cllrbllf>fllbla- :>4.0 em In""""""d i -
T2b liB Wllfti>O!OmOOiallhVMion
l3 Ill Tumo<..- tl> poMc Mil• . . , _ -1111111 of ...na. or
.......,.l!)ldrmec>""'* or nonftmetlonlnJ!:I<I<Woy
l3a II"' Tumo< - -1111111 of "''ln a. no""""*'"*' poMo wd
T3b 1118 T u " " " - t.o poMc wd o r - h)'dron'!>hrosls t1f
non~~~.-~na ~
--·
T4 I\IA Tumot' ~ mutote Gf tUddar « llldum or adDndl baya1d
INa poM!.
NO MO
NO MO
NO MO
NO MO
NO MO
NO MO
NO MO
NO MO
NO MO
NO MO
Nl MO
AnyN MO
Any N MO
AIIJN Ml
F.,,. ~ SB, e,rd DR, Com...... OC:, .. t l , - AJCC C1wor511,py-.ol 7111 ed. N'"'"""'
NV,~-- 2010.llood- p e - .
Chapter 34 • Uterine Cervix I 54 7
I. SPECIMEN TYPES
A. Liquid-based preparations are the most widely used modality for cervical screen-
ing (Pap test). A brush is used to sample the cervix and the sample is placed in
appropriate transport fluid for the proprietary system used in the laboratory, be
it ThinPrep (Hologic, Inc., Marlborough, MA) or SurePath (BD Diagnostics-
TriPath, Burlington, NC). Advantages of this modality include a cleaner back-
ground, a monolayer of cells, increased diagnostic sensitivity, and ease of per-
formance of HPV assays.
B. Conventional smears are made by using a spatula and/or brush to sample the
cervix, smearing the endo- and ectocervical samples on a glass slide, and then
immediately fixing the sample. Papanicolaou staining is performed in the labo-
ratory.
II. EXFOLIATIVE CYTOLOGY OF THE CERVIX
A. Squamous cells. A spectrum of squamous cells, ranging from small parabasal
and metaplastic cells with dense cytoplasm and relatively high N/C ratios
548 I SECTION VII: REPRODUCTIVE TRACT
(e-Fig. 34.20), to intermediate cells with medium-sized round nuclei with open
chromatin and polygonal cytoplasmic outlines (e-Fig. 34.21), to superficial cells
with polygonal shapes and small pyknotic nuclei (e-Fig. 34.22) is seen in normal
cervical smears and liquid-based preparations.
B. Glandular cells. The classic appearance of endocervical glandular cells is sheets of
regularly spaced cells forming a so-called "honeycomb" pattern (e-Fig. 34.23);
varying degrees of disruption of this architecture herald reactive and neoplastic
changes. Endometrial cells are smaller and form more three-dimensional aggre-
gates in cytology preparations; they are also typically more degenerated than
endocervical cells. Careful attention to nuclear details such as the chromatin
pattern and nuclear contours is required to separate glandular neoplasia from
reactive atypia.
Ill. THE BETHESDA SYSTEM was developed in 1988 with the objective of standard-
izing terminology to promote better communication between the laboratory and
clinicians. It is in its second edition (2001).
A. Adequacy. A statement of adequacy is required. Cellularity should be 5000 cells
for a liquid-based preparation, and this can be reproducibly and quickly esti-
mated with experience and with the use of reference images. Having 75% of
squamous cells obscured by blood or inflammation renders a specimen unsatis-
factory, as does improper labeling or slide breakage.
B. Negative for intraepithelial lesion or malignancy
1. Microorganisms should be reported. Various causes of vaginitis can be
detected cytologically.
Trichomonas vagina/is is seen as a pear-shaped structure, "'30 ~-tm in
diameter. It has a small pale nucleus and red cytoplasmic granules. The flag-
ella are often difficult to appreciate (e-Fig. 34.24).
Bacterial vaginosis is marked by the presence of "clue cells," which are
coccobacilli-coated squamous cells (e-Fig. 34.25). This is reported as "shift
in flora suggestive of bacterial vaginosis."
Actinomyces infection shows filamentous organisms and the "sulfur gran-
ules" described above. This infection is associated with runs.
Candida albicans is responsible for most cases of vulvovaginal candidi-
asis. Although the fungus can be a commensal microorganism, when it is
accompanied by acute inflammation, the infection is usually symptomatic.
Budding yeast and/or pseudohyphae are seen (e-Fig. 34.26).
Herpes genitalis is usually caused by HSV type 2. Nuclear enlargement,
chromatin margination, multinucleation, nuclear inclusions, and nuclear
molding are evident (e-Fig. 34.27).
2. Other nonneoplastic findings
a. Reactive changes/repair in squamous cells include nuclear enlargement (up
to twice the size of an intermediate cell) with or without multinucleation,
smooth nuclear contours, small nucleoli, and mild nuclear hyperchroma-
sia. In reparative change, there is vacuolization of the cytoplasm and some
of the cells may be elongated, clustering together in a streaming pattern.
Repair implies disruption of the epithelium with subsequent increased pro-
liferation. The relative abundance of cytoplasm and lack of true nuclear
atypia differentiate this from neoplastic squamous proliferations (e-Fig.
34.28). Reactive endocervical cells may show even greater nuclear enlarge-
ment, multinucleation, mild hyperchromasia, and prominent nucleoli
(e-Fig. 34.29).
b. IUD. The recognition of run-related changes is particularly important in
cervical smears, as the singly dispersed cells with vacuolated cytoplasm
and large nuclei seen in this reactive condition can mimic adenocarci-
noma. Despite nuclear enlargement, truly atypical nuclei are not seen.
Correlation with the history is key when run-type changes appear.
Chapter 34 • Uterine Cervix I 54 9
I. NORMAL STRUCTURE. The vagina is derived from the miillerian ducts and is com-
posed of three layers: mucosa, muscularis propria, and adventitia. The mucosa
is composed of squamous epithelium overlying a lamina propria that contains a
rich vascular and lymphatic network with scattered stromal cells that may show
multinucleation.
II. BENIGN CONDITIONS
A. Infectious diseases
1. Vulvovaginal candidiasis is a common condition that predominantly affects
adult women in their second and third decades. Up to 70% of women
will experience at least one episode in their lifetime. Common predis-
posing factors include antibiotic use, steroid use, oral contraceptive use,
immunosuppression, and uncontrolled diabetes. Pruritus, erythema, and
thick white vaginal discharge are the most common symptoms. Histolog-
ically, squamous epithelial hyperplasia with hyperkeratosis and/or parak-
eratosis is seen. Foci of neutrophilic infiltration of the squamous epithe-
lium are commonly present. Candida can be present in the form of bud-
ding yeasts as well as pseudohyphae, highlighted on GMS or other special
stains.
2. Bacterial vaginosis is most commonly found among adult women. It is caused
by Gardnerella vagina/is, a bacillus which usually grows when the vagi-
nal flora shifts toward a more acidic environment. A watery, malodorous
discharge without significant inflammation is a common symptom. Micro-
scopically, the bacteria overgrow and cover the squamous cells, producing
so-called clue cells.
3. Trichomoniasis, a sexually transmitted disease, is caused by Trichomonas
vagina/is, an oval protozoon with flagella. Microscopically, the organisms
are identified by their bluish-pink body, elongated nuclei, and flagella.
4. Herpes simplex infection is a sexually transmitted disease caused by herpes
simplex virus (HSV). Grossly, the virus causes a mucosal ulceration within a
few days to 2 weeks following the exposure. These lesions are highly infec-
tious until crusting, with final scarring occurring within 2 to 3 weeks of
initial symptoms. The majority of cases are caused by HSV-2, and recurrence
is higher with infection by HSV-2 than by HSV-1.
Microscopically, the ulcerated lesions are characterized by epithelial
necrosis with associated degenerated cells containing viral inclusions, best
identified at the periphery of the ulcer. The cells with viral inclusions have
characteristic features including multinucleation and ground glass nuclei with
a rim of chromatin condensation at the nuclear border surrounded by a cyto-
plasmic halo.
5. Actinomyces-like organisms are most commonly seen in women with noncop-
per intrauterine contraceptive devices.
B. Inflammatory diseases
1. Atrophic vaginitis occurs most commonly in postmenopausal women, but
can also occur during the postpartum period. Grossly, the primary finding is
punctate hemorrhage of the vaginal mucosa. Microscopically, the squamous
cells show decreased glycogen due to lower estrogen levels. Atrophy can be
distinguished from vaginal intraepithelial neoplasia (VAIN) by the monotony
551
552 I SECTION VII: REPRODUCTIVE TRACT
of the cell population, uniform chromatin, the lack of cytologic atypia, and
the low mitotic rate.
2. Crohn disease can result in rectovaginal fistula formation and is associated
with fibrosis, chronic inflammation, and granulomas. The differential diag-
nosis includes vaginal fistulas of other etiologies including radiation therapy,
perforated colonic diverticulum, or as a complication of hysterectomy.
3. Stenosis, ulceration, and necrosis are well-described sequelae of radia-
tion therapy. Stenosis can also follow severe bullous erythema multiforme
(Stevens-Johnson syndrome).
C. Cysts
1. Miillerian cysts are the most common type of vaginal cyst and can be lined
by endocervical-, endometrial-, or endosalpingeal-type epithelium (e-Fig.
35.1).*
2. Epithelial inclusion cysts are lined by keratinizing squamous epithelium and
filled with white sebaceous and keratinous debris. They most commonly arise
in areas of previous trauma such as episiotomy sites.
3. Mesonephric cyst. Also known as Gartner duct cysts, they are usually located
along the anterolateral wall of the vagina (along the path of the mesonephric
duct). This type of cyst is lined by low cuboidal, nonmucinous epithelium.
4. Bartholin gland cysts are thought to develop from obstruction of the ducts of
Bartholin glands, which normally open on to the vestibule. The cyst lining
varies from squamous to transitional to mucin secreting (e-Fig. 35.2).
D. Adenosis occurs in about 30% of women who were exposed to diethylstilbe-
strol (DES) in utero and is associated with an increased risk of clear cell adeno-
carcinoma (see section on clear cell adenocarcinoma below). Adenosis usually
involves the upper third of the vagina, but the middle third or lower third are
affected in about 10% of cases. Grossly, adenosis presents as a red erythematous
granular lesion. Microscopically, adenosis is defined by the presence of colum-
nar epithelium of endometrial or endocervical type in the vaginal mucosa or
underlying submucosa (e-Fig. 35.3).
E. Endometriosis of the vagina comprises < 10% of cases of pelvic endometrio-
sis. The diagnosis requires the presence of miillerian-type epithelium (most
commonly endometrioid) and endometrial-type stroma. Hemosiderin-laden
macrophages are often present as well. The presence of endometrial type stroma
can be used to distinguish endometriosis from adenosis (e-Fig. 35.4).
Ill. BENIGN NEOPLASMS. The WHO classification of vaginal tumors is given in Table
35.1.
A. Epithelial
1. Squamous papilloma is usually asymptomatic and can occur at any age.
Grossly, it usually presents as a cluster of papillary lesions. Microscopically,
squamous papillomas have a fibrovascular core and are lined by benign squa-
mous epithelium.
2. Fibroepithelial polyps most commonly occur in adult women during their
reproductive years. They occur in the lower third of the vagina and grossly
have a soft and papillary surface. Microscopically, they are composed of squa-
mous epithelium with underlying hypocellular fibrovascular stroma. Atypi-
cal myofibroblasts are common in the stroma, and scattered multinucleated
cells with bizarre atypical nuclei may also be seen (e-Fig. 35.5). Howevet;
rhabdomyoblasts and a cambium layer are not present and mitotic figures
are rare; these features, together with patient age, distinguish fibroepithelial
polyp from sarcoma botryoides.
*All e-figures are available online via the Solution Site Image Bank.
Cl'l•ptw 35. Ylgil\1 I Ill
--.
Squln'dl& 011.....,1.,.,., notdlle- opodllod
Non-~11\11
Buolold
v.,._
W.l\'
Squlll'IOUllnln!optlllolll nooplalla
BoniiJI> II<IIMMOI.It l!olono
Caut.rtoma ac~~mlnltlm
Squunoua paplloml (...,nol ml:topo~llontmbl
FI>IOOI'illlolal DOtrl>
G_,lu tumo<s
O.r <:61 a.deKoeardnOI'I'llll
Eildomdltold&llonoarcl...,.
Mudnow:.tdlnOCIIIUtoma
MOIIollon 1101>llomo
Adonomo
01t.or •ptholllltl.mOra
NJIIfiCI:Iqulll"'IOUt carcfnana
Adonold ""*"",.,.,..,'
Ad.,.ld--non
carcmld
Slnol coil.,.,...,_mo
Und!l!'w-..s camo1111
Mil ,,,.........11 ... Dt•..:t . .
So....,. boO)ddoo
Ltlomyourcoma
Eildomdltold ....,.,ll_,o,la.olpde
Und!l!'o.-IOI,Ihll......,...
Ltiolll)'lmO
Gon!lll rllobdoii'I)Qnl
Deep Jt\lbii)XOiila
"-DOI"""OIII'Idlo coiM<k!lo
-.s ........... ,•. ,,,..,_
~ (m.,.,ont m0-n miud IIUMI)
Ad-
Malpnl mbotd l!.lrnot rMOmlllll\ll synoW~•rama
Bani&1'1 rnb<od tum«
-·OCJII·-
Malpnlm-•
Bblnowa
Mollnct)1Sc nowa
. . 55,....... , _ .
llunora of .conn coillYI>O
Yalk uc: tumt:~
Donroold..,...
00..
"""'hn Plfm"*' _...,.ltumo~tumOf
Ad-rnalold bJMOf
._.."_
~~~-, ........ke-n
Malpnl !lm1JIIom•
~.a~mlo
554 I SECTION VII: REPRODUCTIVE TRACT
I. NORMAL ANATOMY. The vulva or external female genital region encompasses the
mons pubis, labia majora, labia minora, clitoris, and vestibule. The entire vulva
except for the vestibule is covered by keratinized, stratified squamous epithelium.
The epithelium of the vestibule is glycogenated squamous epithelium. The lateral
aspects of the labia majora and the mons pubis contain hair follicles. Sebaceous
glands are present in the labia majora and the perineum. The clitoris is lined by
keratinizing stratified squamous epithelium overlying paired corpora cavernosa
that contain vascular spaces surrounded by nerves.
The urethral meatus, major vestibular glands (Bartholin glands; e-Fig. 36.1), *
minor vestibular glands, paraurethral glands (Skene glands), and vagina all open
onto the vulva. The Bartholin glands are paired glands that open posterolaterally
on the hymenal ring; they are composed of acini lined by cuboidal mucus-secreting
epithelium that drain into a duct that may be lined by mucus-secreting, transitional,
or squamous epithelium, depending on the location from deep to surface. Skene
glands open on either side of the urethral meatus and are composed of acini lined by
mucus-secreting epithelium that open into ducts lined by transitional epithelium.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION
A. Vulvar biopsies. Vulvar biopsies should be oriented as for skin biopsies (see Chap.
38) and three H&E stained levels examined.
B. Vulvar resections It is helpful to ask the surgeon to orient the specimen with a
diagram or labeled sutures so that orientation can be maintained during process-
ing. The margins of resection should be inked and, depending on the location of
the resection, the periurethral, vaginal, and perianal margins need to be noted.
In cases with an obvious malignant neoplasm, one section per centimeter of
tumor, including the areas closest to the deep margin; lateral margin, and/or
other margins are recommended. In cases where no tumor is observed grossly,
the entire specimen should be submitted. Because many gynecologic oncologists
consider resection for squamous cancer in this area to be adequate only if tumor
is >8 mm from the margin (Cancer. 2002;95:2331), radial rather than shave
margins should be taken of all but the most obviously negative margins so that
the distance from tumor to margin can be measured.
Ill. DIAGNOSTIC FEATURES OF COMMON DISEASES OF THE VULVA. Many inflammatory
and neoplastic conditions that affect the skin will also affect the vulva. These are
discussed in the skin chapters (see Chaps. 38 and 39). This section only covers
those conditions for which the vulva is a common site of disease.
A. Inflammation
1. Bartholin abscess presents as a painful swelling in the area of the Bartholin
gland. Microscopically, there is acute inflammation of the Bartholin duct,
glands, and connective tissue, with purulent luminal contents. The etiology
includes Neisseria gonorrhea, Staphylococcus, or other aerobic or anaerobic
organisms. Treatment includes excision, drainage, and appropriate antibi-
otics.
2. Hidradenitis suppurativa presents as painful subcutaneous nodules in areas
containing apocrine glands, particularly the vulva and axilla. Initial changes
*All e-figures are available online via the Solution Site Image Bank.
558
Chapter 36 • Vulva I 55 9
include acute and chronic inflammation around hair follicles, which progress
to abscess formation, sinus tractformation, and dermal scarring (e-Fig. 36.2).
Treatment may include laser ablation or total excision of the involved area.
3. Crohn's disease may present as vulvar or perianal erythema, ulceration,
abscesses, or fistulas between bowel and vulva or between two different
areas of vulva. Microscopically, there is acute and chronic inflammation of
the deep dermis, often with associated noncaseating granulomas, fistulas, or
sinus tracts (e-Figs. 36.3 and 36.4).
B. Infection
1. Candida infection is often a chronic inflammatory condition of the vulva that
may be associated with diabetes. It often presents as pruritis and clinically
shows areas of redness with thickened, edematous skin. Microscopically,
there is acanthosis with acute and chronic inflammatory cells in the epithe-
lium, and parakeratosis with neutrophils. Often fungal organisms are visible
on H&E stain in the keratin layer; they are easily identified by silver stains.
2. Syphilis is a sexually transmitted disease caused by the spirochete Treponema
pallidum. The primary lesion of syphilis, the chancre, develops in about
half of the women within 3 weeks of infection and is characterized by one
to sometimes multiple painless, dean-based ulcers. The ulcer heals in 2 to
6 weeks without a scar. Secondary syphilis develops within 6 weeks to
6 months and is characterized by the development of a rash on the palms,
soles, and mucosal surfaces, as well as elevated plaques and papules (termed
condyloma lata) on the vulva and mucosal surfaces. On microscopic sections,
the chancre shows epidermal ulceration, dermal acute and chronic inflam-
mation with numerous plasma cells, and severe arteritis. Condyloma lata are
characterized by marked epidermal acanthosis and hyperkeratosis, dermal
inflammation with numerous plasma cells, and arteritis. The organisms may
be detected on Warthin-Starry, Steine.t;, or Dieterle stains; no organisms are
seen in some cases of active infection.
3. Human papilloma virus {HPV) infection. Condyloma acuminatum, also referred
to as genital warts, is the result of sexually transmitted infection caused by
HPV types 11 (75% of cases) or 6 (25% of cases). They present as asymp-
tomatic, usually multiple or confluent, papillary or papular lesions, and may
occur anywhere on the vulva or perianal region.
Microscopically, condylomata of the vulva typically have a fibrovascu-
lar stalk. The epithelium exhibits acanthosis, papillomatosis, hyperkeratosis,
dyskeratosis, and an accentuated granular cell layer (e-Fig. 36.5). Viral cyto-
pathic effect, termed koilocytosis, takes the form of cytoplasmic clearing
around enlarged nuclei with irregular nuclear outlines and clumped chro-
matin (e-Fig. 36.6). Vulvar condylomata usually follow a protracted course.
They may grow rapidly during pregnancy and then regress after delivery.
Small condylomas may be treated with topical agents while large ones are
excised, or treated with laser ablation or cryotherapy.
4. Herpes simplex virus {HSV). Infection with HSV type 2, or less commonly type
1, is typically heralded by feve.t;, dysuria, and severe pain. Painless vesicles
then appear which progress to an intensely painful ulcer. The ulcer typically
heals in about 2 weeks. Microscopically, epithelial ulceration is surrounded
by virally infected keratinocytes that exhibit multinucleation, "ground glass"
nuclear chromatin, or eosinophilic nuclear inclusions (e-Fig. 36.7).
5. Molluscum contagiosum is a sexually transmitted disease in adults caused by
infection with the Molluscum contagiosum poxvirus. The lesions are small,
3 to 6 mm diameter papules with a characteristic central depression or umbil-
ication, and are usually asymptomatic although perianal lesions may be pru-
ritic. Microscopic features (e-Fig. 36.8) include formation of a cup-shaped
papule with marked epidermal acanthosis, and intracytoplasmic inclusions
560 I SECTION VII: REPRODUCTIVE TRACT
that are initially eosinophilic but become more basophilic as the lesion ages.
Most lesions regress spontaneously.
C. Noninfectious squamous lesions
1. Lichen sclerosus presents as symmetric plaque-like areas of white, thinned
epithelium that may be superficially ulcerated. In advanced cases, there may
be scarring of involved areas and stenosis of the introitus.
Microscopically, there is typically a band-like lymphocytic infiltrate in
the upper dermis with spongiotic changes to the basal layer of the epider-
mis and loss of melanocytes from the overlying epidermis. Thinning of the
epidermis with flattening of the rete and a zone of collagenous connective
tissue immediately beneath the epidermis is frequently present (e-Fig. 36.9)
and was the basis for the previous nomenclature "lichen sclerosis et atroph-
icus." However, it is now recognized that a spectrum of histologic changes
can be seen in the disease, ranging from a predominantly lichenoid infiltrate
without dermal collagenization to prominent dermal scarring with minimal
chronic inflammation. Treatment involves high-dose corticosteroids. Post-
menopausal women with lichen sclerosus have a small risk of developing
differentiated VIN (see below) and squamous cell carcinoma.
2. Lichen simplex chronicus (formerly "squamous cell hyperplasia") typically
occurs in adults and presents as a localized area of pruritus (] Reprod Med.
2007;52:3). It is thought to be a nonspecific response triggered by a vari-
ety of irritants. Clinically, the area is white or red, with accentuated skin
markings and sometimes areas of excoriation. The characteristic feature on
microscopy is marked acanthosis without atypia, increased mitotic activ-
ity, inflammation, often with features that overlap other specific dermatoses
(e-Fig. 36.10). Hyperkeratosis may be present. The dermis is normal. Treat-
ment includes limiting exposure to irritants, topical corticosteroids, and
antipruritic agents.
D. Cystic lesions
1. Banholin cyst. Obstruction of the Bartholin duct leads to the accumulation of
secretions and the formation of a cystic dilatation of the duct. The epithelium
lining these cysts may be squamous, transitional, or mucinous. Cysts can be
treated by drainage, marsupialization, or excision of the gland.
2. Keratinous cysts occur at any age and typically affect the labia majora. They
are small, measuring just a few millimeters in maximal dimension, and are
filled with white cheesy material without hair. Microscopically, they are lined
by stratified squamous or flattened epithelium. They can be excised if symp-
tomatic.
3. Mucus cysts occur in the vestibule and are lined by mucinous epithelium with
or without squamous metaplasia. They are probably the result of occlusion
of minor vestibular glands.
IV. TUMORS. The WHO classification of tumors of the vulva is presented in Table 36.1.
A. Benign tumors and tumor-like lesions
1. Fibroepithelial polyps are also known as acrochordons or skin tags. They may
be hyperpigmented, hypopigmented, or flesh-colored, and typically occur on
hair-bearing skin. They usually have a papillomatous or pedunculated growth
pattern and a soft cut surface. Microscopically, the epithelium may be thick-
ened with hyperkeratosis, or may be flattened. The stroma contains loose
bundles of collagen and may be edematous. Fibroepithelial polyps are clini-
cally insignificant but can be excised if they are cosmetically unacceptable.
2. Papillary hidradenoma is a benign tumor that originates from apocrine sweat
glands. It presents as a dome-shaped mass, usually <2 em in diameter, arising
between the labium majus and labium minus. The mass may ulcerate and
bleed but is usually asymptomatic. Microscopically, papillary hidradenoma
forms tubules and acini lined by a luminal layer of epithelial cells and an outer
Clllptlr 315. Yuhrl I Ill
-·~-""'
Ill I SECTION VII· REPRODUCTIVE TRACl
,,(,I,!IJf.J'I WltOHid*lfcCIIMIItlcl1!on•fr-ot1MMa(~)
-rtllftpd....,ma
Upgs~"""""
il<mll1o!l-rcoma protubonlnl
DooPtn&bnYJoomo
SUporfleio.. r9otn)'I!OI'I\l
All....l)'OIIbroi>-
Co!llllr ~lroma
l.olon\)'Om.t
Gnnullr tal tumot"
Olhe!l
r a . . . .. . . .
Mo..,. .............
eon..,a.l mMIOCjllc"""""
kquhd moloo....,U.""""'
Bhnt l"'ee'MM
Alrolcol m~ """"'of1ho ..,..,"""
Dill>-......tqtle ,_..
. . . . . e-n
Yolk ta<: lllmor
MotlcdCIItumOf
ar.twtumOI/IIO!thettl 011mat.oo nw-etmal111nor
H. . .,........ , Itt U t1n1n
......,.....
Mo..,_.niJtmphoma ("'*'f(y typo)
Lou- !•1*1'1~
-
FI'Otne THBII ~ fllal.otee P,ada.. ltblt1 HfttiJIIf; ~~a/~ ~Md
- - . -dllls--Rimo.IISMaol~lp> IARCA'.... ~ u... ....,
....
Clllptlr 315. Yuhrl I Ill
..._.. ._..
''I.I II lf:!.! II TN II aa.lllel1ioft of Cltd- .tlllt Vulwl
TMM ~aa
I. NORMAL ANATOMY. The normal, unfixed term placenta weighs 350 to 550 g,
trimmed of membranes and cord. The placenta consists of three parts: fetal mem-
branes, umbilical cord, and placental disk.
The fetal membranes insert at the edge of the disk and envelop the fetus and
amniotic fluid. Microscopically, they are composed of a cuboidal amniotic epithe-
lium with underlying connective tissue, a chorionic layer (composed of connective
tissue, intermediate trophoblast, and degenerated villi), and sometimes a layer of
decidua (gestational endometrium) (e-Fig. 37.1).*
The umbilical cord is composed of two umbilical arteries (e-Fig. 37.2) and
one umbilical vein (e-Fig. 37.3) surrounded by Wharton's jelly, a paucicellular
connective tissue matrix. Its outer surface is lined by a layer of cuboidal amniotic
epithelium.
The placental disk is typically oval and microscopically composed of chori-
onic villi surrounded by maternal blood in the intervillous space. The chorionic
villi contain vessels of the fetal circulatory tree embedded in mesenchymal stroma
(e-Fig. 3 7.4). A layer of cytotrophoblast encompasses the villous stroma, and this is
surrounded by a layer of syncytiotrophoblast that is in contact with the intervillous
space. The maternal surface of the placental disk, which is adjacent to the uterine
wall, contains variable amounts of fibrin, intermediate trophoblast, and decidua.
The umbilical cord inserts near the center of the placental disk, and branches of
the umbilical cord vessels arborize over the shiny fetal surface of the disk. Micro-
scopically, the fetal surface of the disk is lined by amnion and chorion.
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION
A. Fetal membranes. The fetal membranes should be assessed for completeness. The
presence of green, blue, or brown staining, indicating meconium or hemosiderin
staining, should be noted. The membranes should be inspected for amniotic
bands, nodules of amnion nodosum or squamous metaplasia, and hemorrhage.
The distance from the insertion to the disk edge should be measured as this gives
a rough estimate of where the placenta was implanted in the uterus. A strip of
membranes should be cut from the rupture site to the disk insertion site, one end
grasped by a forceps, and the strip rolled around the forceps; this membrane
roll should be eased off the forceps into formalin. At least one cross-section of
this membrane roll should be examined.
B. Umbilical card. The length of the cord is measured, including any detached
segments. Note should be made of marginal insertion (at the disk edge), or vela-
mentous/membranous insertion (into the membranes). Abnormalities of cord
color (meconium staining) should be noted. Focal abnormalities such as stric-
ture, hematoma, knots, nodules, plaques, or amniotic bands should be noted
and measured. Cross-sections should be made at regular intervals throughout
the cord length, and the number of vessels and the presence of thrombi should be
noted. At least two cross-sections of cord should be examined microscopically,
avoiding the area just above the insertion site where the two umbilical arteries
fuse.
*All e-figures are available on line via the Solution Site Image Bank.
566
Chapter 37 • Placenta I 56 7
C. Disk. The disk should be assessed for completeness and measured in three dimen-
sions. After examining and removing the membranes and cord, the unfixed disk
should be weighed. The fetal surface of the disk, which is covered by amnion
and chorion, is examined for the same abnormalities as the membranes. The
branches of the umbilical cord vessels are examined for lacerations, calcifica-
tions, and thrombi. The maternal surface of the disk is examined for retropla-
cental hematomas, indentations, or other focal abnormalities. The disk is then
sliced at 1-cm intervals, and each slice is examined and palpated. The color,
location (central vs. peripheral), size, texture (finn vs. spongy), demarcation
(whether well circumscribed or ill defined), and number of all focal lesions are
recorded. An estimate of the percentage of the placental parenchyma involved
by each type of process is noted. Any organized blood clot in the container
is measured. At least two sections of central placenta that include fetal and
maternal surfaces as well as sections of focal lesions should be submitted.
D. Multiple gestation. Placentas from twin gestations may have completely sepa-
rate disks, a fused disk with two gestational sacs, or a fused disk and just one
gestational sac (monoamniotic). If present, the dividing membranes should be
inspected. The percentage of placental parenchyma associated with each twin
should be determined. A roll of the dividing membranes should be made as
for the fetal membranes, and at least one cross-section of the roll should be
examined microscopically to confirm the gross and ultrasound impression of
chorionicity. The chorionic plate vessels should be inspected for anastomoses.
In monochorionic or monoamniotic placentas, the type of anastomoses (artery
to artery, artery to vein, vein to vein) should be investigated by air injection
and recorded, keeping in mind that arteries cross over veins. Note should be
made of unpaired large vessels as these likely represent areas of physiologically
important deep artery-to-vein anastomoses.
Ill. DIAGNOSTIC FEATURES OF COMMON DISORDERS OF THE PLACENTA
A. Fetal membranes
1. Meconium. With recent meconium passage the fetal plate and membranes
will be yellowish-green and slimy. With longstanding meconium passage,
the membranes, fetal plate, and even the umbilical cord will be dull
brown. Microscopically, the amniotic epithelium is stratified and tufted with
pyknotic nuclei. There is marked edema between the amnion and chorion.
Macrophages in this area are filled with yellowish-brown, waxy, meco-
nium pigment (e-Fig. 37.5). Pigmented macrophages may also be seen in
the chorion and decidua.
Meconium passage may be the result of neurologic maturity in the fetal
intestines, but may also be associated with chronic in utero hypoxia, or
stressors closer to delivery. Rarely, meconium induces vascular necrosis in
umbilical vessels.
2. Hemosiderin deposition may stain the fetal plate and membranes brown or
green. Often there is old blood clot where the disk meets the fetal membranes.
Circumvallation (see Section III.D) may also be present. Microscopically, the
membranes do not show the epithelial stratification, tufting, and edema seen
with meconium. Membrane macrophages contain refractile pigment that is
positive with an iron stain. Sometimes a layer of hemosiderin is deposited
in the basement membrane beneath the amniotic epithelium (e-Fig. 37.6).
Diffuse chorioamniotic hemosiderosis is an indication of chronic peripheral
separation and is associated with oligohydramnios in the absence of mem-
brane rupture, preterm delivery, and chronic lung disease.
3. Amnion nodosum forms small, grayish-white, discrete nodules or plaques that
may occur anywhere on the cord or fetal membranes but are most common on
the fetal plate near the cord insertion. These nodules, which represent vernix
caseosa, are easy to remove with a cotton swab. Microscopically, they consist
568 I SECTION VII: REPRODUCTIVE TRACT
of fetal squamous cells, amniotic epithelial cells, and sometimes fetal hair
(e-Fig. 37.7). Sometimes nodules of amnion nodosum become re-
epithelialized by contiguous amniotic epithelium.
Amnion nodosum is the result of oligohydramnios. It therefore serves as
a marker of conditions such as renal agenesis that may cause decreased fluid
production, and can also alert to possible complications of oligohydramnios
such as pulmonary hypoplasia and limb positioning abnormalities.
4. Squamous metaplasia. Plaques or nodules of squamous metaplasia are present
in nearly every placenta. They are tan-white and may be seen anywhere on
the cord, membranes, or fetal plate of the disk but are most common on the
fetal plate near the cord insertion. Squamous metaplasia is difficult to remove
with a cotton swab. Microscopically, squamous metaplasia in the placenta is
identical to that present elsewhere in the body. Squamous metaplasia is not
clinically significant.
5. Amniotic bands may appear as shredded amnion on the fetal surface of the
placenta or as thin adhesion-like threads connecting one part ofthe fetal plate
to another, connecting the fetal plate to the umbilical cord, or attached to the
fetal digits or other fetal parts. Microscopically, they are composed of fibrous
tissue often with no attached amnion. Amniotic bands are associated with
a wide variety of abnormalities in the fetus including digital amputations
(e-Fig. 37.8), deft lip and palate, and body-wall defects. Characteristically,
the defects are asymmetric, no two cases are identical, and the spectrum of
defects in any given case does not fit into a recognizable genetic syndrome.
Amniotic band syndrome only extremely rarely recurs in a subsequent ges-
tation.
6. Fetus papyraceous. Occasionally, a mummified remnant of an embryo from
much earlier in gestation will be compressed on the fetal membranes
(e-Fig. 37.9), referred to as fetus papyraceous. It may represent an unrec-
ognized twin gestation or may be the result of selective termination of a
higher order gestation.
B. Umbilical cord
1. Length abnormalities. The normal umbilical cord is 55 to 60 em long. Short
cords (<35 to 40 em) occur in about 5% of cords; they are usually associated
with conditions of decreased fetal movement such as amniotic bands, oligo-
hydramnios, body-wall defects, fetal neuromuscular disorders, and arthro-
gryposis. Long cords (>80 em) occur in about 5% of cords; long cords are
associated with an increased likelihood for encirclement around the fetal
neck or other body part, knots, cord prolapse, and marked cord twisting.
2. Single umbilical anery (SUA}. The incidence of SUA (e-Fig. 37.10) is about
1%, and SUA is about 4 times more common in twins. There is a strong
association between SUA and congenital malformations, mortality, and low
birth weight. SUA is likely an acquired defect as the incidence is lower earlier
in gestation; in fact, the absence of the artery may be the cause of associated
malformations. In some cases of SUA, a small, atrophic remnant of the second
artery can be seen (e-Fig. 37.11). SUA is also strongly associated with other
cord and placental abnormalities such as velamentous insertion, marginal
insertion, extrachorialis, and shape abnormalities.
3. Abnormal cord insenion
a. Marginal insenion. In marginal insertion, the umbilical cord inserts at the
edge of the disk (e-Fig. 37.12). This occurs in 6% to 18% of placentas
and is not clinically significant.
b. Velamentous insenion. In velamentous insertion, the umbilical cord inserts
into the fetal membranes (e-Fig. 37.13). This insertion abnormality is seen
in about 1% of placentas. In about 75% of these cases, the vessels branch
within the membranes before the branches insert into the placental disk;
Chapter 37 • Placenta I 56 9
in 25% of cases, the cord vessels run through the membranes without
branching. Because the branches of the umbilical cord are not protected
by Wharton's jelly, they are at risk for compression, thrombosis, and lac-
eration.
4. Umbilical cord knots. About 1% of umbilical cords have a true knot (e-Fig.
37.14), which may be loose or tight. Differences in the diameter and color
of the cord on either side of the knot should be noted. Cords with size
differences, particularly with a dusky appearance between the knot and the
fetus, are likely to be associated with an adverse outcome. The cord on either
side of the knot should be examined microscopically for thrombi, a feature
that suggests a clinically important knot. The knot should be untied in the
fresh state to look for persistent grooving of Wharton's jelly, a feature that
suggests chronic tightening. The fetal mortality rate for umbilical cord knots
is reported to be between 5% and 11%.
5. Umbilical cord coiling. The normal cord has a left-handed twist, a feature
that is thought to increase turgor, preventing compression of cord vessels. A
coiling index can be determined by counting the number of complete turns
divided by the length of the cord (which can be compared to a standard
reference). About 5% of cords will have no twist, a finding that has been
correlated with increased fetal mortality, operative delivery for fetal distress,
abnormal karyotype, preterm delivery, and fetal heart rate abnormalities.
Hypocoiled cords, with a coiling index below the 1Oth percentile, and hyper-
coiled cords, with a coiling index above than 90th percentile, have also been
associated with a variety of adverse outcomes.
6. Umbilical cord stricture is a focal area of cord that is markedly narrowed
with a depletion of Wharton's jelly, fibrosis, and often thrombosis of the
umbilical cord vessels. The most common location for a stricture is the area
adjacent to the umbilicus. There is a high association between cord stric-
ture and stillbirth, especially early in gestation. Most cases also show excess
twisting.
7. Umbilical cord hematomas occur once in every 5 500 deliveries, although small
hematomas have been documented in 1.5% of cases following ultrasound-
guided cord blood sampling. Hematomas nearly always occur in the portion
of cord closest to the fetus and present as a fusiform swelling with a dark,
hemorrhagic color. It is important not to confuse a true hematoma, which
will be obvious at the time of delivery, with blood accumulation in the cord
as a result of clamping, blood drawing, or other manipulation during or after
delivery. Large hematomas have a perinatal mortality rate of 50%, whereas
small ones have very low fetal morbidity or mortality.
c. Circulatory disorders
1. Infarcts are firm and well circumscribed, with one edge usually abutting the
maternal surface of the disk (e-Fig. 37.15). Early infarcts are red whereas
older infarcts are white. Sometimes there is central hemorrhage. Micro-
scopically, there is collapse of the intervillous space, which crowds the villi
together (e-Fig. 37.16). Depending on the age, the trophoblast may be pale
and degenerative (recent) or show little staining with only ghost outlines of
villi (longstanding).
Infarcts are common, occurring in 10% to 25% of term placentas from
normal pregnancies, typically at the periphery. Extensive infarction, infarcts
>3 em, infarcts that occur in the central placenta, and infarcts in the first or
second trimester of pregnancy are clinically significant and often indicate
significant underlying maternal disease such as pre-eclampsia, collagen vas-
cular disease, or a hereditary thrombophilic condition. Infarcts are caused
by an interruption in the maternal blood supplied by a given spiral artery
to an area of placental tissue.
570 I SECTION VII: REPRODUCTIVE TRACT
The normal placenta can lose 15% to 20% of the parenchyma with-
out adversely affecting the fetus. However, in placentas that are chroni-
cally underperfused, such as in pre-eclampsia, a lesser degree of infarction
may be clinically significant. Extensive infarction may cause fetal hypoxia,
intrauterine growth restriction, periventricular leukomalacia in preterm
infants, or fetal death.
2. Massive perivillous fibrin deposition. When large amounts of fibrin are
deposited in the placenta, a firm, white or yellow, slightly gritty, ill-defined
mass is formed (e-Fig. 37.17). Often small pockets of red, villous tissue are
interspersed within strands of white fibrin. Microscopically, there is expan-
sion of the intervillous space by eosinophilic fibrinoid material pushing
the villi away from each other (e-Fig. 37.18); clusters of cytotrophoblast
proliferate in the fibrinoid material, and the villi entrapped in this fibri-
noid material become ischemic. The amount of fibrin deposition needed
for diagnosis of massive perivillous fibrin deposition or to be associated
with an adverse outcome for the fetus is not well established. Some studies
have found that entrapment of 20% of the central-basal terminal villi is
associated with adverse outcomes (Arch Pathol Lab Med. 1994;18:698);
others have defined clinically significant fibrin deposition as fibrin extend-
ing from the fetal to maternal surface and entrapping 50% of villi on at
least one slide (Pediatr Dev Pathol. 2002;5:159). Massive perivillous fibrin
is associated with intrauterine growth restriction, periventricular leuko-
malacia in preterm infants, and fetal death, and may recur in subsequent
pregnancies.
3. Maternal floor infarct. The term maternal floor infarct is a misnomer in that
it is a form of fibrin deposition, not infarction. It is defined as perivillous
fibrin deposition surrounding at least one-third of the villi adjacent to the
basal plate, often with extension of fibrin into the underlying decidua. An
alternative definition is that basal villi of the entire maternal floor be encased
in fibrin at least 3 mm thick on at least one slide. As in massive perivillous
fibrin deposition, the villi in maternal floor infarct that are surrounded by
fibrin undergo ischemic changes.
Maternal floor infarct is quite uncommon, seen in far < 1% of placen-
tas. It is associated with stillbirth, intrauterine growth retardation, preterm
delivery, and neurodevelopmental impairment. It may recur in subsequent
pregnancies.
4. Subchorionic fibrin deposition is common and appears as firm, oval, tan-
white, slightly raised plaques of the fetal surface of the placenta, beneath
the amnion and chorion. On cut section, it is laminated and clearly beneath
the membranes but above the villous tissue (e-Fig. 37.19). Microscopically,
sections show layers of blood and fibrin beneath the chorion. Subchorionic
fibrin plaques are not clinically significant.
5. Retroplacental thrombohematomas occur in about 4.5% of placentas. They
are organized blood clots beneath the maternal surface of the placenta that
indent the placental surface. Recent retroplacental hematomas are soft, red,
easily dislodged, and are often seen in the specimen container rather than
adherent to the placenta by the time the placenta arrives in the labora-
tory. Older hematomas are firm, brown, and densely adherent with def-
inite placental indentation (e-Fig. 37.20). Microscopically, retroplacental
hematomas consist of organized blood clot and fibrin, and the underly-
ing placental parenchyma may be infarcted depending on how long the
hematoma has been present (e-Fig. 37.21). Sometimes the villi immedi-
ately beneath the thrombohematoma exhibit villous stromal hemorrhage
in which the vessels are disrupted and the stroma contains extensive red
cells (e-Fig. 37.22).
Chapter 37 • Placenta I 57 1
howevet; are larger and heavier than normal. The microscopic findings are
not specific but are nonetheless characteristic. The villi are often edematous
and immature for gestational age. The cytotrophoblast is prominent. There is
irregular thickening of the trophoblastic basement membrane. Chorangiosis,
avascular terminal villi, and SUA are increased in frequency.
Women with diabetes are more likely to deliver stillborn infants or infants
with malformations and/or macrosomia. There is no relationship between
these adverse outcomes and the severity of the placental findings.
3. Maternal thrombophilic disorders. There is increased interest in the relation-
ship between hereditary thrombophilic disorders (such as protein C and
S deficiency, factor V Leiden, and hyperhomocysteinemia) and pregnancy
complications. Although controversial, it appears that various hereditary
thrombophilic conditions, alone and in combination, are associated with an
increased number and larger infarcts, acute atherosis, spiral artery thrombi,
retroplacental hematomas, and fetal thrombotic vasculopathy.
4. Sickle cell disease. The placentas from women with sickle cell disease may
be small and may have an increased number of infarcts. A characteristic find-
ing is the presence of sickled maternal erythrocytes in the intervillous space
(e-Fig. 37.37). Sickled maternal red cells may also be seen in the placentas of
women with sickle cell trait.
F. Multiple gestations
1. 1Jpes of placentation
a. Diamniotic dichorionic. In this type of placentation, the placental disks may
be completely separate or fused. Each fetus is enveloped by its own ges-
tational sac composed of amnion and chorion. The dividing membranes
are thick. Sections show amniotic epithelium from each twin with fused
chorion from both twins (e-Fig. 37.38).
Dizygous (fraternal) twins exhibit diamniotic dichorionic placenta-
tion, but about 25% of monozygous (identical) twins also exhibit this
type of placentation if the blastocyst splits within the first 3 days post-
fertilization. Because diamniotic dichorionic twins do not share vascu-
lar anastomoses, these twins are the least likely to have complications
such as fetal loss, preterm delivery, and twin-twin transfusion syndrome
(TITS).
b. Diamniotic monochorionic placentation. In this type of placentation, the
placental disks are typically fused. Each fetus is enveloped by its own
gestational sac lined by amnion. A single chorionic layer surrounds both
sacs so that the dividing membranes are composed of only fused amnion
from each sac with no intervening chorion (e-Fig. 37.39).
Twins with monochorionic placentation are monozygous. This type of
placentation is seen in about 75% of monozygous twins and results when
the blastocyst splits between 4 and 7 days after fertilization. Twins with
monochorionic placentation usually share vascular anastomoses and are
therefore at risk for complications such as fetal loss, preterm delivery, and
TITS.
c. Monoamniotic placentation. In monoamniotic placentation, the twins
share a gestational sac, and therefore there are no dividing membranes
(e-Fig. 37.40). Twins with monoamniotic placentation are monozygous,
but only 1% of twins are monoamniotic. This type of placentation results
when the blastocyst splits between 8 and 13 days after fertilization.
Monoamniotic twins have a very high rate of complications, with only
50% surviving to term. They share vascular anastomoses, so are at risk
for the associated complications noted above for monochorionic twins.
In addition, because they share the same gestational sac, these twins have
a high rate of umbilical cord accidents.
Chapter 37 • Placenta I 57 5
Skin: Nonneoplastic
Dermatopathology
Samuel J. Pruden II, Kimberley G. Crone, and
Anne C. Lind
have a coffee-bean shaped nucleus. Langerhans cells are visible only via spe-
cial stains or in aggregates in chronic inflammatory disorders and in Langerhans
cell histiocytosis. Merkel cells are located along the stratum basale and are also
histologically invisible. They have recently been determined to be derived from
progenitor keratinocytes and are presumed to serve in tactile perception.
Small, slender, regularly spaced downward extensions of the epidermis (rete)
divide the superficial dermis into papillae. The papillary dermis, located immedi-
ately beneath the basement membrane, is composed of fine collagen and elastic
tissue fibers and contains the capillary loops of the vascular plexus. The reticular
dermis, with its haphazardly arranged thick collagen bundles and elastic tissue
fibers, is separated from the papillary dermis by the superficial vascular plexus.
Elastic fibers, a component of both the papillary and the reticular dermis, are
usually visible only with the aid of special stains. The dermis provides structural
support and flexibility to the skin.
The dermis also contains adnexal structures, arrector pili muscles, nerves,
and blood vessels. Adnexal structures in the skin include hair follicles and the
eccrine, apocrine, and sebaceous glands. Eccrine glands develop as downgrowths
of the epidermis. They are present as secretory coils in the deep dermis and have
a vertically oriented duct that communicates directly through the epidermis by
way of a pore called the acrosyringium. Alternatively, apocrine glands develop
from the follicular unit and communicate to the surface via a connection through
the follicular infundibulum. Like the eccrine gland they have a deep coil and
a vertically oriented duct, although the apocrine coil has a larger central space
than the eccrine coil and has the classic eosinophilic cytoplasmic apical bleb. Like
the apocrine gland, sebaceous glands are outgrowths of the follicular infundibula.
They form lobules that connect to the follicle via a small duct and are composed of
peripheral basaloid cells and central mature sebocytes with vacuolated cytoplasm.
Hair follicles have varied features depending on the type of hair they produce
and, if they are a terminal hair, whether they are actively growing (anagen), rest-
ing (telogen) or involuting (catagen). The infundibular portion of the follicle is
histologically identical to the epidermis. This portion extends from the epidermal
surface to the sebaceous duct. The isthmus has trichilemmal keratinization and
extends from the sebaceous duct to the insertion point of the arrector pili muscle
(the bulge). The lower segment of the follicle extends from the bulge to the bulb,
the classic ball-and-claw that is seen at the base of every hair follicle.
The subcutis is composed of mature adipose tissue separated into lobules by
fibrous septae. Fully lipidized adipocytes have a slender, crescentic, barely visible
nucleus that has been displaced to the periphery of the cell by the accumulated
lipid. The deep vascular plexus separates the subcutis from the reticular dermis.
II. COMMON DESCRIPTIVE TERMS
A. Acantholysis: Loss of attachment(s) between keratinocytes (e-Fig. 38.1).*
B. Acanthosis: Thickening of the epidermis (e-Fig. 38.2).
C. Bulla: Fluid containing space in the epidermis, > lcm in size.
D. Dyskeratosis: Abnormal keratinization that results in altered eosinophilic cyto-
plasm; individual dyskeratotic cells may be referred to as Civatte or colloid
bodies (e-Fig. 38.3).
E. Epidermotropism: Migration of malignant cells into the epidermis (e-Fig. 38.4).
F. Exocytosis: Migration of benign, nonepithelial cells into the epidermis, com-
monly seen in association with spongiosis (e-Fig. 38.5).
G. Hypergranulosis: Thickening (increased number of layers) of the granular layer
(e-Fig. 38.6).
H. Hyperkeratosis: Thickening of the stratum corneum.
*All e-figures are available online via the Solution Site Image Bank.
Chapter 38 • Skin: Nonneoplastic Dermatopathology I 581
Punch Biopsy
J lll
11111
,, ,cut
,
,,
Demonstrate
nearest margin
Figure 38.1 Gross processing
of a punch biopsy.
582 I SECTION VIII: SKIN
Shave Biopsy
Punch Bio~psy
for Alopecia
cut
cut
A4 A3 A2 A4 A3 A2
cut cut cut cut
cut cut cut cut
Figure 38.4 Gross processing of an elliptical excision specimen.
B. Granular parakeratosis
1. Clinical: Brownish keratotic plaques at sites where skin rubs against skin;
most common in the axillae; may be pruritic.
2. Microscopic: Keratohyaline granules are present in a confluent, thick layer
of parakeratotic keratin (e-Fig. 38.58).
c. Parakeratosis
1. Clinical: Multiple clinical types; most commonly multiple erythematous
macules with a thread-like palpable scale at their border on sun-damaged
skin (disseminated superficial actinic porokeratosis [DSAP]).
2. Microscopic: All lesions of porokeratosis have one or more cornoid lamel-
lae consisting of a thin spire of parakeratotic keratin arising from an area
of the epidermis without a granular layer and usually associated with iso-
lated, scattered, dyskeratotic keratinocytes. If the entire lesion is sampled, a
cornoid lamella can be seen at both edges/sides of the lesion. DSAP has an
atrophic epidermis between the cornoid lamella and a patchy/discontinuous
band of lymphocytes in the superficial dermis (e-Fig. 38.59).
Nonmelanocytic Tumors
of the Skin
Nathan C. Walk, Yumei Chen, Anne C. Lind,
Friederike Kreisel, and Dongsi Lu
*All e-figures are available online via the Solution Site Image Bank.
595
596 I SECTION VIII: SKIN
l3 hi&l>i111<
Tumor '""""""'<11 ITIIOdllo, mendlbb. orUI, Of Mportil bono
\011111-n
T_. Tl/morlOIIth l,.,....n <hbiiiCOfl (aldol Cit el>llOndlcoJal) <t po!Mnl
- o f olaJI baoo
bil nl.,....-{11
Nit R<P>nnl ~pi! nodoocoMal b6 · -
NO No "*"til ~ph n o d o -
Nl Mrta.-ln o '*'P> ipolll\<flallfmph -.3om« lim in-"'"
dlmOi'IOion
N2 lllelao!.eolllho,...~l r,np11 nodo.>3cm bul1101>6cmln
111-• dim"'*"; orin mtJifplo 1!>111.,.,.1 IJmph nodes, nme >6om
., .,..... dimension; or lh blllenl1 at~ t;'mph nodii!ISJ none
>6 an I n , -elm,_
r& Mrta.-ln o '*'P>
•-dim.....,
!.,.._ t~mpll node. >3om bit not >6 em In
ii. Microscopic: One or more cystically dilated hair follicles with radiating
follicles that project into a relatively cellular stroma. The secondary hair
follicles are of variable maturity and may give rise to more hair follicles
(e-Fig. 39.8).
b. Trichoepithelioma
i. Clinical: Usually solitary, skin-colored papule, most common on the
central face; if multiple, associated with AD inheritance. The desmo-
plastic variant is a solitary, firm annular lesion with a raised border
and central depression that occurs exclusively on the face.
ii. Microscopic: A relatively well-circumscribed dermal tumor composed
of islands of basaloid cells with pilar differentiation in a cellular stroma;
papillary mesenchymal bodies are characteristic. Small keratinous cysts
and foci of calcification are often present. The tumor epithelium may
mimic basal cell carcinoma, but there is no retraction artifact or muci-
nous stroma (e-Fig. 39.9). The desmoplastic variant shows a well-
circumscribed lesion in the upper and mid-dermis composed of cords or
small nests of basaloid cells in a sclerotic stroma (e-Fig. 39.10). It may
be confused with a syringoma or a morphea-form basal cell carcinoma.
c. Trichoadenoma (of Nikolowski)
i. Clinical: Rare nodule on the face or buttocks.
ii. Microscopic: A well-demarcated dermal tumor composed of multiple
cyst-like structures lined by multilayered keratinizing pilar-type squa-
mous epithelium. The cysts contain keratinous debris and no hair shafts
(e-Fig. 39.11).
d. Dilated pore of Winer
i. Clinical: Common, usually solitary, comedo-like lesion on the head and
neck, or trunk.
ii. Microscopic: A cystically dilated hair follicle, filled with loose keratin
and lined by acanthotic stratified squamous epithelium with irregular
budding (e-Fig. 39.12).
e. Trichilemmoma
i. Clinical: A solitary, small, skin-colored/pink or brown papule on the
face and neck; multiple lesions are associated with Cowden syndrome.
ii. Microscopic: A well-circumscribed, endo-exophytic tumor composed
of clear squamoid cells with glycogenated cytoplasm. The tumor
extends from the epidermis with a lobular configuration. There is a
thin peripheral rim of palisading columnar cells and variable, thick,
eosinophilic basement membrane (e-Fig. 39.13).
f. Pilomatrixoma (Calcifying epithelioma of Malherbe)
i. Clinical: Firm, deeply located nodule, most common on the face and
upper extremities; onset frequently in childhood.
ii. Microscopic: A sharply demarcated tumor in the lower dermis and,
often, the subcutis. The tumor is composed of large, irregularly shaped
tumor islands and intervening stroma. Two basic cell types are present:
basaloid cells and "shadow" or "ghost" cells. The basaloid cells resem-
ble the cells in basal cell carcinoma and are present at the periphery
of the tumor islands. The shadow cells, which have eosinophilic cyto-
plasm, distinct cell borders, and no nuclear staining, occupy the center
of tumor islands. Several layers of transitional cells with intermediate
features may be present. In addition, dystrophic calcification, a mixed
inflammatory infiltrate, hemosiderin, melanin, bone, and foreign body
giant cells can be seen (e-Fig. 3 9.14).
g. Fibrofolliculomaltrichodiscoma
i. Clinical: Skin-colored papules, most common on the face; may be
solitary or multiple. Multiple lesions are seen in Birt-Hogg-Dube
Chapter 39 • Nonmelanocytic Tumors of the Skin I 59 9
within a loose, vascular stroma. Ducts and, rarely, small cysts may
be seen in the tumor columns. There is a sharp demarcation from
the epidermis (e-Fig. 39.20).
{b) Dermal duct tumor-islands of basaloid cells similar to those of
eccrine poroma, located entirely in the dermis. An epidermal con-
nection may be found if multiple sections are examined.
{c) Hidroacanthoma simplex-islands of basaloid cells similar to those
of poroma, confined to the epidermis.
f. Acrospiroma {nodular hidradenomai solid-cystic hidradenomai apocrine/
eccrine hidradenomai clear cell hidradenoma)
i. Clinical: Solitary nodule, 0.5 to 2.0 em or more, no site predilection,
disputed histogenesis. Nomenclature for this entity is confusing.
ii. Microscopic: Variable histologic appearance, as reflected by the nosol-
ogy; usually a circumscribed, nonencapsulated, multilobula.r; cen-
tral dermal tumor with variable proportions of cystic and solid
areas; tumor cells have variable clear or eosinophilic cytoplasm and
are round, fusiform, or polygonal. Duct-like structures are typi-
cally present and may have a squamous appearance. The stroma
varies from rather fine fibrous tissue to dense hyalinized collagen
(e-Fig. 39.21).
3. Sebaceous hyperplasia and tumors
a. Sebaceous hyperplasia
i. Clinical: Yellow to whitish papules with central umbilication, typically
on the face of older individuals. It may mimic a basal cell carcinoma.
ii. Microscopic: Multiple large, but otherwise normal, sebaceous lobules
centered on a large central orifice; solar elastosis is frequently present
(e-Fig. 39.22).
b. Sebaceous adenoma
i. Clinical: Rare, solitary or multiple, pink or flesh-colored, usually
< 1 em nodule(s) on the face or scalp of adults; may be associated
with Muir-Torre syndrome (visceral carcinoma).
ii. Microscopic: Multiple circumscribed sebaceous lobules usually cen-
tered in the superficial to mid-dennis; composed of peripheral basaloid
germinative cells, central mature sebaceous cells, and a variable zone
of transitional forms. Mature cells usually outnumber the basaloid
cells (e-Fig. 39.23 ).
iii. Genetics: Loss of protein expression of DNA mismatch repair genes
(MSH-2, MSH-6, MLH-1, and PMS-2) has been found in a number of
patients with sebaceous adenoma. There is a high correlation between
lost expression of mismatch repair (MMR) proteins and microsatellite
instability (MSI).
4. Apocrine tumors
a. Apocrine hidrocystoma {apocrine cystadenoma)
i. Clinical: Solitary, translucent to bluish nodule, predominantly occur-
ring on the face.
ii. Microscopic: Several large cysts in the dermis; the cyst wall is com-
posed of an outer myoepithelial layer and an inner layer of cuboidal to
columnar cells with apocrine decapitation secretion; pseudopapillary
projections may be present (e-Fig. 39.24).
b. Syringocystadenoma papilliferum
i. Clinical: Varied; most commonly a raised, warty plaque on the scalp;
associated with nevus sebaceous in approximately one-third of the
cases.
ii. Microscopic: Deep invaginations of duct-like structures extend from an
acanthotic, variably papillomatous epidermis into the dermis; they are
Chapter 39 • Nonmelanocytic Tumors of the Skin I 601
cytoplasm and serpiginous nuclei that appear parallel to each other within
a single fascicle. No intervening fibrous tissue is present (e-Fig. 39.41).
3. Neurofibroma
a. Clinical: Soft, skin-colored, pedunculated papules or nodules. Multiple
lesions in a segmental or widespread distribution are related to neurofi-
bromatosis.
b. Microscopic: A nonencapsulated dermal or subcutaneous tumor charac-
terized by loosely arranged, wavy spindle cells in a pale-staining stroma.
There are increased small caliber vessels and mast cells in the stroma. The
spindle cells tend to surround adnexal structures rather than displace them
(e-Fig. 39.42).
4. Cutaneous schwannoma {neurilemmoma}
a. Clinical: Uncommon, slowly growing, usually solitary tumor/nodule with
a predilection for the limbs of adults. The neoplasm can be either sporadic
or associated with neurofibromatosis 2 (NF-2). Schwannomas are more
commonly seen in the deep soft tissue, intracranially, or intraspinally.
b. Microscopic: Similar to their soft tissue counterpart; form a circumscribed
and encapsulated subcutaneous/dermal nodule. The tumor cells are spin-
dled Schwann cells with indistinct cytoplasmic borders arranged in inter-
lacing fascicles. There are hypercellular areas (Antoni A) containing rows
or palisades of nuclei aligned around eosinophilic cellular processes (Vero-
cay bodies) and hypocellular (Antoni B) areas. No axons are present
(e-Fig. 39.43).
5. Merkel cell carcinoma
a. Clinical: A rapidly growing, often ulcerated, red nodule or plaque usually
arising on the sun-exposed skin of the elderly, particularly on the head,
neck, and extremities. An association with polyoma virus infection (specif-
ically, Merkel cell polyoma virus [MCPyV]) has been demonstrated in the
majority of cases. AJCC TNM staging scheme for Merkel cell carcinoma
is given in Table 39.2.
b. Microscopic: Trabeculae, nests, and sheets of small cells with scant cyto-
plasm, indistinct cytoplasmic borders, vesicular nuclei with nuclear mold-
ing, and multiple small nucleoli that infiltrate the entire dermis and some-
times the subcutis. There are numerous apoptotic forms and mitoses. Local
intralymphatic spread is commonly seen. The tumor cells are positive for
neuron-specific enolase, chromogranin, and synaptophysin and have a
characteristic "paranuclear dot-like" staining pattern for low-molecular-
weight keratin such as CK20. The tumor cells are negative for CD45,
S-100, and TTF-1, allowing distinction from hematopoietic and
melanocytic neoplasms, as well as metastatic small cell carcinoma of the
lung (e-Fig. 39.44).
c. Genetics: Deletion of chromosome lp36 is commonly seen; numerous
other chromosomal abnormalities have been described, of which trisomy
6 is the most common.
6. Granular cell tumor
a. Clinical: Asymptomatic, solitary, skin-colored nodule <3 em in diameter;
multiple lesions in ""10% of cases; most common in Mrican American
adults and women. Malignant counterpart is exceedingly rare.
b. Microscopic: A nonencapsulated dermal-based tumor composed exclu-
sively of large polyhedral cells with abundant fine to coarsely granular
eosinophilic cytoplasm, and small oval centrally located nuclei. The gran-
ular cells infiltrate between collagen bundles and surround adnexa. The
epidermis may be markedly acanthotic as in pseudocarcinomatous hyper-
plasia (e-Fig. 39.45). Tumor cells are usually positive for S-100, except
in a subset of congenital lesions reported as CD34-positive granular cell
dendrocytosis.
l'l.t II fUJI ~IWI.ttalll(TNIII).._s.:~~em.,d... c.~l
,,, , . . . . (!)
1X Prtniluy tutnOf' ¢611not be UllilliiiG d
ro N o - d plmuyll.mor ( o . t , , n c d - """"n11tion
IOithoul aooocii1Dd PrtiMrti
n. In .111tu 1Jflmuytum"
n S2 <:m mofmum tumor dlrnell&lor\
l2 >2cmbutnd>5cm-ml!.mot<l-•
l3
T4
bill ol,.. nodltOII
o.....san.-umtumOfd-n
'*"'
PrtnouytumO< """""bono, m...:lo, fln<:lo, ....
2. Dermatofibroma
a. Clinical: Brownish, round, firm dermal nodules, usually < 1 em in diame-
ter; most common on the legs of young adults.
b. Microscopic: Poorly circumscribed dermal proliferation of spindled fibrob-
lasts, histiocytes, and blood vessels. Fibroblasts at the periphery surround
the collagen bundles ("collagen-trapping"). Dermatofibromas are histo-
logically varied, and there are many corresponding named variants such
as cellulat; aneurysmal, "Monster" cell, and so on. The epidermis typically
shows acanthosis, basal keratinocyte hyperpigmentation, and a broad flat-
tened rete or a basaloid proliferation which can be mistaken for basal cell
carcinoma in a superficial shave biopsy (e-Fig. 39.47).
3. Dermatofibrosarcoma protuberans
a. Clinical: Solitary or multiple polypoid nodules arising in an indurated
plaque on the trunk or extremities of adults; slowly growing locally aggres-
sive tumor; rarely metastasizes.
b. Microscopic: A cellular dermal tumor composed of homogeneous spin-
dle cells arranged in a storiform or cartwheel pattern. The tumor cells
infiltrate between adnexa, and there is characteristically extension into
the subcutis with fat trapping. Occasional mitotic figures can be found,
and atypia is mild. The epidermis is normal, atrophic, or ulcerated (e-Fig.
39.48).
c. Genetics: The tumor characteristically exhibits the translocation
t(17;22)(q22;q13), which results in production of a COLIA1-PDGFB
fusion protein.
4. Giant cell fibroblastoma: A histologic variant of dermatofibrosarcoma protu-
berans, primarily affecting children; it has a strong male predominance and a
similar anatomic distribution; harbors the same translocation as dermatofi-
brosarcoma protuberans.
a. Clinical: Grossly, it is a firm yellow or gray tumor with a gelatinous or
rubbery consistency without hemorrhage or necrosis.
b. Microscopic: The tumor is usually hypocellular and composed of wavy
spindle cells and scattered giant cells with hyperchromatic and angu-
lated nuclei. The stroma is variable from myxoid, to collagenous, to scle-
rotic. Scattered mast cells are seen within the stroma. Irregular branch-
ing "angiectoid" spaces resembling dilated lymphatics may be seen, lined
by spindled or multinucleated cells morphologically identical to those of
the surrounding stroma (e-Fig. 39.49). The lining and stromal cells are
immunopositive for CD34 and immunonegative for CD31, S-100, actin,
desmin, and EMA.
5. Angiofibroma
a. Clinical (major types}
i. Fibrous papule of the face: A solitary, firm, dome-shaped, often flesh-
colored lesion on the nose or central face.
ii. Pearly penile papules: Tiny white papules, 1 to 3 mm in diametet;
arranged in groups or rows on the coronal margin of the penis.
iii. Adenoma sebaceum (tuberous sclerosis): Multiple papules or nodules
with a predilection for the butterfly area of the face.
iv. Digital fibrokeratoma: A solitary, thin, tall horn on a digit.
b. Microscopic: Dermal fibrosis; dilated small vessels; and variably enlarged,
angulated, or stellate fibroblasts. Concentric fibrosis around the blood
vessels is more prominent in adenoma sebaceum than in fibrous papule
of the face (e-Fig. 39.50). The epidermis of digital fibrokeratomas shows
hyperkeratosis and acanthosis of acral skin (e-Fig. 39.51).
c. Genetics: Mutations of two genes, TSC1 on chromosome 9 and TSC2 on
chromosome 16, are identified in patients with tuberous sclerosis.
Chapter 39 • Nonmelanocytic Tumors of the Skin I 6 07
-.w.--
l'olleukl:rople MF
c,.,~.~om--"""
S6Diy ~""""
Adu~ T...,.l b'fnpllome
Pltnoly-C~ lflnph~l<lfwod\'o<loon:lero
Mlwy«tlnnO\\UIII!>iude 111111' ooll b'fnJiloma (MlCL)
L,..Ji>om_ P<!>UiaUI
S.-l*!rKUibll... TGII i)mplloma
i:>.tronodll NM'<4IIIjmJiloma. n.,..llyp!
Pmwy .....,_,. poolplloral T.... lflnphonwl, unapodllod
Mlwy ~·-- ec>l<lonn~ CO&+- T..... limPhOII'III CI>IO'oillonel)
Qrta1100114 yn+ T...alllim~- (I>IO'oilloi!Oil
M>wy...anooo• CD4+ om""""'ll"'""""" ploomorpllle T..... IIMPh"""' (Pft)'otllonll)
c.t n ....ur,. .. ...
Pmwy-.-..mefilnei....,.S....IIIflnphoma
Mlwy~•lmll'lll~""'
Mlwy~. . . . ~
l'olleulor hyperpluls w1111 ~ plurna oelk
pm.y_,_,. fdlli:lo- tjmph<ma
G,_ pai!Jms: lollcoJIIr, follaar and dlfM&, dlfM&
Pmwy-.-..dlffuoo~rplkdlflnphomo.lol~
Pmwy-.-..dlffuoo~rplkdlflnphomo.-r
-le
Pltnoly-lntt-IIIQ 9-<dll!mJilomo
p......., hotnlll>lo!Jje .......
C~056+ neq>lum ti\lm..ty blude NKoel t,omi>honwll
110 I SECTION Vlllt SKIN
NO
Nl
Nit
Nib
N2
N2a
N2b
N3
BQo
Bllb
Bl
Blo
Bib
82
UJd,
Nl
·--IUI*.,...IICt-¥a--ll
Creoo 1: <krmiiiDp•lllio l)mphodiiiiOP'III!Y LNO: no otJplcol tjmplooc:)b
(OL) LNl: - n o l and boll!1od IWiool
l!mllhoeYIDo C!lotemrwod In clJoUtl
LN2: rnf<fy <typbollymphoe)«&.,.ln
$-6oollol-ra
C - 2: Dl; .. ~~y -men! l>y MF LN3: _ , . . . , 1\Y!llalllym~
(pr-of -lform nuclei noclll•rclllloclura p -
>7.6pm
Ored& 3: 1>0111al e1lacetnollt of LN LN4: l•n11ol'oornp!AI .-non~ of
,...hbolu~&: monv o~Yll~ool-llonn nodal•rclllloclun> tr.r W!>lcol
monM.-oeiii{CI.ACI) l)mplloeyloaorhnlcti ""'plulle-.
C-<4: complollo-ont
612 I SECTION VIII: SKIN
diagnostic or surgical procedure. Metastases tend to occur on the skin near the
primary malignancy; metastasis to sites distant from the primary tumor is more
common in tumors that demonstrate angioinvasion (e.g., primary tumors of the
kidney or lung).
Rarely, a cutaneous metastasis is the first indication of a visceral malignancy; the
umbilicus, and less frequently the scalp, is a particularly common site of involve-
ment in this setting. Adenocarcinoma is more frequently observed as a cutaneous
metastasis than is squamous cell carcinoma or urothelial carcinoma. Cutaneous
metastases are variable in clinical appearance and can occur either as solitary or
multiple papules/nodules, or as mimicks of inflammatory/infectious conditions.
A. Sister Mary Joseph nodule
1. Clinical: This condition was named after Sister Mary Joseph (1856-1939), a
surgical assistant for Dr. William Mayo, who noted the association between
paraumbilical nodules observed during skin preparation for surgery and
metastatic intra-abdominal cancer confirmed at surgery. It is usually a soli-
tary, firm, indurated nodule, sometimes with surface fissuring or ulceration;
it is variable in size and can be painful. Common underlying tumors include
gastrointestinal malignancy (>55%, male predominance) and ovarian malig-
nancies (34%).
Sister Mary Joseph nodules account for 60% of all malignant umbilical
tumors (primary or secondary). Most patients die within months after the
appearance of the umbilical tumor.
2. Microscopic: Most commonly, a dermal-based adenocarcinoma with histol-
ogy resembling the primary malignancy. Signet-ring cell morphology may be
present, especially from a gastric primary (e-Fig. 39.60).
B. Metastatic breast carcinoma
1. Clinical: Typically small papules, ranging from 1 to 2 mm in diameter to large
tumor masses, on the anterior chest wall. Intralymphatic spread of tumor
cells (inflammatory carcinoma) can manifest as a diffuse, warm, indurated
plaque (carcinoma erysipeloides). Scalp metastasis can present as alopecia
(alopecia neoplastica), which is also seen in metastases from other visceral
sites, especially from a lung primary.
2. Microscopic: Usually a poorly differentiated adenocarcinoma. Ductal and
sometimes lobular arrangements are present. The proportion of intravascular
tumor varies (e-Fig. 39.61).
C. Metastatic renal cell carcinoma
1. Clinical: Typically erythematous, vascular papule or nodule, may be misdiag-
nosed as lobular capillary hemangioma or Kaposi sarcoma; solitary in 15%
to 20% of cases. The scalp is a common site of involvement.
2. Microscopic: Usually well-circumscribed, dermal nodule(s) composed of
sheets and nests of polygonal tumor cells with clear cytoplasm, distinct cyto-
plasmic borders, and enlarged hyperchromatic nuclei; associated with a del-
icate rich vascular stroma, red blood cell extravasation, and hemosiderin
deposition. The histologic features can be bland, and the differential diagno-
sis includes other primary and metastatic tumors with clear cell morphology
(e-Fig. 39.62).
Skin: Melanocytic
Lesions
Anne C. Lind, Nils Becker, Emily A. Bantle,
and Louis P. Dehner
*All e-figures are available online via the Solution Site Image Bank.
613
114 I SECTION Vlllt SKIN
...,..
MaiiJ ..rrt Colllllll!*'l
lillnllltln ar •IIJIIIIII
JIO\d!onol ~
..tholltAD
·-•willa' Mo~I'IOITIIIn ollu, ~rqjnous« tuparl\l:llll
Ol>food ... lYilO
Oorn-'hd m~ fiOWO\IIth ... Me~- of .-1:1 !YDO. melollorn..llotra:
-IliAD t\ f'IIWl.a or maJr,ant Mllili!IKOII'I'll8
0...1 mllilncc!lllt...,. Mltaalltlc at r.cumM'i millnoma
Spll<n~Wa "Splmld' mo~noma «'bordorlht' ~leoI
SjlltllumOf
Pilln- "'llndlo cd """"'"'Rood Splndlo..n m-•
-~··~
Splndlo..,ll melollorno
Hllo I'IM\Il Mo~-wllll-hOOI-n
Bab•ooiMYI.It ""~""""'will,,..,.., oon....,....
CU\pnllll
BMniMII
""""'will pdf'ntii'O mdu~(sJ ~~~~~- lflllnaln cmpnllll """''
,...~_will...,_
melanocytes may have a bland, nevoid appearance; have a size greater than
that of the basal keratinocytes (mild cytologic atypia); have increased size and
variable chromatin patterns (moderate cytologic atypia); or have increased
size, variable chromatin patterns, pleomorphism, and nucleoli (severe cyto-
logic atypia). A nevus with architectural disorder and severe cytologic atypia
should be considered a borderline melanocytic proliferation; a conservative
but complete excision should be encouraged (e-Fig. 40.15).
N. Nevi on special sites
1. Clinical: The clinical appearance varies with site. Special sites include umbili-
cus, acral skin (palms/soles), areola, and mucocutaneous sites such as the
conjunctiva, anus, and external genitalia.
2. Microscopic: In addition to the features associated with junctional and/or
compound nevi, the junctional melanocytic proliferations at these sites may
have an increase in the number of individual melanocytes with either a lentig-
inous or pagetoid pattern, loss of cohesion, and asymmetry.
0. Combined melanocytic nevus
1. Clinical: They vary in appearance. They may be flat, raised, or both and
may be variably pigmented. Although they can present at any age, they are
most common in the first three decades. The combination of features in these
lesions (asymmetry, color variations, and possible border irregularities) often
raises concern for melanoma.
2. Microscopic: Combined melanocytic nevi may have any of the features
of the previously described nevi in a side-by-side arrangement or top-to-
bottom arrangement. One of the more common combinations is a com-
pound melanocytic nevus and a blue nevus; the most worrisome combina-
tions include a compound melanocytic nevus with a deep penetrating nevus,
or a side-by-side Spitz nevus and compound nevus.
P. Recurrent melanocytic proliferations
1. Clinical: These are characterized by a sudden appearance or reappearance
of pigment in a scar from a previously biopsied or excised nevus and/or
melanoma. In some instances, the patient may not be able to provide reliable
information regarding the biologic nature and/or diagnosis of the previous
lesion.
2. Microscopic: Lentiginous melanocytic hyperplasia and randomly scattered,
irregular nests of melanocytes are present above an immature or mature
dermal scat. The features of a recurrent nevus may be indistinguishable from
a recurrent melanoma and have been referred to as "pseudomelanoma."
Adjacent lesional tissue outside of the scar and/or knowledge of the initial
biopsy or excision findings is imperative for arriving at the correct diagnosis
(e-Fig. 40.16).
Q. Nevus ambiguous. This category could include any/all melanocytic lesions that
have some of the classic features of a distinctive subtype (such as a Spitz nevus)
but are variant in others.
Ill. MELANOMA is a malignant melanocytic neoplasm that can occur in any tissue on any
body site. Ninety percent of all melanomas arise in the skin, and only cutaneous
melanoma will be considered in this section.
Melanoma accounts for only a small percentage (3% to 5%) of primary cuta-
neous malignancies; basal cell carcinoma and squamous cell carcinoma are far more
prevalent (95% to 97%). However, melanoma accounts for >50% of cancer deaths
from a primary cutaneous malignancy. The incidence of melanoma has increased
over the past 25 years, but the recognition and diagnosis of lower stage lesions
account for a substantial proportion of this increased incidence. As expected, the
overall survival has shown improvement as pathologically lower stage lesions are
diagnosed. Melanoma is most common in individuals >50 years of age but affects
persons of all ages, including infants. It is slightly more common in males, in whom
620 I SECTION VIII: SKIN
TO No"'"~"""""
...tlenoo d PlfiMJYtumor
n. Mel!no!M In o~u (dyplool '""~"""Y'!e tm>etplo&la,ll\&knocylle
T1
~ nd •n-........
Tumot :sl mm In thfc:tcnMs
rot loslon)
na: MlhoiA uwatm and m - <llmnr
n~~o- ulo!follmor mltoola ~llnm'
1\Jmor l.ol-UAO mm., 111~
T2t: MlhoiA uloetllllon
T2b:WJI!ul:er-
Tumor 2.01-4.00 mm In 111-
T.la: MlhoiA ulcl!fatm
lllb: vm. ul::allllm
1\Jmor >-1.00 IMlln 11\kicnOIIO
T4a: MlhoiA ulcotl1br
...,..,,.,.,.
r«
T4b:WJI!ul:er-
-~~~~
R...,.lljmjtl nodos..nnot bo "''"'"~
NO No roalohol b'ml>lr 1\«io r..-.
Nl ,.,_.In...., roalohol b'ml>lr node
Nit: Mao-mnk (d~ oflat .811111MIIympll nodo blo!>t;l)
Nib: Mlaom.WWII (dnlclllf dlltaclal>lund porth.,.,.,.,llf corrlll1nlll)
N2 Mil-In t.oo a Illroo • • " lymph ncdos
N21:Mao-•
Nil!: """""'"'_,II
~ SDite «ln-l!anolt rne-*Mihola 18111onolll0dol ~
.......,1... (10
mo-
Me-lnfolrcr """" ,...., tJmpll ncdlll, arnamd-
l'l!j!jonol ""'""node~,,_ ....-'""-...,.
In • • I I tjmpll nodll(l)
t.OC
MO
D1111trt-•..
NodiiWrt-oll
nllotbea-
Ml Dtawrt I'IWM"sk
Mia: Sllfn, tubc:Wn...,. U&sut, Of lymph ncdos bejmd 1111 noponol
tJmpnnodlll
Lu,.
Millo mart.-"""111
"'*"
Mlc: Ill cctrer W...10l o1teo. orony •~~~~t~•I<Mitad oorum
...""".......
SlqtO
Slqj:IIA na
~(IJ)Hl
lis NO
NO
MO
MO
Slqj:IIB nb NO MO
T2t NO MO
fllqo IIA T2b NO MO
13a NO MO
~)
Pdnlotfc 1'iiiMII Nocle, M111AW1l (TNM) Cllllltlcatfon ~
.........rrt ~,. CAl ct. 2010) (t:MI'IINI)
Slqo liB T.lb NO MO
T4a NO MO
5lqo I!C T4b NO MO
$QII!IA n-41. Nlo, 2a MO
Slqa 1118 n-41 Nlb,2b,:l<> MO
n-41> Nl•J 2a MO
Slqo IIIC n.-~~> Nlb,2b,:l<> MO
AnyT N3 MO
AnyT AnyN Ml
624 I SECTION VIII: SKIN
Central Nervous
System: Brain, Spinal
Cord, and Meninges
Richard J. Perrin, Sushama Patil, and Arie Perry
and is formed by the processes of neurons (axons and dendrites) and glial
cells. Neuronal morphology varies significantly, with cell body size ranging from
< 15 f.'ID (e.g., small neocortical granular stellate neurons) to 100 /A-ID (Betz cells of
the primary motor cortex). For descriptive purposes, neocortical pyramidal neu-
rons are often considered the morphologic prototype. These cells contain abun-
dant amphophilic cytoplasm, clumpy basophilic Nissl substance, a large round
central nucleus, a prominent nucleolus, coarse proximal cytoplasmic processes,
and a prominent apical dendrite oriented perpendicular to the cortical surface.
Ependymal glial cells form a ciliated cuboidal epithelium that lines the ventricles
and central canal and focally transitions with epithelium of the choroid plexus.
The choroid plexus, which produces cerebrospinal fluid (CSF) within the ven-
tricles, is papillary; its branching fibrovascular cores are lined by a specialized
epithelium with a hobnailed apical surface.
Ill. INTRAOPERATIVE EVALUATION, GROSS EXAMINATION, AND TISSUE SAMPLING. Eval-
uation of a surgical neuropathology specimen often begins with an intraoperative
consultation, which may be requested by the surgeon (1) to confirm the presence of
lesional tissue, (2) to provide a preliminary diagnosis that will guide surgical man-
agement (e.g., aggressive surgery for ependymoma, limited biopsy for lymphoma,
culture sample to microbiology for abscess), and (3) to sample fresh or frozen tis-
sue for ancillary studies (e.g., Western blot for Creutzfeldt-Jakob disease [CJD],
molecular pathology, tumor banking, karyotyping). For optimal evaluation, spec-
imens should ideally be submitted on Telfa nonstick gauze pads saturated with
normal saline. Tissue that has been soaked in saline is certainly acceptable, but is
more likely to fragment during transport and may demonstrate more severe freez-
ing artifacts. Fresh brain tissue, especially small biopsy specimens, should never
be placed on dry gauze or tissue paper, because subsequent tissue retrieval from
these materials is almost impossible. Water content may be reduced through very
gentle blotting on a clean dry plastic surface, but fresh brain tissue is very fragile,
and improper handling can introduce cellular "touch" and "crush" artifacts.
For intraoperative diagnosis, small portions of the fresh specimen should be
chosen for freezing, for cytologic "smear" or "touch" preparations, and for possi-
ble ultrastructural examination. Freezing or otherwise exhausting the entire spec-
imen for intraoperative diagnosis should be avoided, for several reasons. First,
the techniques available during intraoperative examination seldom yield suffi-
cient information for a definitive final diagnosis; most diagnoses require the fine
histologic detail afforded by paraffin sections and information from immunohis-
tochemical stains and other ancillary molecular tests. Second, freezing introduces
artifacts (e.g., ice crystals, clumping of nuclear chromatin). Third, on some occa-
sions, surgical attempts to obtain additional diagnostic material from the patient
cause bleeding or other complications that prevent further tissue acquisition.
The manner in which intraoperative specimens are processed for diagnosis is
important. Before any tissue is frozen, a block of embedding compound (com-
monly referred to as "OCT"), formed within an empty tissue well within the
cryostat, should be frozen fast to a cryostat "chuck." After freezing, the chuck
and OCT block are removed sharply and inverted; the tissue should be placed
centrally on the flat surface of the frozen OCT block, immediately covered with
a minimal amount of liquid OCT, and frozen from above by a flat, prechilled
metallic weight (e-Fig. 41.1)."' This process maximizes rate of freezing and min-
imizes (but does not eliminate) ice crystal formation. Cytologic evaluation by
smear preparation is extremely helpful because it lacks freezing artifacts and
preserves nuclear details. Smears can be prepared by gently compressing a very
small amount of representative tissue between two glass slides, gently sliding them
apart, and immediately fixing both smeared slides in 95% alcohol (e-Fig. 41.2).
Lastly, a small tissue fragment (1 mm3 ) should also be fixed in glutaraldehyde and
"'All e-figures are available online via the Solution Site Image Bank.
Chapter 41 • Central Nervous System: Brain, Spinal Cord, and Meninges I 6 27
"'
QBM(tnoo~)
..... !+)
QFAJ'
QFAJ', VIIL-40, •r-.&-100, CK
_ . . . •IISIIIrtC:I:I
lt'"OM_I'P"'_
. ...,...(-)
II)K.I, VIIL-40
II)K.I,CN.I!>:!
-....,_
DA ~100. QFJ.P.IDH-1* cot'>, LJ:ol.lm, Hlf8..4S
0 ............ S-100. Q-,IOK-1 lm- cot>. u:A
S-100. Q~. M<IO.""' CI)W lh•- -lh.ri~CI( LCA.II'III, II)K.I
~100. Cf(. V1Jf, hrwlh)• eth' QFN' ElfA.CEA
-·
-, plloul""""'
... ·~ tlltdi'O'I\a EMil""' CIC. CIO Uq). Clt!D-1. TlJl Uq). Co\1.15.2 CEA, £.141),l'IIC CFAP. LCA. HNB-4S,IDH-1
S-1110. -..s, -<>A UAART-11 QFAI\ Cl(, lCA.IIIH-1
llm_ ~ CO.!O(L2e). COI!Io ell, GFAI\ tee-45, S'lli, ooa
PI».---1
o.A. VIM, PR ', 8AF47 S-100. COM, W GFM.HMB-45
HPC Yllf, Clm,DC>;t, F_.XIII .. COM ElfA. C11. GFAP, £.141)
-...
,_,UI~ m,BAF47 £.141). Gf"AI' CIC.LCA.DIII
---......
AAf(l Yllf, EMil. Cll.-. r;m, GfAP.dMtnll\ MP
GIG S'rll,tiF,CG,Ci~ ell, £11111. !\AI'
Clrftii'JIU~ S'rll,f.iiii.HC G9.P,&ol00 OF, CG, Cll. LCA
S-leol,cm4. QoiiVI GFAI\....W ElfA. NF. CK
:rtli, CG.&-1(101 GlfN:CIC.-5
H8 S-100. - 111111011 GFN' ell, £1M
Qonnl"""" OCT4. 024)11,.,.. (COli])"', PIN' ,t.H.:IQ •• C« Afl\ ElfA. HM&.IS.I.CA
'M~bii'I'!Cf Afi\CK PIN:,..,. ~CQ.GfMOCT4.EifA
-
-"'"""" jl-hCQ,CIC.EMII lUI' Afl\ OCT4. QFN\ Hta«<i
EtntlrJ";:nf t:.ttr'lfilllt Cll. OCT4.1'W', C030 .-.-.a.~A. ~CQ. u:.l. Hta«<i
CII.EW. Afl\IU!' ~CQ.I>OI
_,01_._
« 4rl\el6 lb'\6fN!Udll'll ~ GSM. f>wt Mt lltii'MIJ <iSII.
6.Z 1'0100mrnol'lt:lll4, bOCtu.o ac; ~ln.IIM':3 ftht ~ ~ tt41Peq~st.thttJt:ll'IWI$.
"?J.""~"'::;,..,"'""':':,:,-::....- ......
=~~~- .,...tl11"10ttW'HO lmo~ we:aw t~~h WHOitfld$llfiM II.
...-·
~1'1'0 11\~w.rkrlt.
~ IM!tWn« ..,...,, MfdUI.Itllmli'IOI'MOit.'t$COII$.
'Jf::Oiw-t11>'1>',1il0!- • • - •
~.:;·tl ~ tof'JIN'lflO!T'IIt,.
lfiOSIMI: tl ~- . . .
_,....,,._....Of lim;-·-•__,_
0Wiefnllf'fii'IOI.$II!MIIm rrt~~il: t1 Of!lb!YONJ ~~~ w.ounor.
G~ fl ~~ llf'M"'th f. ttftftJOfV'JO(ItfiiCit,
GfiiiP. dll~totl ~r.; NF, 1'101.1/011Wirnont I)K.J,I$0CIII:Fft <1~,....,..1: 'tlQ.4), 01'111'\.tM>-~·mat: l;Q(. ~r.; ~.Z. M*l'fCJ\ ~ 11\tlllfCOII$; TTfl, ~
17'tMOrtl1»n1t*' J: ~ ~ OOI"M"''I'I Cit/tll!f!;$YN, ft'!'llli:IIO~: PZ« ~I'\; tli(A,.~~M~fltWitt VIM. fl'riOflftt P'Jit, ~~~«A
oti'OihOOm**~~ P\,.Y.!IIIIJ¢0/'MI~!r..JIM ~ M-P,«41*Croelr.; I~ ~hi.IMIJ\<tlO!'tOrliCrl>'*"'*'**; 8Af4J,fll$l)O*!tlef$11(ffi~hCIIH1l: eG, 01'!~;(011 rt,
~-PI:er.1N.SE.M~MOCMo:.
Chapter 41 • Central Nervous System: Brain, Spinal Cord, and Meninges I 629
difficult, cortical LBs are not argyrophilic. Fortunately, LBs all show
immunoreactivity for ubiquitin and a-synuclein (e-Fig. 41.8). These
lesions (along with similar inclusions [Lewy neurites] that appear within
cell processes) are seen in Lewy body disorders (e.g., Parkinson disease
[PD], and dementia with Lewy bodies [DLBs]).
c. Marinesco bodies are small, eosinophilic, strongly ubiquitin-positive
intranuclear inclusions located chiefly in pigmented brain stem neurons
(e-Fig. 41.9). These have no known pathologic significance.
d. Neurofibrillary tangles (NFTs} (e-Fig. 41.10) are argyrophilic intracyto-
plasmic filamentous aggregates of hyperphosphorylated tau protein.
Although characteristic of AD, they also appear in many other neurode-
generative disorders, and in rare GGs.
e. Hirano bodies are brightly eosinophilic rod-shaped or elliptical cytoplas-
mic inclusions that occur within the proximal dendrites of neurons, par-
ticularly in the hippocampus. Although not specific, they are particularly
numerous in brains with AD pathology (e-Fig. 41.11).
f. Granulovacuolar degeneration (GVD} is common in hippocampal pyrami-
dal neurons in AD, and less common in older brains without AD. GVD
resembles many small bubbles, each with a small basophilic granule.
g. TDP-43 neuronal cytoplasmic inclusions (NCis}, as the name suggests, are
immunoreactive for TDP-43. Although NCis are characteristic of a sub-
set of frontotemporal dementias, they are not specific, and may be seen
in the temporal lobe in other neurodegenerative diseases (e.g., AD).
h. Bunina bodies are eosinophilic ubiquitin and TDP-43 immunoreactive
intracytoplasmic inclusions that form in motor neurons in cases of famil-
ial and sporadic amyotrophic lateral sclerosis (ALS).
B. Astrocytes
1. Reactive astrocytosis. Normally, astrocytes are evenly dispersed (albeit with
regional variation), mitotically silent, and GFAP positive. In most forms
of brain injury, astrocytes become hypertrophic, increase their GFAP con-
tent, and may proliferate. Reactive astrocytes have prominent stellate pro-
cesses and, often, abundant eccentrically distributed glassy cytoplasm that
inspires the moniker "gemistocyte." Nevertheless, reactive astrocytes gen-
erally maintain an even distribution, and do not exhibit nuclear atypia
(radiation exposure is an exception). Grossly, tissues affected by chronic
astrocytosis are usually firm; thus, the term "gliotic" is used to describe
brain tissues that appear unusually firm or rubbery.
2. Bergmann gliosis refers to an accumulation of astrocytic nuclei (usually in
association with neuron loss) within the Purkinje cell layer of the cerebel-
lum.
3. Alzheimer type II astrocytes, with swollen pale nuclei and minimal visible
cytoplasm, appear in hyperammonemic states (e.g., liver failure, Wilson
disease). In the cortex and striatum, these nuclei are round with a prominent
nucleolus (e-Fig. 41.12); in the pallidum, dentate nucleus, and brainstem,
they are irregular and lobated.
4. Corpora amylacea are basophilic, round, concentrically lamellated aggre-
gates of polyglucosan (polyglucosan bodies) that develop within astrocytic
processes. Common in normal brains, particularly near ventricular and pial
surfaces, these become more numerous with age. Similar structures (Lafora
bodies) form in far greater numbers in astrocytes and neurons (and in eccrine
sweat glands) in Lafora body disease.
5. Rosenthal fibers (Rfs} are brightly eosinophilic, somewhat refractile, irregu-
lar/beaded structures that range from "'1 0 to 40 ~-tm in diameter. EM reveals
them as swollen astrocytic processes filled with electron-dense amorphous
granular material and glial filaments. RFs are commonly observed in PA, but
Chapter 41 • Central Nervous System: Brain, Spinal Cord, and Meninges I 6 33
within the rigid skull and dura. ICP increases when this balance is strained,
as may result from diffuse brain edema, increased cerebral blood flow and
blood volume, or development of space-occupying lesions (e.g., tumor, abscess,
hematoma, or large edematous infarct). Elevated ICP, if not treated, can
cause herniation; herniation syndromes include subfalcine (cingulate gyrus),
transtentorial (uncal), and cerebellar tonsillar/brainstem herniations.
G. Duret (secondary brain stem) hemorrhage occurs when penetrating pontine arter-
ies (which arise perpendicularly from the basilar artery) become kinked in asso-
ciation with brainstem herniation; the resulting acute hemorrhagic infarction
of the pons is often fatal.
V. NEOPLASMS OF THE CNS. For most CNS tumors, incidence varies greatly with age
and gender. Among adults, metastases, GBM, and meningioma are the most com-
mon CNS neoplasms; among children, PA, medulloblastoma, and ependymoma
are far more common. Likewise, tumors often differ in their radiographic features
and propensity for certain anatomic sites. For this reason, microscopic evaluation
of a CNS biopsy specimen is incomplete without considering the neuroradiologic
findings that describe the targeted lesion in situ. Indeed, neuroradiologic assess-
ment can be considered to be the neuropathology surrogate for gross examination.
Imaging studies are particularly helpful when evaluating small biopsy samples.
The World Health Organization (WHO) currently lists more than 100 types of
CNS tumors and their variants (Table 41.2). Table 41.3 organizes common CNS
tumor diagnoses on the basis of location, patient age, and imaging characteristics.
Histologic features are also critical for diagnosis. Because a final diagnosis may
not be obvious from an initial histomorphologic examination, it is worthwhile
to begin with a broad differential based on a specimen•s general histopathologic
pattern. Table 41.4lists eight major histopathologic patterns that may be encoun-
tered, along with their most commonly associated diagnostic entities. Once a
differential diagnosis has been formulated on the basis of these data, closer exam-
ination of microscopic details can refine the differential further and suggest what
ancillary tests (if any) are required to arrive at a final diagnosis.
A. Gliomas
1. Diffuse (infiltrating) astrocytomas (DAs), WHO grade II. Diffuse gliomas are
the most frequent primary CNS neoplasms. Because they are diffusely infil-
trative, complete resection is nearly impossible. On MRI, DAs are nonen-
hancing, T1 hypointense, T2/FLAIR hyperintense, ill-defined intra-axial
masses that can occur throughout the neuraxis, but commonly involve
the cerebral hemispheres. Clinically, they present with new-onset seizures
(the most common symptom), headaches, or functional neurologic deficits.
Grossly, these lesions may appear gray-tan to gelatinous and obscure the
native gray-white junction. Microscopically, tumor cells invade adjacent
cortex along white matter tracts. They aggregate around neurons and
blood vessels and beneath pial and ependymal surfaces to produce the
so-called secondary structures of Scherer. The cytologic features of astro-
cytic tumor cells can vary widely from uniform and minimally atypical,
to highly pleomorphic both in cytoplasmic and nuclear features (e-Fig.
41.15). Cells with elongate, irregular, hyperchromatic nuclei with minimal
cytoplasm are seen in fibrillary astrocytomas, whereas cells with eccentri-
cally located nuclei and abundant eosinophilic cytoplasm characterize the
gemistocytic variant. Tumor cells are often, but not invariably, immunore-
active to GFAP. A recently identified immunohistochemical marker,
antibody IDH-1, is proving invaluable for distinguishing the majority of
DAs (but not primary GBMs) from gliosis. Mitotic activity is very low or
nonexistent.
2. Anaplastic astrocytoma (AA}, WHO grade Ill, is a diffusely infiltrating glioma
with a mean age of presentation in the fifth decade. AA may appear
~41 • Central H~• ~' B111ril'l, ~iul Gelrd, tnd Nelli.._ I II&
,,(,I ]I (fill WltOClull'katlon _.Gfldlltlo!CiiSTu-
fl-41 •••III*Mfloll-
I I
•
AMI..:,IIc-.
•
""
PW.
S£Go\ •
•
PXA •
DA •
FI>. .IY~l
G~~""'
-plootnle l.llr'OeyiOn\1
M •
GBM •
Glontcdl GllM •
GS •
Obntmoll......ul •
Ot'ls fF"*'t1iii•IN'I
Ofplond!tVIoml •
Anlj>illllcolip>c!ondrqrfloma •
Ollflo••MJU~-
~ •
~~~~.~., ......
Anlj)illllc olipoi!Oqtllfl>•
S.llop!nll',ln'<ml •
•
httlooDOI>IiiiiY ~- •
~<t.'rrotmo •
Oollt.Mr •
Plpllo,y •
a.rcol •
Tllll'<¥io •
Ani!>IMdcGIIOn.tJrnoma •
ctn•~•--
Q\orold """""'""-.""'
•
•
Choroid """""co-a
Alypk:lldlo<dd """""'PI_,""'
OIM'tMIM!IifMIIIIVMn
•
AB
•
_,..,...,._...,._
CGa ollllal!llld ..-le
AGo
P<ltfl'
RONT of11lef»Lnh ¥011111cio
I
•
•
I
"'
F\orq:lll!jlome •
-llfllll .....l reaton
PI ._tome •
PPTID • •
PIIIOOblu1oma •
P'll'R • •
_,..,_,.,
111.._111. . .
•
Desmoplalllc:fnodu~r moc!LI"'**'"'• •
ModuloblulomllOIII _,.,. ncdulortty •
AnoDiuelc rnedui~IMtM>o •
LA1110 ooll rnoc!LIIo-rnl •
CNS PNET •
•
eNS ...,....,....._me
CNS ......- " " ' •
Moduloepllllalllma •
E'llonct!lno-11111 •
1(1/1'(1' •
r.nlfon.ill.n.,..,, .......
Sctlwlnl"'a"M (nl!lut1ilmoma~ IW.Irfnorr-.) •
Celll~r •
Pki:dfllnn •
Mellho1lc •
-broma •
Pladf'orm
l'<lrhN.rlom1,NOS • • •
Po-• Ontranauroll
Ma..,_llt 1>01tleurlomo
•
•
MPNST$ • •
Ej>llllolold MPNST • •
MPN&'T ..til n8Md!)mol dl!hr.mi81Son • •
• •
Moknollc:MPNST
MPNSTWIII .....,lu
,....llfllll...r.,..
dllo,_ • •
Menln&bna
Manln§'IMI~I •
•
--
Flbloua (ft-lullc)
Tronsttsonll (rniYdl •
•
Anatcrnn>ua •
Ml:to<Y* •
~ •
~phopl.uliiOC!I&olc:h •
-ploslSc •
Chonlold •
Clolrcd •
AlYI:bl •
(-
I I Ill
Pol>llo!Y •
Rhobdold •
Anll>lulle (mol"""'l!l) •
Mttt"'(;hyn\a\ l"d'\Men!~tuMM
u-
AnJioll-
Hlbcrnoma
U~R:Ome.
Fl>rorrootoolo
SfT
lnftomm-r mycftbrotllulletun>Or
FIH'OWtoml
Mllpnt nnuo hlllloq4oma
i.doiT(jOITII
Ldo-!1XItn0
R"'bdotn)<IM
R,.bdo,.,.."""""'
Q\ondnma
Chondrc&ln::ornl
Olbioorw
Ootoooo1<101no
OotoociiOIIdrotno
Ho1Nr9<>1N
Ejlllhotid MINrVoendath ......
HPC •
An11>iulle HPC •
Anatoooroorno
Klllpo~lureorrr:t
E!o1111 •rcornt~NET
Pmwy mollnoqtic llolons
Dllfuto~
Molancq1Dma
-,.,.,..,..
Mal...,nt melol\orno
HPC.ISFT
Soconduy l)mllflornlllouk.lmla
lntruellr Plluluy illlonoma
c.. nlopiiii)'I1JIIom,ll PI!Jitaly- ·
Rllllk.l., cloft"""
R-dalleyst
S.PI Ill ll!ypothllamtf Gormlnan.t!GCT Colokl cyst (3rd v.)
opl!o poii!M)folrd v. c.. nlopiiii)I1JIIom.<!
PA
PMA!El
PI,.., Oennllll:ii!IOIOOT P"-Y!>mo
Pln01l0$l1110 P.....l ey~t
Pln1111:1~
Plnlllqol
'!'tam~.~~; PA AJVGBM
AM:BM ~Ph'""'
c.r.bol\oj:ml!no ill\1!.1& ,_utor -nome (NF2) \l<llllbu~r -nome
Menln.,..,.
01n1:71J M\Jf~ C&t\trtlll n&U~
SEGA <m smA <m
C"'"'Id p-. pa1JIIoma
--
ChO!dd "'"""papilloma
ChO!dd "'""""""'""""' (lnfe.nt) S.bo""""'"'"me
~uroflbnma (NFl)
MPNST (NFI) Monln.....
SecolldOJY l)mllflorno
Nolroltbromo (NFI)
MPN&'T
ICO I SECTION IXt NERVOUS SVtHE ...
,,/,t,il(ft§l --HI~IeP&ItntllfiwllaiNIIII~
I. .........., ........ ~oolllll!lr
• Dlr1.110 aUome.
.,._,.
• ~s !lmpl>om•
• lnillmrndOiydem)oalhringd.....,
·-sbll
• Orpnlllng lnforet
ll.llocrolo - ( l i n !
• Mo!lo1 ...
• E'l*\<tlrnoma
• Sllb&pontt,ornana
• S£GA
• Cenb1J neu~m1
• PlnDOC)Ituna
• Mn-bi)ONIIInnor~.ATIRT)
•• Olor<id ~
CCM.towt.rnola•11~1
""*""'
• HB
•. lolll ... l l ' l - pn-
• PA
• PXA
• GBMI&$
• GG
• DNT
• Mn-bi)'MIIIllmor (6.(1., nw!ui-PNETI)
· ~--QI~Il
• OCT•
• Ol!Nollholl'IIPtna
• CNS!Jmph"""'
•Sa,.,..,.
• A- and-'"""'
f#.-llcoltlr.pniOII
d lof_,
• CNS !Jmphomo
• cympl>om-.. ..,-Ia
• ,..........,..... t,ompllorns
• v-.llla
• Maoln&~><n~Jorn-
• Ac:IJ1D d~rc onoapllllorTr,joii!IIADEMl
• ~ •llllloi>OI!tJ •lid iol>or hetnct~I
• ABRA
• Ano-.ol&
• CAOASIL
• Vlscullr mlillfwmlltlon
• l n f - ea.,..~
• ~urooorcoklooio
· ~boombolbd...._
~41 • Central H~• ~' 811!rit1, ~iul Gelrd, tnd Nelli.._ I 1•1
.,..,..,...
• S.reorn•
• SeNmOII"'''Il
• -nomalrna~I'IOC)iorne
• Sa<:oniii1Jf.lmplana/lau-lllpiosm'"'71onw!
• f'lll•nllolllll
• S.~nu..,..,....d--...
• r.-outii>•Y _..,..m .....
• ~ lll*'dolumor of ,.,lUll
• H~lt (O.JI', Roai-Ommen d - )
Vl.lhi..... II11Nt
• Mei'll,.... Cllleln011101ooh lor tl'lnllilomotMo. a!lolroltoolo, melollorn....,
....,...,,.._,£,)
+ Men!-
• San:ddOIIt/ii'IIIUiom_d_
• Oolllpn """"'"' d -
• lnflami!IIIQydhoocw <•·I'·
Cosll>mii1-IO)
W.l. . tiiMlc'*"oc.t
• C.robhllinfon:t
• Tumor- -lnllln••..rtntoffod
• lnllclon
• Vuculb
• Hypoltilllmlc hlnWiorna
· ~aloma
• Oorbl dylplulll/lubor
•· HelD...,.
-llanponl_. .
·---d-
• llldwnlcd-
• ~~~n-., <)l1b
•R_I_
• liojooijc ..,..p~~,ollpllhJ
• C.rob!Oiedema
• \'h! .... ...,hollll&
642 I SECTION IX: NERVOUS SYSTEM
cellular and populated by bipolar cells with long hair-like (piloid) pro-
cesses. RFs and EGBs commonly occur within the compact (dense)
regions and, although not specific for PA, provide very helpful diag-
nostic clues. Mitotic activity is low. The blood vessels within PAs are
commonly hyalinized, and often exhibit linear glomeruloid tufts of EH.
This feature, particularly when coupled with the marked nuclear atypia
that commonly occurs in PA, can lead to confusion with high-grade
glioma. However, the EH and nuclear atypia of PA have no prognostic
significance. PAs usually follow a benign clinical course.
b. Pilomyxoid astrocytoma (PMA}, WHO grade II, is a piloid neoplasm closely
related to PA that presents in infants at a median age of 10 months, and
favors the hypothalamus and optic chiasm. On MRI, PMA appears cir-
cumscribed and solid, hypointense on T1 and hyperintense on 1'2, and
shows homogenous contrast enhancement. The histologic hallmark of
PMA is the presence of monomorphous bipolar cells in a markedly
mucoid matrix, with a prominent angiocentric arrangement that resem-
bles the perivascular pseudorosettes of ependymoma (e-Fig. 41.20). As
defined, PMA does not show RFs or EGBs. Nuclear atypia is uncom-
mon. Mitoses can be present. Vascular proliferation resembling that in
PAis characteristic. Necrosis may be seen. The tumor cells show strong
and diffuse immunoreactivity for GFAP, S-100, and vimentin. A subset
of tumor cells is also positive for SYN. Clinically, PMAs behave more
aggressively than PAs, with frequent local recurrences and, often, CSF
seeding at the time of diagnosis.
6. Pleomorphic xanthoastrocytoma (PXA}, WHO grade II, is an epileptogenic
neoplasm commonly located in the superficial cortical regions of the tem-
poral lobe, often with meningeal attachment. Histologically, this tumor is
composed of large pleomorphic, variably GFAP-positive astrocytes inter-
mingled with less conspicuous spindled cells (e-Fig. 41.21). Some of the
tumor cells have bizarre nuclei and nuclear pseudoinclusions, and multi-
nucleated giant cells are common. EGBs are almost always evident. Xan-
thomatous tumor cells, although diagnostically helpful, only occur in 25%
of the cases. Other features include perivascular plasmalymphocytic cuff-
ing and scant mitoses. Reticulin silver stain highlights a network of basal
laminae that surrounds individual tumor cells. Neuronal differentiation
is often evidenced by reactivity for SYN and NF protein; reactivity for
CD34 is frequently observed. Grade II PXAs have a relatively favorable
prognosis (81% 5-year survival). Anaplastic (WHO grade III) transforma-
tion, characterized by less pleomorphism, increased proliferative activity,
necrosis, and/or microvascular proliferation, occurs in 15% of the PXAs;
reticulin staining is often diminished in these cases.
7. Subependymal giant cell astrocytoma (SEGA}, WHO grade I, is almost exclu-
sively found in children and young adults with tuberous sclerosis (TS).
These well-circumscribed, intraventricular tumors usually occur near the
foramen of Monro and cause symptoms associated with obstructive hydro-
cephalus. Imaging studies reveal contrast enhancement and calcification.
Histologically, the tumor cells may be spindled, epithelioid, or gemistocyte-
like, and may appear in sweeping fascicles, large clusters, or perivas-
cular pseudorosettes. The gemistocyte-like cells have abundant glassy
eosinophilic cytoplasm and nuclei resembling those of ganglion cells,
with fine granular chromatin and prominent nucleoli (e-Fig. 41.22). The
hybrid astrocytic and neuronal features of SEGAs are also reflected in their
immunohistochemical profile, with focal reactivity for both GFAP and one
or more neuronal markers (e.g., SYN). Mitoses are rare. Microvascular
proliferation and necrosis are typically absent.
644 I SECTION IX: NERVOUS SYSTEM
8. Oligodendrogliomas, WHO grade II, comprise 10% to 25% of all the adult
gliomas, behave less aggressively than astrocytomas, and show slower pro-
gression and longer patient survival. The most useful histologic feature
for distinguishing oligodendroglia! neoplasms from astrocytic tumors is
nuclear morphology; oligodendroglioma nuclei are nearly spherical, with
delicate chromatin, crisp envelopes, and inconspicuous nucleoli (e-Fig.
41.23). Other helpful features include dear perinuclear haloes that often
result as an artifact of routine histologic processing; a delicate hexag-
onal array of capillaries that resembles chicken wire; and mucin-rich
microcystic spaces, microcalcifications, predominantly cortical involve-
ment, and perineuronal satellitosis. Immunohistochemically, classic oligo-
dendroglioma cells show no reactivity for GFAP. However, oligoden-
drogliomas often contain three cell types that do show such reactivity,
specifically mini-gemistocytes, gliofibrillary oligodendrocytes, and reactive
astrocytes. The mini-gemistocyte, named for its superficial resemblance
to the larger astrocytic gemistocyte, has a belly of glassy, eosinophilic
cytoplasm and an eccentrically placed oligodendroglia! nucleus. Gliofibril-
lary oligodendrocytes are indistinguishable from classic oligodendroglia!
tumor cells on H&E stained sections, but show a GFAP-positive rim of
cytoplasm and a tadpole-like tail. Entrapped, nonneoplastic astrocytes
generally retain their characteristic stellate morphology.
In addition to oligodendrogliomas, several other tumors exhibit the
so-called fried egg appearance with rounded nuclei and dear perinuclear
haloes; most notably dysembryoplastic neuroepithelial tumor (DNT),
PA, central neurocytoma, dear cell ependymoma, and mixed oligoas-
trocytoma (MOA). Nevertheless, this differential diagnosis can usu-
ally be resolved using clinical information, other histologic clues (e.g.,
RFs, EGBs, atypical astrocytic nuclei), immunohistochemical stains (e.g.,
GFAP, Neu-N, SYN, NF, IDH-1, EMA, CD99, D2-40, MIB-1), and FISH
studies.
Ancillary genetic testing of oligodendrogliomas (Adv Anat Pathol.
2005;12:180) is now routinely performed. Several studies utilizing FISH
techniques have established the presence of 1p and 19q codeletions in
60% to 90% of the oligodendrogliomas (e-Fig. 41.24); these tumors
behave less aggressively, are more sensitive to chemotherapy and radia-
tion, and are thus considered genetically favorable. Apart from the obvi-
ous prognostic implications, identification of 1 p and 19q codeletions also
helps to differentiate oligodendroglioma-like mimics from true oligoden-
drogliomas. Unfortunately, pediatric oligodendrogliomas generally do not
show codeletion; those that do harbor the codeletion frequently occur in
teenagers and older children and likely represent the adult type oligoden-
droglioma(] Neuropathol Exp Neurol. 2003;62:53).
9. Anaplastic oligodendroglioma, WHO grade Ill. On the basis of WHO criteria,
anaplastic oligodendroglioma must have "significant mitotic activity, EH,
or necrosis" along with increased cellularity and marked atypia (WHO
Classification of Tumors of the Central Nervous System. Lyon, France:
IARC Press; 2007). High-grade oligodendrogliomas additionally often
show greater cytologic pleomorphism, epithelioid morphology, prominent
nucleoli, and sharper cell borders. Necrosis is more often infarct-like rather
than pseudopalisading. The exact number of mitoses required for diag-
nosis has not been established, but a mitotic index of six or more per
ten high-power fields (40x) is often used to assign anaplastic grade Ill
(based on J Neuropathol Exp Neurol. 2001 ;60:248) in the absence of EH
or necrosis. Anaplastic transformation can be focal or widespread. Some
oligodendrogliomas appear otherwise low grade, but contain hypercellu-
lar nodules with increased mitotic activity; such cases may be designated
Chapter 41 • Central Nervous System: Brain, Spinal Cord, and Meninges I 645
2. Anaplastic ependymoma, WHO grade Ill. Several grading systems have been
proposed for ependymomas, but no consensus has been achieved. According
to the WHO 2007 criteria, anaplastic ependymomas are characterized by
increased cellularity, brisk mitotic activity, pseudopalisading necrosis, and
microvascular proliferation. Necrosis by itself in the absence of pseudopal-
isading does not warrant the diagnosis of anaplasia, because lower-grade
ependymomas also can show degenerative changes.
3. Myxopapillary ependymoma, WHO grade I, is a slowly growing tumor of young
adults that occurs almost exclusively in the cauda equina region, where it
arises from the filum terminale. Grossly, these tumors are encapsulated,
intradural, sausage-shaped masses with a gelatinous interior. Histologi-
cally, the tumors show variable proportions of ependymal and papillary
patterns. In papillary areas, the tumor cells form irregularly ovoid rings
of cuboidal-to-columnar epithelium, each ring formed around a hyalinized
central vessel, some fibrous stroma, and a rim of pale basophilic mucin.
Pale basophilic often bubbly mucinous (myxoid) material also separates the
papillae. In ependymal areas, the tumor cells are spindled and slendet; and
are arranged in loose fascicles and rather extravagant perivascular pseu-
dorosettes (e-Fig. 41.26). The tumor cells are immunoreactive for GFAP,
S-100 protein, and vimentin, with variable staining for EMA, CD99, and
podoplanin (antibody D2-40). These tumors have a favorable prognosis fol-
lowing complete resection. If untreated or incompletely resected, rare cases
can progress and invade perispinal tissues. Occasional ependymal tumors
in this location show a mixture of myxopapillary and typical ependymoma
features; the clinical behavior of such tumors is incompletely understood.
4. Subependymoma, WHO grade I, is a benign, slowly growing, solid tumor
related to ependymoma. They are often asymptomatic, discovered inciden-
tally on CT and MRI studies performed for other reasons. Occasionally,
they undergo intratumoral hemorrhage and become life threatening by
exerting mass effect. Prognosis is excellent following resection. Grossly,
they are sessile or pedunculated masses within the ventricles (lateral >
fourth > third). Histologically, they are lobulated and well demarcated.
The tumor nuclei resemble those of ependymoma and appear in irregu-
lar clusters within a dense fibrillar background formed by tumor cell pro-
cesses (e-Fig. 41.27). Mitotic figures are rare. Occasional pseudorosettes
are not unusual. Secondary degenerative changes include microcyst forma-
tion, hemosiderin deposition, vascular hyalinization, myxoid change, and
calcification. Nuclear pleomorphism and microvascular proliferation are
occasionally seen; these do not warrant a higher-grade diagnosis. It is worth
noting that subependymoma may appear as part of a compound ependymal
tumor; in such cases, tumor grading should be assigned according to the
ependymal component.
C. Choroid plexus tumors
1. Choroid plexus papilloma, WHO grade I (e-Fig. 41.28), is a benign lesion com-
monly located in the lateral ventricles in children, and in the fourth ventricle
in adults. It typically presents with symptoms associated with obstructive
hydrocephalus. Histologically, choroid plexus papilloma resembles normal
choroid plexus. Howevet; its fibrovascular cores are lined by a cuboidal-
to-columnar epithelium that lacks the superficial intercellular spaces that
impart a cobblestone appearance to normal choroid plexus. Mitotic activity
is low. Clear cytoplasmic vacuoles are noted in some cases. Occasionally,
infarct-like necrosis may be present, but has no prognostic significance.
Focal ependymal differentiation is common. Tumor cells show immunore-
activity for S-100, CAM 5.2, transthyretin, and GFAP (focally) but not for
EMA and carcinoembryonic antigen (CEA).
Chapter 41 • Central Nervous System: Brain, Spinal Cord, and Meninges I 64 7
2. Choroid plexus carcinoma, WHO grade Ill (e-Fig. 41.29), usually presents in
children <3 years of age as an enhancing, intraventricular mass. These
tumors are highly aggressive, often metastasize via the CSF, and are nearly
uniformly fatal. Histologically, the tumors are more solid and complex
than papillomas. High-grade cytology and frequent mitoses are the rule.
Foci of necrosis are characteristic, and microvascular proliferation may be
seen. Focal small cell features may mimic embryonal tumors (e.g., PNET).
Immunoreactivity for S-100, CAM 5.2, and transthyretin is characteristic; is
variable for GFAP; and is not typically observed for EMA and CEA. The dif-
ferential diagnosis includes anaplastic ependymoma (GFAP+, CAM 5.2-,
EMA+/-), GBM (GFAP+, CAM 5.2-), medulloblastoma/PNET (SYN+,
CAM 5.2- ), and metastatic carcinoma (in older patients, EMA+, CEA+/-,
S-100 +1-, CAM 5.2+).
D. Other glial neoplasms.
1. Angiocentric glioma {AG}, WHO grade I (e-Fig. 41.30). First reported in 2005,
AG is a rare supratentorial tumor associated with epilepsy in children and
young adults. Radiographically, AG appears as a T2/FLAIR hyperintense,
nonenhancing mass that expands the cortex with minimal mass effect. In
some cases, a projection of the tumor extends to the ventricle. Histologically,
AG shows uniform, slender spindled glial cells, often arranged either radi-
ally from or parallel to vessels, and, occasionally, perpendicular to the pia.
AG shows some ependymal features, including dot-like immunoreactivity
for EMA, and microvilli and zipper-like junctions by EM. The proliferation
index is low.
2. Astroblastoma {AB}, not yet graded {e-Fig. 41.31 ). As with AG, AB is asso-
ciated with epilepsy in children and young adults, but AB is also associ-
ated with headaches and vomiting, and shows a female predominance. On
MRI, AB is usually large, lobulated, well demarcated, solid with an occa-
sional cystic component, T2-isointense with gray matte.t; and enhancing.
Histologically, AB bears some resemblance to ependymoma, but has stout
(rather than fibrillar) cell processes that form the defining astroblastic pseu-
dorosettes. Vascular hyalinization is usually robust; in some tumors, this
architectural pattern assumes a papillary appearance. Like ependymoma
and AG, AB shows dot-like immunoreactivity for EMA; reactivity for GFAP
highlights the abbreviated cell processes within pseudorosettes. ABs show-
ing >5 mitoses per ten 40x fields, anaplastic nuclear features, increased cel-
lularity, microvascular proliferation, and pseudopalisading necrosis are con-
sidered anaplastidmalignant, but may still be resectable due to this tumor's
noninfiltrating growth pattern.
3. Chordoid glioma of the third ventricle {CG}, WHO grade II, is a rare tumor of
adults that occurs almost exclusively in the vicinity of the anterior third ven-
tricle and hypothalamus. Radiographically, CG is solid, well demarcated,
T2 intense, and strongly contrast enhancing. Histologically, as its name
suggests, CG focally resembles chordoma, showing cords and dusters of
eosinophilic epithelioid cells within a bubbly basophilic mucinous matrix;
where mucin is lacking, the cells (strongly GFAP positive, modestly reac-
tive for EMA) appear in sheets. Lymphoplasmacytic infiltrates are typically
seen. Immunoreactivity for GFAP (and absence of whorls, psammoma bod-
ies, and physaliferous cells) distinguishes CG from chordoid meningioma
and chordoma.
E. Neuronal and glioneuronal neoplasms
1. Ganglion cell tumor {GG and gangliocytoma}, WHO grade I. Ganglion cell
tumors are epileptogenic and appear most often in the temporal lobes. On
imaging, they commonly enhance and appear solid, cystic, or both. A cyst
with an enhancing mural nodule is a common (but not specific) pattern.
Microscopically, they are usually cortically based, microcystic, variably
648 I SECTION IX: NERVOUS SYSTEM
fibrotic, and calcified, and they often show perivascular lymphocytic cuffing
(e-Fig. 41.32). Dysmorphic neurons (that may exhibit cytomegaly, vac-
uolated cytoplasm, coarse Nissl substance, irregular multipolar processes,
and/or nuclear abnormalities including binucleation) are the defining
feature. Architecturally, the ganglion cells are clumped or haphazardly
arranged in comparison with the laminar well-ordered arrangement of the
normal cortex. GGs have a variable glial component, typically astrocytic;
glial predominant GG may resemble DA or PA. Other features commonly
noted include EGBs and RFs (although the latter is more common at the
tumor periphery, and may result from piloid gliosis). Cortical dysplasia may
be seen adjacent to GG. High-grade glial transformation is exceedingly rare
and difficult to define. The glial component is immunoreactive for GFAP;
the neuronal component is variably immunoreactive for SYN, NF, and chro-
mogranin, but is usually immunonegative or minimally positive for Neu-N.
A subset of cells both within and adjacent to the tumor often shows
immunoreactivity for CD34; these CD34+ cells are "spider-like,, charac-
terized by long, stellate, ramified processes, and are thought to represent
progenitor cells. GGs have a favorable prognosis after surgical resection.
2. Desmoplastic infantile astrocytoma and desmoplastic infantile ganglioglioma
(DIAIDIG), WHO grade I (e-Fig. 41.33), typically occur in children younger
than 2 years, and are located superficially within the frontoparietal region.
Radiologically, they are very large, attached to the dura, brightly enhanc-
ing, and are associated with a very large, occasionally multiloculated cyst.
Histologically, the tumors are biphasic. In the collagen N- and reticulin-rich
phase, the tumor cells are arranged in a storiform or fascicular pattern. The
astrocytic cells, spindled and gemistocytic, are often inconspicuous within
the desmoplastic background, but can be identified by GFAP immunos-
taining. The neuronal component (present in DIG, absent inDIA) is also
subtle because the polygonal neuronal tumor cells are often considerably
smaller than those of conventional GG. The other component of the tumor
resembles PNET, with abundant small cells and many mitotic figures, and
lacks reticulin and collagen. EH and necrosis may be observed but are not
associated with an unfavorable prognosis.
3. Dysplastic cerebellar gangliocytoma (DCG} (Lhermitte-Duclos disease}, WHO
grade I. This unique cerebellar neoplasm is often associated with Cowden
syndrome (autosomal dominant PTEN/MMAC-1 mutation). Patients with
DCG often present with cerebellar dysfunction and/or obstructive hydro-
cephalus. DCG, which is Tl hypointense and T2 hyperintense, has a char-
acteristic striped appearance with both modalities. Microscopically, DCG
presents as a unilateral expansion of cerebellar folia wherein the internal
granular layer is replaced by dysmorphic ganglion cells; there is no glial
component. Abnormal vascular proliferation is sometimes noted, and white
matter is occasionally vacuolated. Evaluation for other features of Cowden
syndrome (breast and gastrointestinal lesions) is warranted in patients with
DCG.
4. Central neurocytoma, WHO grade II (e-Fig. 41.34), occurs in the lateral ven-
tricles near the foramen of Monro in young or middle-aged patients with
obstructive hydrocephalus. On imaging studies, these masses are large, glob-
ular, enhancing, and often calcified. Histologically, the tumor cells have a
uniform appearance with round nuclei, finely granular chromatin, incon-
spicuous nucleoli, sparse eosinophilic or clear cytoplasm, and delicate fib-
rillar cell processes (in contrast to the coarse processes of glial tumors).
The cells often appear in solid monotonous sheets that may also exhibit
prominent chicken-wire vasculature, and/or neurocytic (Homer-Wright)
rosettes, and/or perivascular pseudorosettes, and/or calcifications. Thus, the
Chapter 41 • Central Nervous System: Brain, Spinal Cord, and Meninges I 64 9
nuclei and a small amount of cleared cytoplasm; classically, they form sin-
gle or clustered rosettes and pseudorosettes within a relatively hypocellular
eosinophilic matrix of astrocytic tumor cell processes. The centers of these
rosettes and pseudorosettes are more intensely eosinophilic and contain
fine, SYN -immunoreactive cell processes. The surrounding matrix shows
immunoreactivity for GFAP, and may exhibit EGBs and RFs. Surgical resec-
tion is curative.
F. Pineal parenchymal tumors
1. Pineocytoma, WHO grade I, is more common in adults. On MRI, pineo-
cytoma appears as a Tt hypo- or isointense, T2 hyperintense, uniformly
enhancing, often calcified, discrete mass in the region of the pineal gland.
Through mass effect, pineocytoma often causes increased ICP and upward
gaze palsy (Parinaud syndrome). Histologically, pineocytoma tumor cells
(SYN+, Neu-N+) have round-to-oval nuclei, salt and pepper chromatin,
inconspicuous nucleoli, and poorly demarcated cell borders, and often
form pineocytic rosettes (large, exaggerated Homer-Wright rosettes) (e-Fig.
41.38). Mitotic activity is very low. Pineocytomas may be difficult to dis-
tinguish from normal pineal tissue in a small biopsy, although a lobular
pattern with gliovascular septae favors the latter.
2. Pineoblastoma, WHO grade IV, is a rapidly growing malignant tumor that
predominantly occurs in children and very young adults. Occasionally,
pineoblastomas occur in association with bilateral retinoblastoma (trilat-
eral retinoblastoma). On MRI, pineoblastomas are inconsistent: hypo- to
isointense on Tt, hypo- to hyperintense on T2, with homogeneous or het-
erogeneous enhancement. Their borders appear more infiltrative than those
of pineocytoma, and leptomeningeal metastasis (through CSF dissemina-
tion) is common at presentation. Grossly, they are soft, friable, and poorly
demarcated. Hemorrhage and necrosis may be present. Calcification is rare.
Histologically, they resemble other small blue cell tumors (e.g., medul-
loblastoma, PNET). The primitive-appearing cells have little cytoplasm,
hyperchromatic molded nuclei, and are densely packed in sheets occasion-
ally interrupted by Homer-Wright rosettes, Flexner-Wintersteiner rosettes
(which have a central lumen), and zones of necrosis. Mitotic activity is
generally high. Invasion of the adjacent pineal gland and leptomeninges is
common. Immunoreactivity for neuronal markers (e.g., NF, SYN, chromo-
granin, and neuron-specific enolase) is weak and focal relative to that seen
in pineocytoma. Occasionally, these tumor cells may additionally express
antigens related to photoreceptor or pineal differentiation, including retinal
S-antigen, rhodopsin, and melatonin.
3. Pineal parenchymal tumor of intermediate differentiation (PPTID), WHO grade
11-111, is a recently recognized entity that occurs more commonly in adults.
Histologically, PPTID is rather cellulat; lacks pineocytic rosettes, and has
slightly more pronounced cytologic atypia and a higher mitotidproliferative
rate than pineocytoma (MIB-1 indices from 10% to 16% for PPTID vs. 1%
to 2% for pineocytoma). PPTID appears in two main patterns: diffuse, with
mildly irregular round-to-oval nuclei arranged in a sheet-like pattern, and
pseudolobulated, wherein tumor cells are divided into lobules delineated
by vessels. A higher grade (Ill) is assigned to PPTID when the mitotic index
exceeds six mitoses per ten 40 x fields or when immunoreactivity for NF
protein is nearly absent; for this reason, definitive grading is best reserved
for resection rather than limited biopsy specimens. Recurrence and survival
rates are intermediate between those of pineoblastoma and pineocytoma.
4. Papillary tumor of the pineal region (PTPR), WHO grade II or Ill, has been
reported in children and adults, with a peak in the third decade. MRI shows
a large, well-circumscribed, T2-intense, enhancing (occasionally cystic)
Chapter 41 • Central Nervous System: Brain, Spinal Cord, and Meninges I 6 51
mass in the vicinity of the pineal gland. Histologically, PTPR exhibits papil-
lary features; large, pale-to-eosinophilic tumor cells form columnar epithelia
around hyalinized vessels (e-Fig. 41.39). In other, more solid areas, tumor
cells with round-to-oval nuclei and clear or vacuolated cytoplasm may form
true ependymal rosettes. Mitotic activity is variable (0 to 10 per 10 high-
power fields), and necrosis may be seen; EH is usually not observed. PTPR
does not infiltrate the pineal gland.
This histologic appearance resembles that of ependymoma. Further,
both tumors can show focal membranous and dot-like reactivity for EMA
(consistent with the fact that PTPR is thought to arise from the specialized
ependymal cells of the subcommissural organ). The key features that dis-
tinguish PTPR are minimal reactivity for GFAP and strong reactivity for
CK18. Because PTPR has only recently been described, official grading cri-
teria have not yet been defined; howeve.t; a mitotic index of five or more per
ten 40x fields has been associated with poorer prognosis.
G. Embryonal tumors, most common in children, are so-named because their "small
blue cell" histology is reminiscent of elements of the embryonic nervous sys-
tem. These tumors are bulky, grow rapidly, and seed the CSF pathways. All
embryonal tumors are inherently malignant (WHO grade N); howeve.t; the
prognoses for these tumors vary widely, as some respond very well to current
treatments.
1. Medulloblastoma, WHO grade IV, the most common embryonal CNS tumo.t;
is thought to originate either from remnants of the external granular layer
or from the fourth ventricular germinal matrix. On imaging, they are often
T1 hypointense, T2 hyperintense, noncalcified, homogeneously contrast-
enhancing cerebellar masses with restricted diffusion. In approximately
one-third of the classic cases, the tumor has already seeded the CSF path-
ways focally (with so-called drop metastases in the lumbosacral spinal cord)
or diffusely (with so-called icing of the subarachnoid space) at the time
of presentation. Distant metastases, most often involving bone and lymph
nodes, are rare. Several histologic subtypes of medulloblastoma have been
described including classic/undifferentiated, desmoplastic/nodula.t; "with
extensive nodularity," large cell, and anaplastic.
Classidundifferentiated medulloblastoma (e-Fig. 41.40), the most com-
mon subtype, appears as a patternless sheet of abundant small tumor cells
with hyperchromatic nuclei and minimal apparent cytoplasm. The nuclei
show a degree of molding and are often described as round-to-oval or
carrot-shaped. Nucleoli are inconspicuous. Mitotic figures and apoptotic
bodies are often abundant. Microvascular proliferation and necrosis may
be present, but are not prominent. About 40% of the cases exhibit Homer-
Wright rosettes. In addition to seeding the CSF, this tumor often reinvades
the cerebellar parenchyma from the subarachnoid space via Vrrchow-Robin
spaces.
The desmoplastidnodular variant (e-Fig. 41.41) has a characteristic low-
power appearance of rounded, pale islands separated by dark internodu-
lar tissue. The pale islands (reticulin free) contain cells with a relatively
neurocytic phenotype that features round to oval nuclei, open chromatin,
moderate cytoplasm, cell processes that form a neuropil-like stroma, and
little mitotic activity. The internodular tissue (reticulin rich) is formed by
densely packed, primitive-appearing cells with hyperchromatic malleable
nuclei, smudged chromatin, little cytoplasm, and high proliferative activ-
ity. This variant is genetically distinct and has a slightly more favorable
prognosis than other subtypes (Acta Neuropathol. 2006;112:5).
Another prognostically favorable variant is medulloblastoma with
extensive nodularity. This rare tumo.t; found almost exclusively in infants,
652 I SECTION IX: NERVOUS SYSTEM
remission, is more common in young adults (women > men with a ratio of
2:1) and is themostcommonformin the United States (""80% of cases). Less
common forms involve rapid, unremitting clinical deterioration-either de
novo (primary progressive MS [PPMS]) or in the setting of RRMS (sec-
ondary progressive MS [SPMS]). Demyelinative MS plaques in these sub-
types most often involve white matter periventricularly, and in the optic
pathway, brain stem, and/or spinal cord. Radiographically, chronic inac-
tive plaques are hypointense on T1 and diffusion-weighted images; plaques
with active inflammation are hyperintense on T2 and show postcontrast
enhancement. This latter feature can be responsible for some radiolog-
ical diagnostic uncertainty in cases of Marburg disease or acute tume-
factive MS. In acute tumefactive MS, a large isolated plaque appears as
a T2-hyperintense, peripherally enhancing lesion that resembles a high-
grade glioma or lymphoma. Although the rim of enhancement around
the demyelinative lesion may be incomplete at the cortical side (forming
a horseshoe-shaped profile that would be highly unusual in a neoplasm),
this feature is not robust enough for conclusive diagnosis; consequently,
such lesions, although uncommon, are often biopsied. At frozen section,
tumefactive MS can represent a great diagnostic challenge; in such cases,
examination of a cytologic smear preparation is often very helpful as it
preserves important cellular details that can be obscured by freezing arti-
fact. Microscopically, an active demyelinative MS plaque shows (1) loss
of myelin, (2) relative preservation of axons, (3) perivascular nonneoplas-
tic lymphocytic infiltrates, (4) numerous foamy macrophages, (5) reactive
astrocytosis, and (6) cerebral edema. Mitotic activity among astrocytes and
macrophages may be brisk, but nuclear atypia should be minimal. Gran-
ular mitoses and Creutzfeldt cells (which contain multiple micronuclei),
although not specific for MS lesions, are characteristic. Myelin loss with
axonal sparing (which distinguishes this lesion from an axon-destroying
infarct) is best visualized by comparing Luxol fast blue-PAS (LFB-PAS)
stained sections to others stained with Bielschowsky silver or NF protein
immunohistochemistry (e-Fig. 41.60). On LFB-PAS sections, the border of
demyelination is usually abrupt, and blue-green debris (representing par-
tially metabolized myelin) is visible within macrophages. In rare cases that
cannot be satisfactorily diagnosed with special stains, FISH studies can be
applied to evaluate for glioma-associated chromosomal changes.
2. Acute disseminated encephalomyelitis (ADEM}Iperivenous encephalomye-
litis represents an unusual autoimmune response among children and young
adults that is induced by recent infection by measles (most commonly,
but also), mumps, varicella, influenza, rubella, Campylobacter jejuni, or
Mycoplasma pneumoniae or vaccination (smallpox or rabies). Symptoms
include headache, fever and vomiting, followed by weakness, ataxia, and
visual and sensory loss with progression to stupor and seizures. Radio-
logically, ADEM usually appears as many small T2-weighted and FLAIR
hyperintensities in subcortical and periventricular white matter and the
spinal cord; the deep gray matter and cortex may also be affected. This
clinicoradiographic pattern is characteristic enough that biopsy is not
always performed. Nevertheless, when obtained, biopsy material shows
perivenous demyelination with axonal sparing, mononuclear infiltrates,
macrophages, activated microglia, and occasional petechial hemorrhages.
ADEM is thought to result from a myelin-directed, cross-reactive immune
response to a foreign antigen.
3. Demyelinating viral infections. Viral infections that commonly cause CNS
demyelination include human immunodeficiency virus (HIV leukoen-
cephalopathy and vacuolar myelopathy [HIVL, HIVVM]), human
T-celllymphotropic virus-1 (HTLV-associated myelopathy/tropical spastic
662 I SECTION IX: NERVOUS SYSTEM
presents in the fihh or sixth decade of life with classic migraine, multiple sub-
cortical ischemic strokes, psychiatric disturbance, and later, dementia. In this
context, T2 hyperintensities in anterior temporal lobes and external capsules on
MRI are strongly suggestive. Although the symptoms of CADASIL arise from
the brain, the disease also affects peripheral tissues including the skin. His-
tologic and ultrastructural examination of arterioles in the skin or brain pro-
vides the diagnosis. Notably, a rare, otherwise identical clinical/radiologic syn-
drome with an autosomal recessive inheritance pattern (appropriately named
CARASIL, linked to the HTRA1 gene) lacks granular osmiophilic inclusions.
E. Cerebral infarcts. Most often, biopsies of infarcts are performed when "glioma"
is in the differential diagnosis, particularly in young patients. Gross and micro-
scopic features of a cerebral infarct change over time. In the acute stage
(1 to 3 days), red necrotic neurons, vacuolated neuropil, and variable neu-
trophilic infiltrates are present. Subacute infarcts (days to weeks) demon-
strate capillary proliferation with prominent endothelial cells, numerous foamy
macrophages, and reactive astrocytes that are typically more common at the
periphery. Because such reparative lesions may exhibit mitotic figures, necro-
sis, and microvascular proliferation consistent with GBM, care must be taken
to recognize the reactive and histiocytic components, particularly during an
intraoperative microscopic evaluation. Large infarcts eventually evolve into
cystic lesions with a weblike network of delicate blood vessels, sparse gliotic
parenchymal remnants, and a few residual macrophages. Infarcts disrupt and
destroy axons, a feature often helpful in distinguishing infarcts from demyeli-
native processes such as MS in which the axons are relatively spared. Once an
infarct is diagnosed, identifying its cause is the next step; if the specimen at
hand provides no insight, clinical correlation is required.
IX. NEURODEGENERATIVE DISORDERS. Although some of these affect children and ado-
lescents, most come to clinical attention after the fifth or sixth decades. An increas-
ing number of neurodegenerative diseases can be diagnosed by identifying changes
in DNA, biologic fluids, peripheral biopsy material, or radiographic images. Nev-
ertheless, few treatments are currently available in clinic for neurodegenerative
disorders, and most of them remain idiopathic, progressive, and fatal. Conse-
quently, CNS biopsy performed explicitly to diagnose a neurodegenerative disease
is rare. These disorders usually appear in a surgical pathology specimen inciden-
tally (e.g., when some brain is removed to evacuate a life-threatening hematoma)
or when unusual clinical/radiologic data suggest that securing a definitive tissue
diagnosis warrants the risk of brain biopsy. Recognizing these disorders in surgical
specimens may become more critical as treatments become available.
A. Alzheimer disease (AD}, the most common neurodegenerative disorder, is char-
acterized histologically by neuron loss, gliosis, and the presence of extracellular
beta-amyloid peptide deposits (diffuse, cored, and neuritic plaques) and NFTs
(NFT, intraneuronal aggregates of hyperphosphorylated tau protein) in the
neocortex and hippocampus (e-Fig. 41.73). Plaque deposition begins 10 to
15 years before the onset of very mild dementia of the Alzheimer type (DAT,
the appropriate term for the clinical manifestations of AD). The degree of
cognitive impairment appears to correlate with the severity and extent of NFT
pathology, which begins in the mesial temporal lobes and subsequently spreads
outward to involve the parietal and frontal lobes; the occipital lobe is relatively
spared. Neuroimaging techniques (to assess atrophy, metabolism, and amyloid
burden within the brain) and CSF biomarker measurements (e.g., of amyloid-
beta peptide and tau protein) are likely to allow reasonable antemortem con-
firmation of AD pathology in the near future (Nature 2009;461:916-922), but
recognition of the histologic changes will remain important.
Currently, there are three leading sets of criteria for the neuropatho-
logic diagnosis of AD: Khachaturian, Consortium to Establish a Registry for
670 I SECTION IX: NERVOUS SYSTEM
biopsy specimen. It is also worth noting that biopsies of individuals with CJD
may also show preexisting histologic changes associated with other dementing
illnesses, therefore, identification of abundant amyloid plaques, NFTs, or LBs
does not rule out the possibility of CjD. For this reason, immunoblotting and
immunohistochemical analysis for the abnormal protease-resistant protein are
essential for definitive diagnosis.
D. New variant CJD (nvCJD) is a rare form of TSE linked to bovine spongiform
encephalopathy (BSE). Unlike CJD, nvCJD affects a younger demographic
(adults younger than 40 years), typically has a longer clinical course charac-
terized by behavioral changes more often than dementia, and shows an addi-
tional thalamic abnormality on DWI, T2, and FLAIR MRI. Microscopically,
the most characteristic feature is the presence of numerous amyloid plaques
(the so-called florid plaques that are PRP-positive, amyloid-beta negative) in
the cerebral and cerebellar cortices. The few hundred cases reported since 1996
have been clustered in the UK and France and are thought to stem from an
outbreak of BSE that affected the UK cattle industry in 1986. Perhaps reflecting
the incubation period and changes in cattle industry practices, the incidence of
nvCJD in the UK has dwindled over the last decade, so the diagnosis is now
quite rare.
I. SPECIMEN TYPES. The most common specimen type obtained for cytologic evalua-
tion of pathologic conditions of the central nervous system (CNS) is cerebrospinal
fluid. Rarely, stereotactic fine needle aspiration (FNA) of cysts or masses of the CNS
is used to provide specimens for cytologic diagnosis.
A. Cerebrospinal fluid (CSF}. Several methods have been developed in the last decade
for concentration of samples obtained via lumbar puncture, including cytocen-
trifugation and membrane filtration. Slides prepared via the cytocentrifugation
technique using a cytofunnel (http://www.shandon.com) are usually Diff-Quik
stained since leukemic involvement of the CSF is the most common malignant
diagnosis; the limited amount of material obtained (0.5 to 1.0 ml) usually pre-
dudes allocating material for Papanicolaou and other stains. Although the precise
proportions vary by practice setting, most CSF specimens obtained for headaches
and mental status changes show reactive (e-Fig. 41.75) or nonspecific features,
and usually contain benign lymphocytes andmonocytes (e-Fig. 41.76). Other nor-
mal cells which can be found in CSF specimens include ependymal and choroidal
cells (e-Fig. 41.77).
1. Traumatic tap. CSF samples that consist predominantly of peripheral blood are
diagnosed as "negative for malignant cells" with the caveat that the sample
may represent a traumatic tap (which occurs in about one-quarter of the cases).
When a CSF sample shows leukemic involvement in the presence of peripheral
blood, the possibility of CSF sample contamination by peripheral blood blasts
should be raised and a repeat sample should be obtained.
2. Infectious processes. Peripheral blood contamination can result in the presence
of variable numbers of polymorphonuclear neutrophils in a CSF sample; how-
ever, abundant neutrophils in the absence of blood contamination should raise
the possibility of acute meningitis. Prompt diagnosis is crucial, since it can be
fatal if not treated promptly (e-Fig. 41.78). Aseptic meningitis gives a picture
672 I SECTION IX: NERVOUS SYSTEM
Successful use of nerve biopsy requires a discussion between the clinician and neu-
ropathologist since the clinical differential diagnosis may dictate an unusual sampling
scheme, for example, vasculitis in which multiple cross-sections may be required for
diagnosis.
I. NORMAL PERIPHERAL NERVE AND METHODS OF ANALYSIS. The sural nerve, a cuta-
neous sensory nerve to the lateral foot, is typically biopsied without stretching
or use of intraneural anesthetic. The nerve is divided into a portion for fixation
in formalin for paraffin embedding, and a portion for fixation in glutaraldehyde
for plastic sections and possible electron microscopy. The entire nerve is typically
used rather than dissected into individual fascicles. The portion {"'1 em) of the
nerve fixed in formalin should be cut into 3 mm segments and examined in cross-
section with paraffin-embedded H&E sections, thioflavin-S histochemistry, and
possible immunohistochemistry.
An H&E-stained cross section of the sural nerve {e-Fig. 42.1)* contains 6 to 12
fascicles, each surrounded by flattened perineurial cells. Outside the perineurium
is the epineurium, containing connective tissue and an anastomotic vascular net-
work. Often individual myelinated axons can be seen in H&E-stained material
{e-Fig. 42.2). The endoneurium {the space inside of the perineurial cell layer and
outside the axon/Schwann cell units) contains fluid, collagen, capillaries, venules,
fibroblasts, macrophages/monocytes, and scattered mast cells. Immunohistologic
localization is useful for the demonstration of neurofilaments {a rough measure of
axon number, e-Fig. 42.3 ), amyloid, immunoglobulins, subtypes of inflammatory
cells, and growth factors and their receptors.
Plastic-embedded sections of 1 J.Lm thickness {e-Fig. 42.4) provide a wealth
of information. Myelinated axons range in diameter from 2 to 18 J.Lm and are
coarsely separated into small {mean of 4 J.Lm) and large {"'12 J.Lm) myelinated
axon populations {e-Figs. 42.5 and 42.6) with myelin thickness related directly
to axonal diamete.t The patterns of nerve damage visible in plastic sections may
be characteristic {although rarely pathognomonic) of certain disease entities or
pathogenetic processes. Qualitative information is provided on the degree of
myelinated axon loss, distribution of axon loss, presence of active axonal degen-
eration {AD) or demyelination, identification of regenerative clusters of axons,
swollen axons, onion-bulb formation, the nature of cellular infiltrates, or amy-
loid deposition. Groups of unmyelinated axon populations are detectable in plas-
tic sections {e-Fig. 42.6).
Ultrastructure provides additional detail and is the only definitive method
to evaluate unmyelinated axons {e-Fig. 42.7), which are three- to fourfold more
numerous than myelinated axons in the sural nerve and typically are <2 J.Lm in
diameter.
If necessary, lengths of individual lightly fixed, osmicated myelinated axons can
be dissected or "teased, out of a fascicle with pins. Each myelin internode, main-
tained by a single Schwann cell, ranges from 0.2 to 1.8 mm in length, increasing
*All e-figures are available online via the Solution Site Image Bank.
673
874 I SECTION IX: NERVOUS SYSTEM
Figure 42.1 Selective loss of small myelinakd axons. Normal fascicle is on the left.
Axonal Wallerian
Degeneration Degeneration
Figure 42.2 AD vs. WD. AD is typically characteri2ed by simultaneous
AD, regenerative clustm~ (arrow), and axons with pathologic signatures
(crosshatched) compared with synchronous and uniform degeneration of
most axons in a fascicle in WD.
~i(.t:!l(fl\1
11111111 Tllldnt Dlllp
All.mi'IUin N:l:liomldo Alml!r1h8
Alwllc .!ltll cii!Mdo Amloclarone
Oe.dmh.m Buelc!hom -..11>
Cdd Corbm dlll.lllde Chlo"""hl
lad Dmo111)41mmproplonMo c~~
Ulh-.m Db<ln Clloq'*'ol
M<mll't' Ell\11'101 Ook:HclfiO
TeluriLin Eli\'Jieho OJddo CJanl:lo
Thll.,m Hmcorbor...nd """'IIIII Do-
Lalrdaaln lllchm.:.talo
OlpiOjlllosp/lona..,.. llllulllrun
"""h-,- Ooll>lllbli:ln
wr..,. ElllomWI>I
Tob>no ~
'!'ode o11 """'"""" GIIIIOII\mlde
'l!lek~·· ~-
Vocor -
"')1cllioflc!o --
lilian• Mllon-
Nirofln~
NlrouoOlllle
N•..W.dtk&
~lne
p~
Podop/I)Wn
1'$:1\lollnotod '*'h...,.,
PI'OCONmlde
-
l"jfldoxlne (Vbmln llol
Sod~m-
Sllrunln
r .....tnu• (Fll!iOI!)
T.,...,.IPa-1 (TI>O>I), ""'""-0
'TIIoldomlde
YlncuiWoldo ~""· Ylnblo6el
Zlne~-lonN
Chapter 42 • Nerve Biopsies I 87 7
Figure 42.3 Ischemic pattern of axon loss. Intra- and inrerfascicular variability of axon loss
is typical but not pathognomonic of a vasculiti.c/ischemi.c pathogenesis.
Figure 42.4 AIDP. Patchy loss of myelin in large and small axons is accompanied by an inflam-
matory infiltrare (solid dots). Normal fascicle is on the left.
cases. CIDP may be a chronic form of GBS in which there is failure of regulatory
T-cells to suppress the typically monophasic attack of GBS. Its autoimmune
pathogenesis involves CD4+ and CD8+ T-cells, and possibly also antimyelin
antibodies. HLA subtype frequencies (e.g., HLA-DR2) differ between GBS and
CIDP.
C. Antimyelin associated glycoprotein (MAG) neuropathy is a slowly progressive
demyelinating neuropathy with an associated lgM monoclonal gammopathy
(monoclonal gammopathy of unknown significance, MGUS) in which the anti-
body is directed against MAG, a molecule which supports myelin integrity.
Anti-MAG antibodies deposited on portions of the myelin sheaths of periph-
eral nerve axons result in altered myelin periodicity, demyelination, and AD.
Immunosuppression and plasmapheresis have resulted in transient improve-
ment. Rituximab, a mouse-human chimeric antibody against the B cell surface
marker CD20, is reported to have clinical benefit.
D. Antiglycolipid, antisulfatide, and antiganglioside neuropathies have been associ-
ated with a wide range of clinically identifiable acute and chronic neuropathy
syndromes.
E. Anti-Hu antibody neuropathy is associated with paraneoplastic sensory neuropa-
thy and likely reflects development of antibodies against shared antigens of
small cell lung carcinoma and dorsal root ganglion neurons. Infiltration of
CD8+ T cells is also present.
F. Autoimmune autonomic neuropathy resulting in autonomic failure following a
viral illness may be mediated by circulating antibodies against ganglionic nico-
tine acetylcholine receptor.
VII. GENETIC NEUROPATHIES
A. Hypertrophic (onion-bulb) neuropathies
The concentric proliferation of Schwarm cells in response to multiple
episodes of demyelination and subsequent remyelination results in onion-
bulbs, the defining pathologic hallmark of a group of neuropathies. Onion-
bulbs are visible in H&E-stained nerves (e-Fig. 42.24), a pattern accen-
tuated with immunolabeling for Collagen type IV (e-Fig. 42.25). Onion
bulbs are best illustrated in plastic embedded nerve (e-Fig. 42.26) and
with electron microscopy (e-Fig. 42.27). Nerves may be palpably enlarged
and conduction velocities markedly decreased. Genetic analysis has iden-
tified mutations in myelin constituents (Po, PMP22, myelin basic pro-
tein), including those localized to noncompact myelin near the paranode
(MAG, cormexin 32, neurofascin 155) and axonal proteins (Caspr, contactin,
kinesin). A more complete catalogue can be found at several websites (e.g.,
http://www.molgen. ua.ac.beiCMTMutationsl).
1. Hypertrophic Charcot-Marie-Tooth disease (CMT 1) is inherited as an auto-
somal dominant condition resulting from a duplication or point muta-
tion in the PMP-22 gene or a point mutation in Po myelin protein (both
needed for myelin compaction), a defect in the early growth response 2
gene (EGR2), or mutations of periaxin (a Schwarm cell protein regulating
its shape and axonal communication). Animal models have established a
role for macrophage and lymphocytic infiltration in disease pathogenesis.
X-linked CMT demonstrates a defect in the gene for cormexin-32.
2. Other types (Table 42.2). Dejerine-Sottas disease (DSS, includes HMSN-
ill+) begins in early life, some cases of which have a demonstrable genetic
defectinPMP-22. periaxin, GDAP1. EGR2. or myelin protein Po. Refsum's
disease, caused by phytanic acid oxidase deficiency, also results in an onion-
bulb neuropathy.
B. Hereditary neuropathy with pressure palsies (HNPP). Teased fiber preparations
of this dominantly inherited neuropathy demonstrate marked focal hyper-
myelination (called tomaculi or sausages) characterized by redundant myelin
110 I SECTION IXt NERVOUS SVtHE ...
O..IIN"M
CMTIA
CMTIB
CMTIC
llh111'111
AD
AD
JU)
-4-
~i(.1:!1(fiJI Rtlmt~KeNdttlf"IN•,..,_wtttlOIIoftl.41ncllom~ll
PMP22
~
EGR2
,..,..,.
ONon bub (oeJ
0~
08
UTAF!SIMPLE OB
CMTIX )(!) Comsln32 Alon.V08
CMT2A AD - F1mly Mombor IB (!QIIb) Alonol/:1:08
CMT28 AD RAB-7 Alonel/:!:08
CMT21l AD <lt,l:;liiRNA ~ Alone!
CMT2£ AD Noilroll~m~ Alonal
PMP22, Po, COMoodn 32 Alonal
CMTa(DSS) ADorAR PMP22, Po, EGR2 P..-, 08
(of do """') GDAPI
CMT<4A AA <lDAPI 0~
CMT48.1 AR Mlo>tubllloJ1n lle..... +~
lldio1ad1Jf..... 2
CMT482 AR s.t blndtns --2 oa + -~ thldolntns
CMT-4C AR SI-G & -IM!I>tsclo reput 08 +fecal thldolnll\ll
dan~In2
CMT4D AA N-m,rc dowmll"""' OB
IHMSN-~ Re8\11111Dd l!lll&l (NOIIGJ)
CMT4£ ARJMl EGR-2.1'1> SeYelori"(IOihloo>
CMT.U: ARJAD 08(~1)
- · PMP22, 1'1:>, EGR2
HNPP AD PMP22 Dom)OI + ~om ....u
~4.2• HGii'W.Bieplisa I Ill
Lymph Nodes
Anjum Hassan and Friederike Kreisel
I. NORMAL ANATOMY. Lymph nodes are the most widely distributed collections of
lymphoid tissue within the lymphoreticular system, which also includes the thy-
mus, tonsils, adenoids, spleen, and Peyer patches. Due to their easy accessibility,
lymph nodes are the most frequently examined lymphoid tissue for a lymphoretic-
ular disorder. Microscopically, the lymph node shows four compartments: The
most obvious are the primary and secondary follicles, which are usually found
near the capsule, surrounding the follicles and extending deeper into the node
is the paracortex. The third and fourth compartments represent the medullary
region and the sinuses, respectively (e-Fig. 43.1).*
II. GROSS EXAMINATION, TISSUE SAMPLING, AND HISTOLOGIC SLIDE PREPARATION.
Fresh lymphoid tissue should be examined by gross inspection, touch prepara-
tion, or frozen section examination to assess whether the:
A. Tissue represents adequate sampling, and
B. Tissue needs to be allocated for various ancillary studies essential to a correct
diagnosis. The fresh lymph node should be cut perpendicularly along the long
axis, and material for ancillary studies procured as follows:
1. Wet touch preparations fixed in 95% alcohol or formalin, for H&E or
Papanicolaou staining
2. Air-dried touch preparations for Giemsa or Wright-Giemsa staining, cyto-
chemistry (myeloperoxidase, nonspecific esterase, etc.), or cytogenetics [i.e.,
fluorescence in situ hybridization (FISH)]
3. Rapidly frozen tissue for immunohistochemistry, cytochemistry, or genetic
analysis
4. Fresh tissue (in RPMI 1640 medium or saline) for flow cytometry
5. Sterile fresh tissue for microbial cultures or cytogenetic analysis (karyotyp-
ing or FISH)
6. Thin-shaved tissue rapidly fixed in glutaraldehyde for electron microscopy
7. Paraffin-embedded tissue after fixation for routine H&E-staining, immuno-
histochemistry, and special stains [e.g., Giemsa, periodic acid-Schiff (PAS),
elastin, trichrome, Leder, etc.]
Procuring tissue for histology takes priority over other studies. The most
commonly used fixative for permanent sections is 10% neutral buffered for-
malin. B5 fixative is commonly used in addition to 10% neutral buffered
*All e-figures are available online via the Solution Site Image Bank.
683
684 I SECTION X: HEMATOPOIETIC SYSTEM
formalin because of the sharp nuclear detail it produces. Howevet; this mer-
curic chloride-based fixative is very expensive and poses an environmental
hazard. Furthermore, molecular studies cannot be carried out on B5-fixed
paraffin-embedded tissue, because it will generally yield poor polymerase
chain reaction (PCR) amplification results.
Ill. DIAGNOSTIC FEATURES OF COMMON BENIGN DISEASES OF LYMPH NODES. In reactive
lymphadenopathy, there are five different architectural patterns to be recognized,
with many showing a mixed pattern of response.
A. Follicular hyperplasia (e-Fig. 43.2) is characterized by an increase in number
and size of B-cell germinal centers and is common in lymph node-draining sites
of chronic inflammation. This pattern is also present in syphilitic lymphadeni-
tis where, in addition to the marked lymphoid hyperplasia, thickening of the
capsule by chronic inflammation, fibrosis, and neovascularization with arteritis
and phlebitis, and a marked plasma cell infiltrate in the medullary region pre-
dominate. Rheumatoid lymphadenopathy and acute human immunodeficiency
virus (HIV) lymphadenitis are other examples of marked follicular hyperplasia.
B. Diffuse (paracortical) hyperplasia shows expansion of the T-cell paracortical
areas. This pattern is commonly seen in viral lymphadenitis [Epstein-Barr virus
(EBV), cytomegalovirus (CMV), herpes] and vaccinia lymphadenitis revealing
an expansion of the paracortex with increased immunoblasts, imparting a mot-
tled appearance. Follicular hyperplasia and sinus dilation are often concurrent
findings in this entity, resulting in a mixed pattern of lymphoid hyperplasia.
Phenytoin lymphadenopathy represents a relatively pure diffuse hyperplasia
showing an expanded paracortical T-zone with numerous large immunoblasts,
eosinophils, plasma cells, and neutrophils.
C. Sinus hyperplasia describes increased cellularity within the medullary sinuses
of lymph nodes. Sinus histiocytosis is seen in numerous nonspecific responses
to chronic inflammation, as well as in lymph nodes draining solid tumors.
Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman) disease
is characterized by markedly dilated sinuses filled with CD68+, S100+ histio-
cytes showing emperipolesis. Lipophagic reactions causing sinus histiocytosis
with accumulation of phagocytosed fat include mineral oil ingestion, Whipple
disease, and lymphangiography procedures. Prominent vacuoles in the histi-
ocytes can generate a signet-ring cell histiocytosis, a pattern that should not
be confused with signet-ring-cell carcinoma. Finally, vascular transformation
of lymph node sinuses (e-Fig. 43.3) shows a sinus pattern, and it is important
to distinguish this entity from Kaposi sarcoma (e-Fig. 43.4). The latter can be
distinguished from the former by the proliferation of spindle-shaped Kaposi
sarcoma cells forming cleft-like vascular spaces that contain erythrocytes, many
of which are extravasated.
D. Granulomatous lymphadenopathy describes formation of epithelioid granulomas
in lymph nodes. Caseating granulomas are epithelioid granulomas that form
central necrosis and caseation and are typically seen in Mycobacterium tuber-
culosis lymphadenitis. Special stains for mycobacteria in paraffin-embedded
tissue (Ziehl-Neelson, Fite--Faraco) will detect the bacilli as bright red, slen-
der, beaded microorganisms (e-Figs. 43.5 and 43.6). Mycobacterium leprae
lymphadenitis and histoplasma lymphadenitis are other examples of caseat-
ing granulomatous inflammation. Necrotizing, noncaseating granulomas are
present in cat-scratch disease (e-Fig. 43.7) caused by Bartonella henselae. in
which suppurative granulomas with stellate microabscesses surrounded by pal-
isading histiocytes predominate. Kikuchi-Fujimoto lymphadenopathy is char-
acterized by necrotizing granulomas containing karyorrhectic debris, but lack-
ing neutrophils; this form of necrotizing lymphadenitis characteristically occurs
in young Asian women. Nonnecrotizing, noncaseating granulomas are charac-
teristic of sarcoidosis lymphadenopathy (e-Fig. 43.8), composed primarily of
epithelioid histiocytes with scattered multinucleated giant cells, lymphocytes,
Chapter 43 • Lymph Nodes I 6 85
and plasma cells. This type of epithelioid granuloma can also be seen in drain-
ing lymph nodes of Crohn's disease.
E. Acute lymphadenitis is an acute inflammation in lymph nodes draining an
infected focus. Acute lymphadenitis is almost exclusively bacterial in nature,
and morphologic features range from focal infiltration by neutrophils to necro-
sis and suppuration with abscess formation.
It should be mentioned that in most cases of benign reactive lymphadenopa-
thy, a combination of more than one architectural pattern is present in the same
lymph node.
IV. INCIDENCE AND EPIDEMIOLOGY OF NON-HODGKIN LYMPHOMAS
A. 8-celllymphomas. B-celllymphomas constitute the vast majority of lymphomas
in North America and Europe, accounting for nearly 90% of all lymphomas.
Diffuse large B-celllymphoma (DLBCL) (""31 %) (e-Fig. 43.9) and follicular
lymphoma ("'-'22%) (e-Fig. 43.10) are the most common types. Immunosup-
pression, specifically due to HIV infection and immunosuppressive therapy to
prevent graft versus host disease, is associated with a markedly increased inci-
dence of mature B-celllymphomas, particularly DLBCL and Burkitt lymphoma
(e-Fig. 43.11).
Some of the low-grade B-celllymphoproliferative disorders include follicu-
lar lymphoma grades 1 and 2, chronic lymphocytic leukemia/small lymphocytic
lymphoma (CLUSLL) (e-Fig. 43.12), nodal and extranodal marginal zone B-
celllymphoma (e-Fig. 43.13), and lymphoplasmacytic lymphoma, which are
generally indolent, but incurable and usually present in a disseminated stage
with bone marrow involvement. Mantle cell lymphoma {e-Figs. 43.14 and
43.15) and DLBCL represent "intermediate-grade" B-celllymphomas that
generally show a more aggressive clinical behavior, but are potentially cur-
able. The same applies to high-grade B-celllymphomas, which include Burkitt
lymphoma and precursor B-lymphoblastic leukemia/lymphoma.
B. T-celllymphomas. Mature T-cell and natural killer {NK)-cell malignancies are
rare, accounting for only 10% to 12% of all non-Hodgkin lymphoma (NHL),
and usually are more aggressive than B-celllymphomas. The most common
subtypes are peripheral T-celllymphoma, unspecified {"'4% of all adult NHLs
and "'30% of peripheral T-celllymphomas) {e-Fig. 43.16) and anaplastic large
cell lymphoma ("'3% of all adult NHLs) (e-Fig. 43.17). In general, T-cell
and NK-cell malignancies are much more common in Asia and are linked to
viral infection with EBV (NK-celllymphomas) {e-Fig. 43.18) and human T-cell
leukemia virus (HTLV-1) (adultT-cellleukemia/lymphoma) (e-Fig. 43.19). The
current 2008 World Health Organization classification of lymphoid malignan-
cies is summarized in Table 43.1.
V. PATHOPHYSIOLOGY OF NON-HODGKIN LYMPHOMAS
A. B-cell lymphomas. Mature B-cell malignancies mimic stages of normal B-cell
differentiation; therefore, classification is generally based on their morpho-
logic and immunophenotypic resemblance to the normal B-cell counterpart
(Fig. 43.1) (Best Pract Res Clin Haematol. 2005;18:11). Normal B-cell devel-
opment begins in the bone marrow where precursor B-lymphoblasts undergo
immunoglobulin VDJ gene rearrangement and develop into IgM+, IgD+ na1ve
B-cells with surface immunoglobulin light chain and CDS expression. These
resting B-cells circulate in the blood and occupy primary follicles and man-
tle zones of secondary follicles. Malignant counterparts of CDS+ naive B-
cells are believed to be CLL and mantle cell lymphoma. Upon antigen stim-
ulation, naive B-cells undergo blastic transformation, migrate into the cen-
ter of the primary follicle, and form the germinal center. These centroblasts
are large lymphoid cells with vesicular chromatin and several eccentrically
located nucleoli. They express BCL-6 and CD10 but switch off BCL-2 pro-
tein expression, therefore becoming susceptible to death through apoptosis.
Centroblasts undergo intense proliferation that is accompanied by somatic
II& I SECTION x.. HEYATQPQIEliC SY5TEU
<
''I.I II f,,I 2001 WHO C1amtlcatloft., LfmplloiUeojllul!u ,,........
s.llJiy~-
Pmwy-.-..<:030+ T<d ~Ph/lllrollal111Ne dllonlon
Pmwy_,_,. poolpllaral T.... ~phornot,,.,. tu~
Pltnoly -•mmo-4ella T<dl tl!nlli>orno
P r t r . y - coa ra~t~vo aar..W.•pld"moi!'Ojlle ~ T<lll ~Ph"""'
P m w y -<:04 ralllwl ""'""""'"~"' T<d ~pkomo
P._,knl TtCIII tl!n]lllomo, NOS
An&lofmmunoblllllc T<ool tl!n]lllom.t
Anopillllc .... cdll)mp/lome, ALX ]>OIIIMo
Anll>illllc lu• ...1tl!ni>home, ALXn11011<e
fi'CIT!: ~SH, Clmpo £. Kan1a NL.e141l,4!lda:. WHO~ ttl~d~
Nd ~- cyan, IARC ...... 2ll08. u.. """ """'"""'·
888 I SECTION X: HEMATOPOIETIC SYSTEM
• ~Multiple~
l ~myeloma:-j
Figure 43.1 Brief overview of normal B-cell progeny and malignant counterparts.
Migrate
to th ymus
loll Subcapsular ,. I,. Inner ,. 1111( Medulla ••
cortex cortex
Stem cell Precursor T cell
...........
Ill lhltaan_,rnbcllnolsmdtnd lql ....tAtett..,.anl!>lullc?
'* w
Do111o ~ niiCiei\Otila Gtncdll CUIIOII'(,.,.,....Wd)! Or • ~ ohope\Otila
bompyor I1IOidlod ""'"''" O""''l'lull Or a ..nabl&shope- doeplf ftllded ttf.,.......S
nuel&l (CioMiod)t
Hlolo... ..,...
b """""" ~nt1b11>ofaa .,_,!II;(' a_.nootAlem--""""'
llllilo I
!._
UIII,P{loJ-.,....,.... ... lillrJI
..
At1. Ill•-ofB
-~~~
(9anonl~ Cll:!n+) « TI I - l.\illnonl~ Cll$+)1
T lymphocytes normally rearrange four different TCR genes (TCRa. {3. y. and
8) to encode a unique antigen receptor expressed on their surface, and South-
ern blot analysis targets the TCR/3 gene. In contrast to normal tissue, where
only one band corresponding to the germline configuration is visible with this
technique, both B-cell and T-cell malignancies will yield an additional band
corresponding to the clonal population.
I. PCR. Because Southern blot analysis is labor intensive and expensive to per-
form, many laboratories have turned to PCR amplification methods to detect
clonaligH and TCR gene rearrangements.
The 200 variable segments of the lgH gene contain three highly variable and
mutation-prone regions called complementary determining regions (CDR1, 2,
and 3) that are interspersed between four conserved framework regions (FR1,
2, 3, and 4) that provide reliable targets for consensus primers. About 70%
of B-cell malignancies harbor donal rearrangements that are detectable with
framework 3 primers, and about 15% to 20% of additional donal rearrange-
ments can be detected when framework 2 primers are used. Therefore, most
laboratories use primers targeting frameworks 2 and 3 to detect at least 80%
of all B-celllymphomas.
The rearranged TCRy gene is most suitable for clonal detection by PCR
amplification in T-cell malignancies, because most T lymphocytes harbor rear-
rangements in 1 of the 11 y segments. Because many of these segments are
homologous to one another, they can be targeted by a single consensus primer
set. TCR8 and TCRcx cannot be targeted because TCR8 is deleted during TCRcx
gene rearrangement and TCRcx has such a diversity of possible rearrangements
that these cannot be covered by current probe technology.
J. Cytogenetic analysis is a morphologic study of chromosomes to assess changes
in their number and structure.
1. Conventional karyotyping requires viable cells, and analysis of chromosomes
is performed according to their size and banding pattern after chromosome
staining. Although this study has become critical in the diagnosis and clas-
sification of acute leukemias, it is not frequently used as part of a routine
diagnostic workup for lymph node biopsies.
2. FISH uses one or more labeled probes directed toward specific portions of
chromosomes. It is helpful in locating specific translocations, deletions, and
amplifications of chromosomal regions. The advantage of FISH (see Chap.
59) is that it can be performed on nondividing nuclei (interphase nuclei),
eliminating the need for cell culture. Furthermore, interphase FISH can be
performed on a wide range of specimen types, such as peripheral blood
smears, cytospin preparations, or paraffin-embedded tissue. Many of the
classic chromosomal translocations that characterize specific lymphomas
can be identified with interphase FISH techniques. Tables 43.3 and 43.4
summarize the characteristic morphologic, immunophenotypic, and genetic
features of B-cell and T/NK-cell malignancies, respectively.
VII. PLASMA CELL NEOPLASMS. Plasma cells represent terminally differentiated B-
cells. Their neoplasms are characterized by secretion of a single homogenous
immunoglobulin product known as the monoclonal or "M" component, which
can be detected by serum and urine protein electrophoresis. The World Health
Organization categorizes plasma cell neoplasms into monoclonal gammopathy of
undetermined significance (MGUS), plasma cell myeloma, plasmacytomas (PCs),
immunoglobulin deposition diseases, and osteosclerotic myeloma (POEMS syn-
drome).
A. Patients with MGUS are asymptomatic with no evidence of lytic bone lesions,
but reveal a clonal plasma cell population in the bone marrow of< 10% and an
associated small M component of <30 FJL. No lytic lesions or myeloma-related
organ damage is present and a B-celllymphoma with plasmacytic differentia-
tion should be excluded.
o-..
ClUil!.
Cl'-ll>lllf _ _,.,
Mosllt Oljmpll>mollt, tni!Y
IIIU\11li>IIIJ
PB: Srnal colo w!th<OelliO\Y
...........l>l.nlt
al20 (-11), CD19,f&M,
.......
Trllomy 12, dlllllonut
<limped <111>"'"'*' end
~- fl'llll>oo,
.............. homob1i= h»mplcuous nu<!ool
(tO, alll2 (-11). CD~;,
ali'3, CD79o. 0043
13ql4,d-Oil
llqll2-2il
Prolfmp"""¥11c
........... he!>etool>lenom...W.
tlmPhlldenolii>IIIY: 1><1111>11era1
•blciiD lfm"""'*"l&
Mllbd 'l>lln«i"'I\Y...m...l
---of
LN:Prolmte~•~
"paiUnmul'tllllula" •I'd
prot/mpl!oq1as
PB: IIIII! kim..-- o.t11 al20 (brV!I), CD19, ~.
~IIIII&~
~no.llfi
doNIf ,..,.l'fl'd
canp~aa ""~ ...
tlmpllod ....pallly, flll'lcly round nuclous, ~ al22, CD711o, FMC7, common~ JbNM'ml!liM: of
IIMNmll
liiii'C lfmphocoJto eoijnl _oll,..,_,..,d ali'31yplcallr ·-cos TP63 In -tiO'J.. I(H
C>100 " 10'/l.l promNI!t <lllltlel nucteollo Pt&0011tln113ol- cb\aly re.lrnlrcod
tllkYooiiiMJklmll Prodomba.mloid~ men. PB: Sm«J. to MICIIJm-ol!od col& all&, CD20, C022 (b!Wrtl, Nool*llle~
__
Ol'i&lmllllllllr. PII""'''II)f)llls wtth""'rrrfetenllslliii!Y al79tl. CDlie(~. «b_,.lty,llfi clcNiy
LJ!nplql~
t.mphornll
Walkno111!m
wtth~penla
""'·~..ttl-lad
,.,.........,
I!IW.tloooty eym!*mt
pro)dono
Spleen: Rod !>liP ln11-
Crna=ol')': "'JI>ody 11.....1
Mll:lln of """!111m~
~'""""'"'
..... "*- lfmpllo<:JIOO.
Dln:het b>dlao
end
-..
al2S lblWI1!, CD10:!1,
TRAI\DBM4lnllul»
No 1podilc .,.llltic
«b""""lty.llfi doNIf
I<IOiit«JWWd
,...,.,...b<JIInomlo ~o!COS.CD:zil,
Of COlO
Splon~ maiJ,NI Splono,....lf, . , , _ . PB: SmiJ. to MICIIJm--- allS., CD20, 007llo, IJM Allelic ""'old!_,.
mnoB-coll -101111•-.rnune with pollor Yl!l (¥1- and (tO, no - - n o f 7<!21-a2ln40\lloof..-.
t.mphorno lll,..,bo<:ylo..,.... or""""""· IJm~
Spbon: Both-pllpond Old
al6, CD23, COlO, or tbomy31nrn-IJH
doMif ...mlrco<l
""''~ lfmp-nopotny C043
1111t> lnl'l!nltlon
....•
IMOrnii'IOtl
(oMII'o<lold)
••
• 1m=! • '1:5!11 c~~a,.d••tooucr- <If DillS a~ ~.con Mall.ll&tlld• <m•..o
D•a. ~1111 ..1Jn'Millotta.t 1111,..1111J ••••~'"~,. Caallfn
Ealno<llll mqn&l Hlllay alclwnl: lnfllm""""'Y i'olymofplloulln- a1 Cl>19, CD20, CDi'!IQ, .,., no 1(11;lll)(q21;<121) In -25%
"""" BGI Of •!b*nmune diad.,. ~llo> <olb, ""''"""lonal COO, CD23, to !iO'll.o f -
t.mphom.o(INI\1 ~P)'ftlrl"'""' mo~<ell..,al CI>IO 1(14:115)(q32;~.1)
bmphoft\ll Haohmolo111midlll. S!Oiretl limP~ -..s ~1\i.nl, lllti c:lcNIY
~-l. Olnctll'lO'It mmunol>hblo end limr>llcid r&~~mu'CIId
comnm.aball- '*With...,,~
-
dftltwtlltlon, .CIItlfl'UI\
'*""(limpliooplhol~l
~ ,...lltlal zone l<>:$1md OfiOnOrdmd Milllbll zone 0111d ~- CDI9, CD:!l>, CDl'911. _,.,no The U'OIM~
wtlll ~al MZI. _,..
....,...led
lkoiiiJmDhoma 1/mlhdo~ .,... ~llnl1odb!' ""''l'&lllllorl al COS, CDit),
~l .... lkolk, CDIO not .W""''; laH elonll!r
,...,~ lk>el&, til amd roononpd
limP,.,.,_ plosni1 col
d i l l . . - mi!Y 1» ~
Fol...,r~phomo Wkllsprood poolpllonillllld Fdwlllr )llltl.wn alclosoly Cl>19, CD:!l>, CD22, CD7'll, 1(14;lll)(q32,"<!21) ....
<Onbal bmpllad""pa111y, pocl;od ""'plint!o l'dll::loo BCL4 BC~ CDIO RWIIT8l-il8rlt of the
-
""""mlliTOII' -ln40'll'. """poood " ' - ..... BCL·2 IOnOiood~ to
"'- den..-:d ~ « ••
G-l:0-6oo-l!llllllpf
--lorlal111o
BCL-2 prolaln ill1d s..WOI
""........,...lpnl
Gredo2:6-15-pt
Gredo3: >15 -IMb'llpf ...,.,...
c:olb; I&H _I.,
f>OOIII>Io"""'""_'_"'
"""' tloymlo """"nil.
bloi>O!'"' mDilo diM omd and
la•na HlA...,.tnll
-lonboftonebloorrt
IPJI>onl!plold .. l)'lllypo.
pNin<-9p
f)fc11.oo!o-""*
-----
obleond 1>,-
and"""" aJ!!fal:t
Buotoll t.mploom• Hfllo\1' .......... tlmplooml C019, C02D, C02:!, COlD, I&H donoollf ••IYM.II'd;
obn ~na.tm•omel booplollc -IIOIWd C{)JfMJ BCL-61 no 1(11;1<4) Inmoool.-,
- o r • . . . llullllmloo, rllk ~~~ anc1 racukr nii::W -lonoiTdl, BCL-l!, 1(2:11) and t(ll;22) ..,.
for ...- ........... "¥"f'ro wth -...nl .srna!ll'ltl.ldloll 10-Qind.. of IOO!r.
~-.....-. "'lo<<omk:'" 8uri<ltt
tlfnplooma wttn ~em ol
Unalbb-body m•"""'-
lmPIII11!1I._I!Y-OiiY"
- llld ot""' fldll
llln.lo:llns, • •porut t::" 8 uri<ltt
•-mnco
tlfnploomo wtttl•bdomlnol
on ....
CU.- tim~ ....lolmiii;$U. omell irlrcl~ IJm- MAll, onu0011a 1111 ><laW IJm-lllooe; II. Ion,..,oqt:o~xofn QI, iiiiiNiualhot f8, ~I
- Lll; hilt, hlii!--MI; TRAP. tl1lr<lln-.~dl- HLA. humoon lou-•~ Tdl, amlwl~ ~--
....•
i r1'i~Q~:~)~~:f~·S~X~li!}l
o-..
CllaJtdelll1lc Fullnl'lf ~""""""" T~llllld fjlt~IIIIIIIIII'Odet
Cllllr.l Ifill rtt'IM ..,..,,. __,_,.,.. _,.,
r....nprot~mp!Jo<;)11o 11#-d->Oith PS: Mlldl..,..lzld colla wll\ C03, C02, C07, C04+1C08- ln'<(14)(q11;qW In 110\11,
le<Rmla ho~l&non-tt and .......... C$>plinm •.tllblo _, G)'ll',, CD4+1Cil!l+ In 1(14:14) (qll:<jW In 10'll.
IIIII'MIIIIod tim~.
mllbd tim~ ,.Uitj
""'*"'"lo
nucloolle. ellll
I>IOCI\IIJIOno«bloll&
:I:Slt>, co.h-tD$+ In IS'l(. ~ltR...,,6ond
ret.-1. deNlireamtnft)d
>100 X 10'11. · -llld TCR
lltromboc>yt.oponla
r... u~a,...,..,..,, Indo,.,. dnbJ COU/lit~ .tMIIl'6 pg, Llrp .,.,••" tlmpllcq1as coa, TCR...t,6, coa In 110'11., CloNJi ,..,.,pd ltR, no
tim~ wll\ ol>ulllont~ end
~·
-nmlooolTIA-1, unlqw eylopholl<:
lcMicomll onomll, mltl II> m - ,.,80f 0011/118 ODI'Ophk CD57, flOf(orln, IV""'Yl\1& 1\bnonnal~
tlm~ modenl1o !Pralilo:BM:~I B. Fao !CD95l ellll Faol.
'l'iem"'""'lf, rlwJmmld lnfl-
l!lrthrtt:I.!Ji~ du.lfldttrlmmune
compllilcas, rw-IPJI'In...
l)oll..,..,..
_._NKoall Morep-nlomqMiilm; pg, (Jmpllold-lo!p<tllen C02, cCD3 ,, C056~ 'T'JA..lf TCR no! cbldy ,..,......,,
lcMicomlo -.loumlo blood ploUo, nonnal LGL wll\ •1\Chi!J . .noymo 8, porto!fn, EBY. ESV ~ h <~IIIII
"'mill~! "''fllploms.
_
b.,opl\lt::'*"'IMm
_,[f11IJillnU[\)O, no""~'""""".,""'"""' oploomall'onn
~·· hoTJO.toeolr
lm'IIIIII!Y. IIIJiiotpn flllu,.. hypotdl- .......
C03«C057
-ion
ilpndHI
NluiT-all.......,ll £ndornlo In Jlpon,llto Cort!I>Nn pg, Mlldllm·lo IIQilHIZod colla Cl>2, CD3, 0051 CD4, C025, CloNJi lra.,..tld Kll\41,
bonln, porto of C8n1nll-.
tllild '> HTI.V-1. ('11_...,,
wll\ polf!ollobid - -
lodl C07 '*"'".,...."'"""' ltR
li!!Potoe~ll.
llol>'bi>IMon'IIIIII!Y, ti.CH,
.....,lotldT....a
lmmunccloll:ilnc!rwll\
oppo11111lrllo , _
i:XW~I NK-11"-<>ll M010 P,_l'lt b Mil.,..,., ~~~~r.t~. Cl>2. CD56. cCIXh. 11:R notc:ioMIY lllllmii'Cild.
ttmphoiNI, nlllll Centro~,
_ _and m_S..IAI!Amlllb,
__ tnfjocoll!!t: ~nd VIN!JimO 8, TV\-I, £8V P<-ln elonll
t)l>o
·~~ pMo/11\ 111Jifoo&C03 I'd "11-lfann
- ... -
Gl not
cdrancdalllloo
lnc:luda ..... taft t:tasw, tasl:tt,
pc!lom, hypon:llnomllll::
....~~~~~, lnlllmbalcl
lnflomrnalory....
"''"-
~T-<>&1 Abeocla1od Willi <:ollie d lloolt» U~m-lma•b Cl>). CI>8-H-. Cl>l<G. ClcNiin~~~1111n.DldTCR. HLA
ttmphoft\a end~~l'llk )o)Jnum or him. btoed TIA-1. PI\IYIIIO, perfoltl IXIA1'050l.IJQB1'1)21)1
~~ •pedrum,lnbtmlad pno1ype II""!"".......
lnillmrnalo'Y celo
~pllni:T<III
ttmphoiTII
PMk lndd""" ln•do-
olldyou,.od...,morlold
IIIPI1DII!>Iotuo.,..tf"""
tlm~.rn«e
oomnm In.,..- ..ttl
lmmiiii*ITJIIIOIIOion
""'-...,..,..
S*ILIIIOIIIII!nlllrwllonalllw,
oploon, •lid lloni> ITIIrrow by
mol(fii!Himd ....
CD3, TCR:-y/1, llA·l,
l'lllllf1Nv "" 004, CD&
TCR-«<ft, •lid podwln
ClcNJyrumo~
bodw'omotOiillD
trbomy8
TCR,
7q,
-~ "'"'""""
T<CIIItlmpi>Oin& saptel; 6pfd~tmlt urlrwol\'ld;
1'1111 pon<:)ll;'l""""' - · ....S """""'· """""-. rtnmhf
IIIPI1Dill>lotu II....\I' orl<rtoais
t.ttcoollfU~ l.or'C 11111111111 hlltoly, mulllplo ~ ~ldennOO'OO~ ln11i!MI oftmor Cl>2. CD3. CDS. 004, ClcNIY lllllmii'Cild TCR
IGIJioM.I'OIIdl-+ PMUO ... <o~l~dll..,.reMJorm• nue1a1. TCR-«<ft. no <lM181'1Jio~of
t<mr,11'11quenllyon lnllk Pll~t'IJJ mb'adbsca•e CD7
_ , oync~ ..... Aai&llt:w'AI!Wrtof~ PB, l.llnmurn ai!OOO&Izo'Y I n c - CD4.C08 rllll> ClcNJyru"""'JIJI TCR
f-lclol;~. ce!ll per mml. •carebrtfofm,. loci<l,.
lllf1!1101b1 of CD7
tlm~.ond -lei
<R<IIr<thf$6my....
..•... [0>1-
•
••
--a!
'
DlleiA Clllll:ll , _ ' - II.,_IDD ...nplllaaw- a-11:1
Mllclmmunobllo11o lmmu~..c.,ncloiiY" Mtt1Un>of 1>011m01~ Cl>2. Cf>). C04l0tll ClcNIY ,..mii'Cild '!CR.
T,.,.ll tlfnplloma tlfnplloma, 111m lUll,...,..,., tim~ - • e\ur tltlomy S,lrlllt>my S,
plolftle1\lalon,aaellot, ~plolm,.....,.....,
COlO,
folk:tMr dendrt!Sc oolll .,. addlloNIX ell,...,...,.•
qlojlonfll, ~I -pltlo, ond hW!IWr\111 byCI>21,
~mmq,lol>llllnomll, + lmrr&lnobllol>; -~ HlV; CI>ZI, coa!i,
''"""-- proml,.,...,ol FDC """"-k lmmuno-.,CI>20+
o.-.lcla i)tlcia& Oll1d EBV+
Pel\?hint r....11 PradomNI!Cii nodlll-buucl BIOOid ~ -m. T,zono Ct>2. Ct>3. ~ C04, mw Re.!mll\,lld TCR
tlfnpmiNI,
-11\od
AN!>Inllc lqll <Oil
tlfnpmm.s
--"'·--
tlfnpllomu lhot oonrd bo
·--of
diM.ua
cllltlhoo<l
tlfnpllomo- 114ip (Ill
or M will froquont8ldnlnodal
...- . P<....wlio• tl
fdllcM. LMI'MI wi!MtlOtll
many aplholbld ellalrua
Kllmlrl: ..,~ = lrup .... with
abllndlrt ~1o1m end
hoo...._.holped nldil
--coso
II'I'IO!f<IIMI'It
tf(. omll'll kllllr;.HT~hurNn T-<011 lill.bmlt>iN'< LDH, LGl,lll"ffO-1; fB, pell""""l-; Bioi, lloMmo"""' IJ:I, q .,.nu~rljn\"'= HE\\ h~
ll'ollolllollll.._.., :::·< _ r _ l f t l e _ TCR, T- """''b, nA, T-<OIIrlll!t:Oid 1-lullro~i':... al, F•...l>d; £8V, (jloall...a..r ; Al.K:l, ll\lpluli>
lllpc•ll-1; IMA,'I'"""~I memll-. . . .; teL• I, T...al6illo!MII/Ijmpltomo I; HLA, IIIJINf'ollulcoojl&l._,.
~ 43 • I.J'Mp!l Hedee I •••
''I.I II ( fHI Q11111a fof 1M Dlll!*fuf " ' - tdlo!Jeloma
f)o.... • ,..._ ooll ,.,.._
PC WI tb:lvt *'PI'I«
donll bono"""""' plurne<:)tol\11 ("""'ly >lOll plume.,.lll)
M~><*lh In ooturn « u'*">: uouetJ Ontt nct•~•ennn 1&0 >30 etdl «ItA >2Srfdl.
• or~ IIB!rtoholn - · :.I.I)Wd"' ~ t.r!M -.p~~o,_
Rtlobd - · orlliale mpolmum(llnemla, ~mil,
r.curnllnl: fnfec;:tk)ns, ek:.)
t111t-..,
-lnollf!ldlnoy,
llllta• Orpur11aau
~ ,....._.. S.. M c.j~alllt lytle IIIII au llfllllr•n
MC!US <lOlii ~ 111111 11'6'0iomoieWI> tlollo Nono
(lgG ..:$5 e'dl. II>\ <:!S
etdll
PC t1o11o -tllaw P- Nona
700 I SECTION X: HEMATOPOIETIC SYSTEM
type Castleman disease can be cured with surgical excision, systemic therapy may
be required in multicentric forms.
IX. HODGKIN LYMPHOMA. Hodgkin lymphoma typically presents as a primarily nodal
disease characterized by a predominance of reactive cells with a paucity of neo-
plastic Reed-Sternberg (RS) cells or variants. Hodgkin lymphoma comprises two
distinct entities: classical Hodgkin lymphoma (CHL) and nodular lymphocyte-
predominant Hodgkin lymphoma (NLPHL). These two entities differ in their
clinical appearance, morphology, immunoglobulin transcription of the neoplastic
cells, and immunophenotype.
A. CHL accounts for "'95% of Hodgkin lymphoma and is subdivided into four
categories: nodular sclerosis ("'70%), mixed cellularity ("'20% to 25%), lym-
phocyte rich ("'5%), and lymphocyte depleted (<5%). The neoplastic cell is
the classical RS cell or its variants (mononuclear forms, mummified forms, or
lacunar cells).
1. Nodular sclerosis CHL commonly presents with a mediastinal mass. His-
tology reveals scattered RS cells embedded in a mixed inflammatory back-
ground of mostly T lymphocytes, plasma cells, eosinophils, neutrophils,
and histiocytes, compartmentalized into nodules by thick strands of colla-
gen fibrosis (e-Figs. 43.33 to 43.35).
2. Mixed cellularity CHL presents with scattered RS cells in a diffuse or
vaguely nodular mixed inflammatory background without nodular scle-
rosing fibrosis.
3. In lymphocyte-rich CHL, the nonneoplastic cellular background is com-
posed of predominantly T lymphocytes. Other inflammatory cells are rare.
4. Lymphocyte-depleted CHL is rare and occurs most commonly in association
with HIV infection. Histologically, sheets of RS cells predominate without
a significant lymphocytic or mixed inflammatory infiltrate.
The main clinical findings of CHL at diagnosis are painless peripheral
lymphadenopathy, most commonly involving the cervical region, and con-
stitutional symptoms such as fever, night sweats, weight loss, and infec-
tions due to immunosuppression. Many of the pathologic and clinical fea-
tures reflect an abnormal immune response due to the wide variety of
cytokines and chemokines (Table 43.8), produced by the RS cells (Blood.
2002;99:4283). The etiology of CHL is largely unknown. Studies based
on the month of diagnosis have revealed peaks in February and March,
and the lymphoma appears to be more prevalent in adolescents and young
adults with higher socioeconomic status (Hematol On col. 2004;22: 11). The
association between EBV and CHL has been demonstrated in numerous
seroepidemiologic studies in which antibody titers to EBV and viral capsid
antigens have been consistently found to be higher in the lymphoma cases
compared with controls. There also have been reports of clustering of CHL
within the same family, especially among siblings of same sex and close age.
Certain human leukocyte antigen (HLA) types, such as Al, B5, and B18,
appear also to be clearly associated with this lymphoma (Hematol Oncol.
2004;22: 11).
B. Nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) is characterized by
scattered large neoplastic L&H cells (lymphocytic and/or histiocytic cells, pop-
corn cells) residing in nodular meshworks of follicular dendritic processes, filled
with nonneoplastic lymphocytes mostly of B-celllineage (e-Figs. 43.36 and
43.37). Clinically, patients present with localized peripheral lymphadenopa-
thy. The disease develops slowly, with fairly frequent relapses, but remains
responsive to chemotherapy and usually is not fatal. About 3% to 5% of cases
evolve into DLBCL.
C. Pathophysiology. The cellular origin of neoplastic cells in both CHL and
NLPHL was finally determined when clonally rearranged immunoglobulin
~··
&-1
.......llllrlf
Pobintprolnflllmrnoblly~lnd.- -lOIIII B<qmpton,.
-·
.Cr..., and ..U.ph... ~·
w
-Md
l:.oolroolill dl'le-. lltdllera1lon, 01\d AoooxtoledlOIIII blood •nd lloolio
-pllllo
W """'"' 0111 dl'lll••-· tllmulllfon of l01111111-~
ll·l,llld TNF-a p:tldudlon llld- plullllq!Dols
W NtutTo!>hl niCIU-- Auociltad lOI!IIuaue
- p i l l ..
May bo adtw... ...-.th , _
W
&-1~
&-IS
T-col 01\d moot cell-h feclor
AIItoaN~
colla
lOIIII EBV+ . . -
ftlf RS
I tE ~...E
.. v
Cl
"' ..
Cl
v
.c:
.c:
..~
:II
i1 p
'
- 1 ... is
~
A
i "' -'
I i.e I i.
i"' If H
~~~
'I! .e
~
11#
~
I :I Jl t•r
... ... "
!I~
~~B ...'Va ..'
~
A
4::
:i
4::
_..
·~
-
...
~
0
i
~ it
..."' -. ~~
0
"
~·
;;·
,_ ii
-- f
10$
706 I SECTION X: HEMATOPOIETIC SYSTEM
and mycobacteria. Fungal hyphae and yeast forms can occasionally be identified
as negative images on Diff-Quik--stained smears (e-Fig. 43.40).
Ill. SMALL-CELL LYMPHOMA
A. Small lymphocytic lymphoma. The aspirate contains a monotonous population of
small lymphoid cells with round nuclear contours, characteristic checkerboard-
like clumpy chromatin, and indistinct nucleoli (e-Fig. 43.41). Scattered large
prolymphocyteslparaimmunoblasts with vesicular chromatin, prominent cen-
tral nucleoli, and pale cytoplasm are present.
B. Lymphoplasmacytic lymphoma. The smear contains a mixed population of small
lymphoid cells, plasmacytoid cells, and plasma cells.
C. Follicular lymphoma. The aspirate is composed predominantly of small centro-
cytes (that have irregular and grooved nuclear membranes) that are intermixed
with scattered centroblasts (that have a large noncleaved nuclei with multi-
ple small peripheral nucleoli) and immunoblasts (that have single prominent
central nucleolus) (e-Fig. 43.42). Lymphoid aggregates without abundant histi-
ocytes are not infrequently found (Cancer. 1999;87:216; Cancer. 2006;108:1).
Although the cytologic criteria for grading follicular lymphoma are not well
established, the percentage of centroblasts on smears has been proposed as a
basis for grading by several groups (Am] Clin Pathol. 1997;108:143; Clin
Pathol. 2002;117:880).
D. Mantle cell lymphoma. The aspirate is composed of a monotonous population
of small to intermediate-sized lymphoid cells with variable nuclear membrane
contour irregularity, dispersed chromatin, and inconspicuous nucleoli (e-Fig.
43.43) (Cancer. 1999;87:216).
E. Nodal marginal zone B-cell lymphoma. The aspirate contains a rather poly-
morphous lymphoid population composed predominately of small lymphoid
cells with round nuclei and clumpy chromatin, with scattered plasmacytoid
cells and immunoblast-like large cells (Diagn Cytopathol. 1999;20:190). Some
intermediate-sized lymphoid cells that have a monocytoid appearance and
exhibit a moderate amount of pale cytoplasm are also present (e-Fig. 43.44).
Lymphoepitheliallesions are not identified on smears. Due to its polymorphous
appearance, diagnosis based on the cytopathologic findings alone is challenging
without ancillary studies.
IV. INTERMEDIATE CELL LYMPHOMA
A. Lymphoblastic lymphoma. The aspirate is composed of a monotonous pop-
ulation of intermediate-sized lymphoid cells with fine granular chromatin,
irregular nuclear membrane contours, inconspicuous nucleoli, and scant
basophilic cytoplasm (e-Fig. 43.45). Mitoses are frequently identified (Acta
Cytol. 1992;36:887).
B. Burkitt lymphoma. The aspirate contains a monotonous population of
intermediate-sized lymphoid cells with round nuclei, finely dispersed chromatin,
and multiple distinct nucleoli. The cytoplasm is scant, deep blue, and contains
small lipid-filled vacuoles (e-Fig. 43.46). Tingible body macrophages and mito-
sis are prominent (Cytopathol. 1995;12:201) and are indicative of brisk cell
turnover.
V. LARGE CELL LYMPHOMA
A. DLBCL. The aspirate shows a monotonous population of large atypical lymphoid
cells. The centroblastic variant contains centroblasts with irregular nuclear
membrane contours, coarse chromatin, and multiple small nucleoli (e-Fig.
43.4 7). The immunoblastic variant shows immunoblasts with irregular nuclear
membrane contours, open chromatin, and single prominent nucleoli. The T-cell
rich B-celllymphoma variant is composed predominantly of small mature lym-
phocytes and scattered large immature lymphoid cells with polymorphic nuclei
and prominent nucleoli. The cytological diagnosis of this variant is challenging
(Diagn Cytopathol. 1998;18:1).
708 I SECTION X: HEMATOPOIETIC SYSTEM
SUGGESTED READINGS
Immunobiology, (6th ed.) New York and London: Garland Publishing; 2005:149-153.
Swerdlow SH, Campo E, Harris NL, et al., WHO Classification of Tumors; Pathology and Genetics;
Tumors of Hematopoietic and Lymphoid Tissues, 4th ed. Lyon, France: IARC Press; 2008.
Bone Marrow Pathology
Jeffery M. Klco and John L. Frater
I. NORMAL GROSS AND MICROSCOPIC ANATOMY. The bone marrow is generally consid-
ered the fourth largest organ in the human body and is composed of cells derived
from a variety of lineages including stromal cells, adipocytes, lymphocytes, and
hematopoietic precursors. The most frequently sampled areas are the posterior
superior iliac crest and, much less frequently, the sternum and long bones. The
bone marrow has an orderly microscopic anatomy. The most superficial part con-
sists of a layer of dense cortical bone with an adjacent cover of dense fibrous
periosteum. Deep into the cortex are the bony trabeculae, which consist of thin
trabecular bone and the marrow cavity itself. The marrow cavity contains islands
of maturing hematopoietic cells with intervening areas of fat, the latter of which
increase with age.
The cellularity of the bone marrow is defined as the percentage of the marrow
cavity composed of hematopoietic cells. In biopsies from the posterior iliac crest,
marrow cellularity decreases with age, and is expressed by the formula: Marrow
cellularity= (100- patient age)% ± 20%. Thus, a 50-year-old individual would
be expected to have a marrow cellularity of approximately (100- 50)% ± 20%,
or 30% to 70%.
Under normal circumstances, maturing myeloid and erythroid elements occupy
different regions of the marrow cavity. Myeloid precursors lie adjacent to the tra-
becular bone, and erythroid elements form "islands" of cells between trabeculae.
The ratio of myeloid to erythroid elements is roughly 2:1. Megakaryocytes are
irregularly distributed throughout the bone marrow. Under normal circumstances
they are not present in clusters.
It is important to be cognizant of the multidisciplinary nature of hematopathol-
ogy. Diagnoses in bone marrow pathology are not generally the product of mor-
phologic analysis of the bone marrow core biopsy or peripheral blood and bone
marrow aspirate smears alone. Clinical history is extremely important in sepa-
rating morphologically similar diseases and should be provided by the patient's
physicians. Also, the pertinent features of the physical examination, such as lym-
phadenopathy, splenomegaly, and hepatomegaly, are of importance. Other clinical
laboratory information, such as complete blood counts and serum and/or urine
protein electrophoresis, are often of interest. Radiographic data are of importance,
particularly in the evaluation of a monoclonal protein.
II. GROSS EXAMINATION AND TISSUE SAMPLING. The same pathologist should review
both the bone marrow aspirate smears and the core biopsies whenever possible to
avoid ambiguities or outright contradictions. The bone marrow aspirate is generally
performed before the biopsy. There are two kinds of aspirate smears: smears pre-
pared directly from the specimen without pretreatment and smears prepared from
concentrated aspirate fluid. Concentrated bone marrow aspirate smears and touch
preparations prepared from the bone marrow core biopsy are particularly useful
in the evaluation of specimens diluted with peripheral blood. Three to five smears
are stained using the Wright-Giemsa or similar technique, one is stained for iron
using the Prussian blue technique, and additional unstained smears are reserved
for ancillary techniques such as fluorescence in situ hybridization (FISH), if nec-
essary. Although examination of bone marrow cells with enzyme cytochemistry
is likely to be rendered obsolete in the coming years, its use is still highly recom-
mended by the World Health Organization (WHO) committee for the diagnosis of
709
710 I SECTION X: HEMATOPOIETIC SYSTEM
acute myeloid leukemia, and high-quality aspirate smears should be reserved for
this purpose when an acute myelogenous leukemia is suspected. Additional bone
marrow aspirates are obtained for flow cytometric, cytogenetic, and/or molecular
genetic studies. Aspirate smears are generally reviewed using high power (600x to
1000x) and are important for evaluating individual cell detail. However, because
the process of aspiration disrupts cell cohesion, the relationship of the various cell
types and the marrow cellularity cannot be reliably assessed. An adequate bone
marrow core biopsy adds this important information.
Ill. DIAGNOSTIC FEATURES OF COMMON BENIGN DISEASES. The number and scope of
nonneoplastic bone marrow disorders are vast. Emphasis is given to commonly
encountered bone marrow diseases and conditions that may simulate neoplasia.
A. Megaloblastic anemia. It is often not necessary to perform a bone marrow biopsy
in patients who present with anemia, because the most common forms of anemia
(iron deficiency, megaloblastic, and anemia of chronic disease) may be diagnosed
by laboratory analysis of the peripheral blood, clinical history, and response to
iron, vitamin B12 , and/or folate replacement. Bone marrow biopsy is performed
for patients with anemia that is unexplained or therapeutically resistant.
It is important to note that megaloblastic anemia may simulate a neoplastic
condition, particularly a myelodysplastic syndrome. Patients with megaloblas-
tic anemia may present with marked cytopenias and pronounced dyspoiesis
(e-Figs. 44.1 and 44.2).* Clues to discriminate this benign condition from a
myelodysplastic syndrome include normal blast percentage, absence of kary-
otypic abnormalities, and improvement of cytopenias following administration
of vitamin B12/folate in megaloblastic anemia. Also, the degree of dyspoiesis in
megaloblastic anemia often exceeds that encountered in most cases of neoplastic
myelodysplasia.
B. Benign causes of lymphocytosis. Numerous benign conditions may cause a tran-
sient increase in benign lymphocytes in the peripheral blood and can simu-
late chronic lymphocytic leukemia/small lymphocytic lymphoma (CLUSLL),
T-celllarge granular lymphocytic leukemia, or other lymphoid leukemias. Stress
lymphocytosis is a transient increase in morphologically normal peripheral
blood lymphocytes encountered in individuals subjected to physiologic stresses,
including individuals presenting to hospital emergency departments. An abso-
lute increase in lymphocytes accompanied by "reactive" forms with increased
basophilic cytoplasm and inconspicuous nucleoli may be seen in patients infected
with Epstein-Barr virus (EBV, e.g., infectious mononucleosis) or, less commonly,
cytomegalovirus or other viral pathogens. Interestingly, in cases of EBV infec-
tion the virus particles are present in morphologically normal lymphocytes, and
the reactive lymphocytes represent cytotoxic T cells directed at the infected cells.
Correlation with serum viral antibody titers is useful for arriving at the correct
diagnosis and for avoiding unnecessary procedures such as bone marrow and
lymph node biopsies. Many other infections are associated with lymphocytosis,
including pertussis, in which the lymphocytes have characteristic clefted nuclei
and thus may simulate peripheral blood involvement by a non-Hodgkin lym-
phoma. Persistent polyclonal B-cell hyperplasia, as its name implies, is often of
longer duration than other benign forms of lymphocytes. It frequently affects
women who smoke, and is associated with the human leukocyte antigen (HLA)-
DR7 phenotype.
C. The granulocytic 111eft shift." Other cases of leukocytosis represent a granulocytic
shift to immaturity (colloquially known by the archaic term "left shift" that
refers to the traditional placement of immature myeloid cell percentages to the
left of neutrophils in classical peripheral blood smear reports). Because many
*All e-figures are available online via the Solution Site Image Bank.
Chapter 44 • Bone Marrow Pathology I 7 11
H. Granulomas may be encountered in bone marrow core biopsies and are occa-
sionally noted in aspirate smears. Granulomas may be quite subtle and are
usually composed of admixed histiocytes, lymphocytes, and plasma cells. Some
granulomas contain foci of necrosis and infiltrating neutrophils. The causes of
granuloma formation in the bone marrow are similar to those in other sites;
the most common etiologies are infectious, autoimmune, or idiopathic. In addi-
tion, granulomas are occasionally encountered in the bone marrow of patients
with Hodgkin lymphoma, although their presence is not indicative of marrow
involvement by disease. Because it is impossible to predict with certainty the
etiology of bone marrow granulomas, their presence usually warrants the use of
stains to aid in the identification of acid-fast bacilli or fungi. However, it should
be noted that special stains are far less sensitive than microbiologic culture in the
detection of most infectious agents in the bone marrow, so microbiologic anal-
ysis of fresh bone marrow tissue is recommended when an infectious etiology
is considered. An important item in the morphologic differential diagnosis of
granulomas is the lipogranuloma, which is generally considered to be nonpatho-
logic and consists of a collection of histiocytes and lymphocytes surrounding an
area of fat demonstrating microvesicular change.
I. Benign lymphoid aggregates are present in the bone marrow of healthy individ-
uals and at increased incidence in elderly individuals. Because they are com-
mon, it is important to recognize the attributes of benign aggregates and dis-
tinguish them from their malignant counterparts. Benign aggregates are often
well circumscribed and are small to medium sized. They are composed of an
admixture of cell types including small and mature-appearing lymphocytes (typ-
ically CD3+ T cells), histiocytes, and granulocytes. They frequently contain a
central small-caliber vessel. Although they occasionally abut bony trabeculae,
they do not demonstrate the paratrabecular pattern of growth seen in follicular
lymphoma and other non-Hodgkin lymphomas involving the marrow. In some
cases, immunohistochemistry or in situ hybridization forK and).. immunoglob-
ulin light chains can be used to further evaluate aggregates.
J. AIDS. Since the first cases of AIDS were reported in 1981, a number of associated
diseases have been identified in the bone marrow. Commonly encountered mor-
phologic changes in the bone marrow of human immunodeficiency virus (HIV)-
infected individuals include granulomas, lymphoid aggregates, plasma cell
aggregates, and dysplasia in one or more hematopoietic lineages (Haemophilia.
2001;7:47). Since the development of combined drug therapy, the incidence of
secondary infections has decreased. However, infectious agents are still encoun-
tered in the bone marrow of individuals infected with HIV and, because impaired
inflammatory responses are a hallmark of HIV infection, it is recommended that
all bone marrow biopsies from patients with HIV be examined with special stains
for fungi and mycobacteria.
Lymphoid aggregates occur with increased frequency in the bone marrow of
HIV-infected individuals, are sometimes large with ill-defined borders, and grow
along bony trabeculae; these are all features suggestive of malignancy. They may
be extremely difficult to evaluate, especially in view of the increased incidence
of non-Hodgkin lymphomas in this population. Ancillary studies may be useful
in the evaluation of monoclonal B-cell or phenotypically abnormal/monoclonal
T-cell populations.
Dyspoiesis is a common finding in the bone marrow of patients with
HIV infection and can be seen in other viral infections such as hepatitis C.
The significance of this finding may be difficult if not impossible to inter-
pret because of the increased incidence of neoplastic myelodysplasia and
acute leukemias in the HIV+ population. Blasts are not typically increased
in HIV-associated dysplasia, and clonal cytogenetic abnormalities are not
present.
Chapter 44 • Bone Marrow Pathology I 7 13
A. MICIIIOI)oaec••~
D._
HLI1er O!llld""""
(m~lldoolt!Jpe I H)
Scllo1&"1JJdrorne
(m~ octll.llldoslsl!l>t IS)
HLrlor-Gdldo ~rno
(m~llcl"*"" I H.s)
H1111Br llftldromo
(m~Jid. .""" Ill
_l_,po O!llld"'"" type A
(m~ 1tdl.llldoslsl!l>t Ill A)
Sanftl_.po syno~,..,. type 8
(m~lld... "" 1118)
SWI_,DO O!llld""""IYI>O C
Cm~lld. .""" Ill Cl
_l_,po syna"'"" typeD
(m~lldoolt !Jpe Ill D)
Molq""ll(lndrome lypt A /Ma!INII.eclolamlno. 6-oulflla suffllasa
(m~llcl"*"" f'l Al
Molq""ll(lndrorno lypo 8 ji-CIIadoolcluo
(m~Jid. .""" f'l 8)
Ma.-.....Umy~e ~lo<bamln~
w
Sly.,..,.,.,.
(m~lldoolt!Jpe
~!ucuronid.e&e
em~lld"*""~o
.......
t. UJIII-..~Ddllt
c:...:Nrd-
II'IIIM dtl'lclloiiiJ
ji-GI""""'reb,_,..
'kfl••ce..,..
Glu"""""-
Nionwlr>-l'lctdiNaa Sph~ Splllnp>)'Oin
Gqblibla
TI)"&<NdiMuo
Sln<llofl'd -
~
-m-.llossA
-rn-.~~oo&A
GM> ..,.,.lela
GMIII*'Jifooklt
FII>IYdhoooe
-~~- ... QM> lllll'l.llblldo
()nmldo1111<Mooo
716 I SECTION X: HEMATOPOIETIC SYSTEM
DlleiA
htioiold and lfmphotl
-lumowlth
'-••liiii
Fll'1LH'IXiFIIA "*» 1D
<mlile dolollon II
II Cllrbl
M > F;wldo«..IIr«JW~;
Mpa, aplemm~
ll'lo LJl DIIIIIJ
~oohlle Mil ordertt
rrdl-;n..t ..lk
""""'*""
lCD II-Ia
UnCX>IM'IOI\M«Y
be..WLO<'T.u!L
'"····
(•••"'11>
I..UIIb)
I.Jcolf 111>011 r 11\&ro
lllu'IOo!pn
POGF'RA 4q lhi*h may Mod '"'IIMIM ofhi!tl 4rlll f~MUd In ban& don-from
roqulre RT.f'CR or IL'IIt-, ~\r
"""'-'~·
!Unlnpl'l'llnt llf&l\eoslnophl
FISH fDr.W.....,) COliN (>1.5" !G"IU
ll'ddi'C INfiU& dncl
mlma <hronl::
oooiiiOPhl., llull.omlll -~
Clll'dlll<falJre
-pUna-
~ld and lymphoid EIVSJ'D(;F1!8 ,. > ~ w1tte q&l'dJ\II8o; ~lypon:olJ~r boM Ra" typ""!lf I.Jcolf 111>011 r ttw..
-
I«S; 12l~l; pl2)~ --non~•l\d """""with myeloid llnoo!pn
I'OOfll8 .., b6dot.ocbd by lt'*'>qlool&, h:ILIIIn, !P'INI\oqlll: dal•~t~~~trom
,..,.._.,.rt ""'""""""'alii pl'ldomhlnce~ llfat\eoslnophl
ZNFliiB-FGI'f/1
anopll!ls; mlml::t
CMML
M >F. me<IIM'IIO~».
,.,_
lncbiii'C eosinophil,
-pll!ll::
JlrDQ.II!IIOr$ ~ nd
Poor (no)
""lillbl& dina I 001l110phllll to
-~--
lt(8; UXpll;q12! mla!d phon<ilyp&
R;Flllol>normdlloio
(aloo-NBpll
OJI'd....)
rnc:tltoommon;
n LI'TIII'Qota w.rle rD
ldonllllod
-1\edlro,. -w!l, CO-£. Kemo Nl.<i.._, ..,._ WHO- of
tam~
prMintlrtSon; t.d:ln.lhla
MIRmod...JYd-
llll)'bopll!OOflt
T~p""blullc
lymp-
(i.e., with blast percentages of at least 20%) and are broadly categorized as
myeloid or lymphoid according to their immunologic and enzyme cytochemical
properties, the characteristic features of which are summarized in Tables 44.5
and 44.6. There are two important exceptions to the requirement of at least 20%
blasts in acute myeloid leukemia. In the WHO classification, the presence of
t(8;21)(q22;q22) [AML1-ETO], inv(16)(p13;q22)/t(16;16)(p13;q22) [CBFfJ-
MYH11], and t(15;17)(q22;q12) [PML-RARa] is classified as acute myeloid
leukemia regardless of the percentage of blasts (e-Figs. 44.13 to 44.17). The
second exception involves acute erythroid leukemia, for which blasts comprise
at least 20% of the nonerythroid cell population, rather than at least 20% of
all cells; because another requirement for acute erythroid leukemia is that at
least 50% of all cells are erythroid precursors, cases with very high erythroid
percentages may qualify as acute leukemia even in the presence of a very modest
blast percentage. Immunophenotypic analysis of acute leukemia is discussed in
Chapter43.
Acute leukemias, particularly in children and the elderly, may present with
isolated anemia and a lack of circulating blasts. Diagnosis requires detection
of at least 20% blasts in the bone marrow and/or the presence of an acute
myeloid leukemia-specific cytogenetic abnormality. Although replacement of
the bone marrow by fibrosis, carcinoma, sarcoma, or other nonhematopoietic
cell may result in anemia, it is generally accompanied by leukopenia and/or
thrombocytopenia reflecting indiscriminate displacement of normal marrow
constituents.
1. B-lymphoblastic leukemia is arbitrarily separated from its tissue analogue,
lymphoblastic lymphoma, by the presence of a tissue mass and :::;20% bone
marrow blasts in the latter (e-Fig. 44.18) and is no longer subdivided on
the basis of immunophenotype since genetic features are more predictive of
outcome. B-lymphoblastic leukemia is most common in children <6 years
old; patients with purely lymphomatous disease are somewhat younger on
average. Most cases express HLA-DR, terminal deoxynucleotidyl transferase
(TdT), CD10, CD19, CD24, and cytoplasmic CD79a. CD10-negative cases
often demonstrate rearrangements of the mixed-lineage leukemia (MLL)
gene and coexpress one or more myeloid-associated antigens. In the pediatric
population, cytogenetic and molecular genetic findings are highly predictive
of disease outcome. Predictors of good outcome include the following: age 4
to 10 years at diagnosis, hyperdiploidy (51 to 65 chromosomes) in the blast
population, and presence of translocation t(12;21)(p13;q22) [TEL-AML1].
Predictors of poor outcome include the following: age <4 years or > 10
years, hypodiploidy, t(9;22)(q34;q11.2) [BCR-ABL], t(4;11 )(q21;q23) [AF4-
MLL], and t(1;19)(q23;q13.3) [PBX-E2A]. The prognostic significance of
t(1;19) is controversial. Recently, mutations/deletions in the lymphoid tran-
scription factor gene IKZF1 (IKAROS) have been shown to be associated
with a high rate of leukemic relapse and a poor outcome. Mutations in PAXS
are also common.
A special category of lymphoblastic leukemia is the leukemic analogue
of Burkitt lymphoma. Patients with this disease commonly present with
an abdominal mass (Western Europe and North America) or a jaw lesion
(Africa). The bone marrow is commonly involved, and the blasts have deeply
basophilic cytoplasm with many lipid vacuoles, express CD10 and mono-
clonal surface immunoglobulin light chain, and are generally CD34 and TdT
negative (e-Fig. 44.19). Diagnosis requires demonstration of the transloca-
tion t(8;14)(q24;q32) [MYC-IGH] or, less commonly, the variant transloca-
tions t(2;8)(q11;q24) or t(8;22)(q24;q11) involving MYC and the K and A.
light chain genes, respectively.
.....
,. DilliN
A._.....,. _ ..... CIIIICII
,.lll .... lllc ... ___
-MIDJf .....,....,.
k>bln'f'llold 1111/loltrole Mil Often - M i l Blu1* Mllloi'C lllander J\uar CD~. CD$+, MPO+. F""'r«blo
t!S;21l!<!22;<1221: G«<011-..IIIY<hlo.10 roda. «bl'lOITMI.croniiCicll C()~,CD$4+,C056+
(RIJNXUIUNXlTCJ
kibln'f'llold ~~~~~-Mil Qcc.w._"\dlf pres,enb ..t:h Abnom\11 eoolrq>llll& w!ll\ llllfll CDIS+-, CDS$+-,IAPO+; F...,ralll&
tbnormll ~ nwraw fiiCbahiGJIMy <It sesa basophile IJ'AIII. ., - troquonii)'CD4+, all4+,
-lfllslrw(l6)(pl3q22) .. lobotlon CDII~.CDllo+.C~
ttl&;lli)(pl3:q:i2) C036+.~
rtlBF,IIrl'tHllJ
kib 1ft>~ IMJI<.emll c.q,\ojllllly, """"U'IawWBC Abnom\11 prom,..\oq!M Mil co~ (llelo__.a!, con. F...,r«bl&
CAloR 10!1111(15;17!(<122;ql2);
(PN!AWila one~ .._,
klb ..,.!old~~~~~- Mil
(l!y_..,u~rVINJil;
WBC ~"*""llon1l
hl!tl
F~ca:~nln dllldren
m\Oifpl!o A"" rod& ptedomlnd&
lllo,.,.,UC - fi'OIIOmN11
(lr1.!H), HLA-DR-, COS4-
..,._,....
CDlie+, C064+, elm+•
.__..,..... -·--~~-..lool·
I. fdlowii'C a ln'f'l~luCio oYIIdrom& Cl.ltl$l or ~ld>'rn~~~~dklordor
2. I.IDS-nolobd e,t:el- •lln-ltj
!. l.lull ....... d)OI)i&IM ~I&IM In . t - SO'l!. ofh-ln!oo> eollll,_,
e. ..................... .,.. ,......,.., ,tiiiC.., lilllltlwl
• AI~ ...rt...llled; typlco!ly hu• >10,... lotlnq!wttl\ toMOS ....
• ~~II _,hlbloHalaD:I (tome ma:f ba tlfnphold); 1~ lllbio<;ywttl\ no t-MDS phooo. """'""""""' ba~,_,,....-,.ln<:lud~
l!Q~•tmm.llm
l._.....,...
• OIIwMM
._laNII-CII:Ji•IDII
Aclto rnyolotllolloltr:le UollltJ - I n d:.lil>ood, -dlt ol ~ MPO+. dlt o! Ofton CD13+. co~. CD117+. u,.,.,.,bl&
mblmatJcltiW•:d:llad cytoc,oilll:& bhbto NBE+ C())4+, Cll31k-, HLA-DR+
Ai:lb rnyolotllo- - I t UollltJ p-In ad :.~~hood, ..mpr~sa ~ o1
s~~.m Ol!on C013+, co~. C014+, Ullk..,nlblo
IN1:ntlcn qtopolniM, - ! I f..til no!101)11mll "'*; :!:!!!'. ol C0117+, MPO+
mlfk:lcl.,.lna : cf W'8C blull MPG+, ,;;?."of blalll
NBI".t
N.bi!I)'O!iotlla-""" Vorllblo""r'"'P•rol ~ ol bl:lsls MPO+. ~ ol !Ju:llfCD13+, CD~. COlli+; Vllllablo
mav0!11on ~I!IO~io&Y bhbto NBE+: >l'"' ml:tll1rc Y>i:l::bD CD117+, CD34+,
~t~enu~cel& HlMlR+
Ai:lb rnyolom~ IMJI<.eml! AMml!, r-. ~,wac >20!i.blodtO..,....rc U.U.IfCDI3+, CDS3+; 01lan Vuleblo
....I t t - prornoncqte.s); ==~ CD4+, C014+, COil b+,
monocytes. aI'd PMCUJ'SOI'I; CD!l'*' C064+, C0!6+,
llJid ~ noulropl:llo and tfiOZlmO+
P"""'"""" !:3l!'. ol biMI>
MPO+. !:3!1'. o l - "'""!If
NBE-+"
Aclto m~ il<lk.eml! Moot...,mon lndikhn.., ~ ~C>IIIh. olwl:lell V•:tellD CD13+. Cll33+. U~l&
p---"""*'IMy
d~ l:l:!edlrc d~son~
...
~~ arw; mono~ ....::20%
-pi:!~ and PlliCU/liOI'Ii
CD117+; Ol!on C014+,
CD4+, COlli>+, COlle+,
<3~ oT blab MPO+, !!3~ of CD64+, C06&t, CD36+,
bl:lm NBE+ ~
N.bm~l!umlo -common ln...,b. ollan ~~ ..lb. olwl:tl: V•rlablo COI3+. Cll33+. Unf-ru>lo
p-wtll\-tnll(lllfliiY 111o mo)o:l!yer& 1>10~ C0117+: Ofton CD14+,
dr-,l:l:!edlrcd~am~ ... <20% noWophlund CD4+, COlli>+, COlle..,
•ou....m.-... litel-ln AMLincUie c-«<T, H-IlA$. K-IIAS. WTJ, fl(fl{ll.f n1 pe11111- dupl:llion of NU, "'"" m -
1':"1""*tll)l!l:o""' 1111!diD l>e cldmnh:d •IIIII" cohO!tof AML pe1len1o. '"'root""""'10rANL end-
Atulel1l!<i:ll:lleu""""'""'- -lerd C<lii'Ae"'p"""-...- In WI«) (20011.
'IDHJ,/D/12., D - end m:~...-.. h..,. eloo been litel-lndllet m)'dold mel[tnoncb.
~1n>m.ICC.20ll;a47S-486:-~Il:ll&~ .
....
726 I SECTION X: HEMATOPOIETIC SYSTEM
(1•"'"'bit)
Cllilll± (u-.,blo H+1, ponlnobculllf) 17p (ll'S3) clo-
Z.p70 :1: (unf...,rlll>lo H+l (111fm><oblol
Mal'llo ... b'rnPIIoiNI CO&+-, CI>IO-, COl~. C02!>+ U.Witanoll lim~""" ~ 11;14l t8CU -Iaiii
(b!W\1l, Cl>lt)-, FMC7+, cleftodll)ldad ~ dtllloa.
Cl77il>+-, siJIWI! dlah+ 1\ttdi.Mr. lrihw~ or
(WaleIf bo11J11!, qcln Dl+ ~beo::ollu pol!Mno a!
MlmlW-
Fol...,r tymph>mo CD!;-, CI>IO+, COIH, C020+, VIll1oblo "'"""''l'holoilr; dl'fllll, ~14;18) fJlCL2o/GHI
COZI-, '" 111111 <"'*'+ nodiAar, 1~ or
ponrtnobo<:<llllf palbifm a!
M«JJ'CW~
MarltlllZIII» lk>ell COh Cl>lo-, COI9+. C020+, Smlllll)m~. -with ~ll;l8l(Q21:<121l
tymph>INI COl!!-, '"llgttehMI+ omplo~ !API2..uou:rn lclon1Hitd 1n
·-<tf-...,11
""'"*"'"""" tympmm•
LJmplql~ C06-,CI>lll-, COIH.C020+, Smaii1Jm~,....,- t!9:141(ql3;1j32)
tymph>moiW<Itlomlnlm COZ3-,I- illlht<"'*'+ plosm~ lill1lnl ~~ tloi'AIIod ••
~I>Uihomll ~tA-bla II not
llmbd 101llk""" a1
""'"'"'""'
..... tiJ, _,...,.......,....lin; II. hmu-lln.
F"'"': -lowS. C.. E. lllnto Nl.dtL, ods. WHO_, of liRr>ol.l'odHo""'fojootlloM!d ljql>p/lold-., ~ IAJ\C -lllOII. Uoohlh pomhom
728 I SECTION X: HEMATOPOIETIC SYSTEM
I. NORMAL GROSS AND MICROSCOPIC ANATOMY. The spleen is the largest lymphatic
organ. In a normal adult, it weighs 50 to 250 g. Anatomically, the spleen is divided
into white and red pulp, separated by an ill-defined interface known as the marginal
zone (e-Fig. 45.1A).* For a schematic of splenic architecture, see Figure 45.1.
A. White pulp. The white pulp consists of periarteriolar lymphoid sheets (PALS),
which contains lymphoid follicles. T lymphocytes are predominately located in
periarteriolar lymphoid nodules and B lymphocytes are predominately located
in the lymphoid follicles. The latter may contain germinal centers that become
visible to the naked eye when enlarged, forming splenic nodules (malpighian
corpuscles). In routine hematoxylin and eosin (H&E)-stained sections, the white
pulp appears basophilic due to the dense heterochromatin in lymphocyte nuclei
(e-Fig. 45.1B).
B. Red pulp. The red pulp has a red appearance in fresh specimens and in histologic
sections because it contains a large number of red blood cells (e-Fig. 45.1B).
It consists of splenic sinuses separated by splenic cords (the cords of Billroth),
which are composed of a loose network of reticular cells and fibers with a large
number of erythrocytes, macrophages, lymphocytes, plasma cells, and granu-
locytes. Special endothelial cells that express both endothelial and histiocytic
markers (known as Littoral cells) line the sinuses. The sinusoidal lining epithe-
lium is discontinuous, allowing for transport of blood cells between the splenic
cords and sinuses.
II. GROSS EXAMINATION AND TISSUE SAMPLING
A. Biopsy and fine needle aspiration cytology. These procedures are rarely attempted
because of the risk of hemorrhage and the likelihood of undersampling. How-
ever, some studies suggest increased chances of a definitive diagnosis when fine
needle biopsy is combined with flow cytometry, with an overall accuracy of 91%
and a major complication rate of< 1% (Am] Hematol. 2001;67:93 ).
B. Splenectomy. Trauma, staging procedures, and surgical convenience account
for >50% of all splenectomies. Therapeutic splenectomy for known diagnoses
(idiopathic thrombocytopenic purpura [ITP], chronic myeloproliferative dis-
orders, lymphomas, etc.) accounts for most of the remaining cases (Cancer.
2001;91:2001). Unexpected pathology is rarely found in splenectomy speci-
mens, but significant splenomegaly (weight >300 g) or localizing lesions warrant
careful prosection and ancillary studies.
C. Processing. The spleen is weighed, and its outer dimensions are recorded. Hilar
fatty tissue is removed and processed for lymph nodes. The capsule should be
described, noting texture and intactness. The spleen should be thinly sliced (every
2 to 3 mm); lesional distribution should be noted, followed by a description of
the uninvolved spleen. Sections of any lesions (preferably following overnight
formalin fixation of thin slices) and two representative sections of uninvolved
spleen should be submitted for microscopic examination.
If clinically indicated, fresh lesional tissue in 1 mm pieces should be placed
in RPMI medium and directed immediately to the flow cytometry lab with
instructions regarding the appropriate protocol. Cytogenetic studies may be
*All e-figures are available online via the Solution Site Image Bank.
729
730 I SECTION X: HEMATOPOIETIC SYSTEM
sc - .
FT- F
TA-l
TV-.
LN -I
PALS
WP-
RP - I
S- Si
CB - '
Figure 45.1 Diagram of the spleen showing important anatomic landmarks and&- and T-cell
distribution.
b. '**""" _,_,.1"1
•• llt&dlo1ion
c.lmmunoou-lwlqenll,ln-na-rold&
aEn-dllonlors
a.~bm
b. liYI>®IIIIbJtorn
•. Die- rrwlltuo
-4. Cllronlo elcollollam
Chapter 45 • Spleen I 7 33
Usually benign incidental findings, their clinical importance lies in their potential
to mimic neoplastic and nonneoplastic splenic lesions.
IV. REACTIVE SPLENIC DISORDERS. These can be divided into diffuse and localized pro-
cesses. Diffuse disease entities include reactive lymphoid hyperplasia (e-Fig. 45.7A),
follicular hyperplasia, and disorders such as Castleman disease (see Chap. 43 ).
Localized disease includes granulomatous disorders and infectious processes simi-
lar to those in other locations (see Chap. 43 ).
A. Diffuse reactive processes. Reactive lymphoid hyperplasia in the spleen may
occur with or without germinal center formation. Because of the reactive nature
of the latter, and its histologic lack of maturing germinal centers, this entity is
variably referred to as "activated A," "early activated immune reaction," "reac-
tive nonfollicular hyperplasia," and even "immunoblastic hyperplasia" (Am ]
Surg Pathol. 1981;5:551). Nongerminal center hyperplasia is often associated
with viral infections, especially herpes simplex virus and Epstein-Barr virus
(e-Fig. 45.7B), which explains the common occurrence of splenomegaly in
patients with infectious mononucleosis.
Reactive lymphoid hyperplasia with germinal center formation (e-Fig.
45.7C) is commonly referred to as "follicular" hyperplasia. It is the most com-
mon pattern of lymphoid hyperplasia in the spleen and is seen in both acute
and chronic immune reactions. Follicular hyperplasia is frequently observed in
bacterial infection, often as an incidental finding. In fact, splenomegaly is char-
acteristic in subacute bacterial endocarditis, alerting the clinician to this process
in the appropriate clinical context.
B. Focal reactive processes. Localized reactive splenic processes can also present
with splenomegaly.
1. Granulomas. The most common form of a focal benign process is granu-
lomatous inflammation, ranging from lipogranulomatous inflammation (of
unknown etiology) to caseating or noncaseating granulomatous inflamma-
tion. Caseating granulomas are primarily due to infectious disease, including
tuberculosis and fungal infection; however, they are also seen in X-linked
chronic granulomatous disease. Noncaseating granulomatous disease is most
frequently associated with sarcoidosis (e-Fig. 45.8). For most granulomas
in the spleen, no known etiology can be found (Arch Pathol Lab Med.
1974;98:261 ).
2. Infarcts. The spleen is a frequent site of systemic emboli, which commonly
arise from cardiac valve lesions or mural thrombi. Infarcts are usually wedge-
shaped with a hemorrhagic to pale-tan to fibrotic appearance depending
on the age of lesion (see e-Fig. 45.9). Non-wedge-shaped infarcts arise in a
variety of intrinsic hematopoietic and nonhematopoietic processes. Essential
thrombocythemia and chronic idiopathic myelofibrosis are the hematopoi-
etic disorders most frequently associated with infarcts; less common causes
include paroxysmal nocturnal hemoglobinuria, sickle cell disease, and aplas-
tic anemia. Among nonhematopoietic etiologies, vasculitides (e.g., polyarteri-
tis nodosa, infections, and TTP/ITP associated) and splenic artery aneurysms
are common culprits.
V. NEOPLASTIC DISORDERS OF THE SPLEEN
A. Lymphoid neoplasms. Lymphoid neoplasms can involve the spleen as primary
disease or as a part of a generalized lymphomatous process. Splenomegaly is a
nonspecific but classic component of many hematolymphoid disorders. A brief
approach to the evaluation of the normal and neoplastic components of the
spleen is presented in Table 45.4.
1. Primary splenic lymphomas. These account for <1% of all lymphomas and
can of B- or T-cell origin (see Chap. 43). Not unexpectedly, B-celllymphomas
are more common, and diffuse large B-celllymphoma (which usually presents
as single or multiple circumscribed nodules of varying sizes) accounts for the
ClonoraJ Guld&IN&lO Ewluota
lhostran.l Ccmpollmont
Goonoratt•~ 1 M -
blood""
ean-lltoo•- of
rl, moM<,1illlllll<ta"'- and -1111c
Qllb. r..,th«-compllo61ho "'I- unr of
'llloon and n ...,,..,._ of•Cill'doof St-.•
Al Vuoulor Ofldotllelol ...lo Cll34. COOl, F -VIII relotad 011111on ere uoo!UII'I
hW\flill1hl: boCIIIIOITI'lOI 01\d llflOI:Io* ........
Lll!nl.,.lk Sllara...,.r•<>lbolll endOIMI~t-..,d
1'11~1'111l.a'tl~ ElprM.& C08 uttlform~
<:068, ......,_,and anll<llym~ . . woflrl llohl.
WllonNuolb1oflnhdltorrnllarlole<lnloclloua
orprrllmlll OOIIrod, 1'>\S, G"'m W*!, AFB. Jl\d GMS
otol'lo con bo Uled.
Tw<> mlj<r~ lnta~n' ODCl and folleu~r (FDC).
<:068, SlOO, COl•J ~llJ 121 ...t~ rn.qah•
"""bit poo!Myln dllndlti::-.
FOO& . . COOl+ al.15+; lOCo ara SIOO+.
Galofll GWclolnn10 E¥11- In-~ mot\ IJmphornos 1.-111o wlllto pr.lp
111& &- tnd T...,.l ~Didd
Oornpo/lmonto
(hodulo//lollcUar loW-·-"'""''·
. . . . . may P-.t.. di!I'Uoe""IID DUIP ~.
ba-...M
--ol.
II) CoWd 11111; be a m-.olat •I 0045, l f - l o root-..,. tlmplrold.
b) Ia ~a e. or T..,.ll.......-1 b) 9~CD21), CD?Ill) lind T~COS,C045RO) eel llWMra
•ra • ......,.. uliiCI .,_ ta1ceft _,.,....number •nd
llt.
<:1 Allarrant T-edI mlll<.m. <:043, ancl!b< C05.,. ollon
...-.
d)An>ll>o&e_lra1.,or <f) Bd-2 Ia portlcutorly lrolpflrlln dllnnUol d~of
~llwlo/ b'rn"""""' 011d I'08CttJo ~llwlor lt/liorplt>M.
S...lln eiTnlnol """'"' raldllh&col*tYto undo~~~>
•pol*>* and do root _..1M antlopop~OBII ~.
~. Al>out 110!1. Gf follcrJor t~mplromu --llcl-2
lnlhofollcliD.
0) Haw-tlll>o tlmplrold o1 Met lioNI B· •hd r....u m'"'""" rrwt bo usod to furllt"
"'""'"II<Y be oubtYI>od? clloracUnze t>llenottl>6of""llouo llmrilorn .. (Ooo•loo
ClloP. ol3, T®loo o13.3 01\d o13.4l
s.mr. er.mp~oooro In -~ dll«dool ..til lallll"""mpon.,.. of ,.,uld\&
wllll Pnodomnontly Rod col& h - mon><ocl..- red pulp 1.-mert.
Pllpln\IOt.wnonl
I) HI'Yoolloull!mta
bl Hapodooplorrlc r....u
tlmplromo
TRAI'IndDI!A-44
~. C04-. CO!!-, ollan Cllli6+.
m~loo <TIA. fiOitlltt, pn~olll
-11 +, y$
Ill~
e) T·I.GL
d)T·PLL
o) k:UJ>Ioulamta and
mydopmllar- dlolu..,dams
""""--
CO$+,C08+
~.uauli'/CD4+
Forar•nu~tlcor mcH>0<,,1Sc:<:.&lb: MPO, CDM, allt7,
COI58
F« ~l<ll <Olh: Qtl<o>t>l>ctltt 01\d homotlol>l'l
F«"""'lca~: C041, CD4:CI>. CD61, F-VIll
ftJ' P"""""" tJrnplrolclloulc.emtat: Tdt, COi'!lt, CDS, COlO
Chapter 45 • Spleen I 7 35
Soft Tissue
John D. Pfeifer and Louis P. Dehner
If the term "soft tissue" were restricted to only mesodermally derived structures, then
nerves and neural tumors would not be considered in the discussion. Similarly, there
are other examples of "soft tissue tumors (SITs)" which are present in organs but
may not be derived from mesoderm. By convention, however, neural tumors, gastroin-
testinal stromal tumm; melanoma of soft parts, perivascular epithelioid cell tumor,
and so on are regarded as "soft tissue neoplasms" despite the absence of any evidence
of a well-characterized progenitor cell or mesenchymal stem cell (MSC, characterized
immunophenotypically by reactivity for CD73, CD90, and CD105) with the multipo-
tentiality to differentiate into a variety of tissues such as muscle, blood vessels, and fat
(Stem Cells. 2011;29:397;] Pediatr Hematol Oncol. 2008;30:301 ).
I. TISSUE PROCESSING
A. Biopsy specimens. lncisional or core biopsy of a suspected soft tissue neoplasm
is performed to determine the appropriate management based on the patho-
logic type. The biopsy tissue should be placed immediately into 10% forma-
lin or other appropriate fixative. The number of biopsy fragments should be
recorded, as well as their aggregate dimension, and all the submitted tissue
should be processed. Three H&E levels should initially be prepared for micro-
scopic examination. For very small specimens, in order to avoid wasting tissue
when refacing the block, it is strongly recommended that additional unstained
slides be cut from the block during initial sectioning in the event additional
studies such as immunohistochemistry are needed.
B. Resection specimens. Excisional specimens are often complex and varied, and
the macroscopic examination should be guided by tumor location, extent,
and type. The margin of all intact specimens should be inked, and the gross
distance from the tumor to the closest margin documented. The maximum
dimension of the tumor should be recorded, as well as the color and consistency
of the cut surface, and presence of hemorrhage and necrosis. In general, it is
recommended that one section per centimeter of tumor should be submitted for
microscopic examination (scout sections can be used to evaluate whether such
thorough sampling is required for definitive diagnosis). The closest surgical
margin should be evaluated by either shave or radial sections depending on
the nature of the specimen.
For those tumors in which a biopsy did not permit definitive diagnosis,
tissue should be collected and processed for electron microscopy. Considera-
tion should always be given to the need to send a sample of viable tumor for
739
'JCO I SECTION .... SOFT TISSUE AND BONE
I' I
1,1,! I (!-I\ ~~~~uc MHwlltlet a-c~e~~.uc of"'lllooa l1olt
Tl.a r~!~Pol ._lilt •IIIIIIIIIG a lad flriGIMol
_ , rhlbdoiT(jOMn:oma 1(2;13)(q36;q14) PAXJ.FOXOll'wlon
10;13l[l>36;ql4) I'AX7-ffl'(!)ll'wlon
-rdDI!ItWOOitlll d..O7ltOC; 17)(~11.2;<!2.Sl MPI.-17'D Mlon
Alljjoln.mld tbii>UI: t02;22Kqllt\ql2) EWSRZ..ATI'l1\.u!lan
hldSoq!Dma 1(2;22)~;q12) EWI/Sl.ciiEBl
102;16Kql'%qlll FUI'.41l'll'llllon
Al)lbll_.,niiiiDus ~l"ff\um""'Y nns•ndlor Am~ of MOM2.
tumw/ul :lff&ui!UI1Dd m-<ttrar.OIIOIIIOS CDI(4,H/116112p8t
.-....,...,. W.TNroLPSl lOith ampl~- of
12ql4-ql5
C!Mr oelun:orn• t02;22Kqllt\ql2l EWSRZ..ATI'l1\.u!lan
(mollnorna Dl tDII palll) 1(2;22)~;q12) EWSJ!l.cREBll'llllon
aon...,llllnfonlllo 1(12;15)(pl3;qlfi) EJW.Nil!l<3,...,
n"""""""""
Dodl'l&lllfldotod ll_r...,. ~-um•OIY ~nundlor
mill«< <II_,.,
lOith •mpl~- of
12q14-q15
-
O.niiiiDft-rama 107;22Kq:!21q13l•l'od COL!Al-P£161'B l'wlon
~bcnnlfPont ...n d_n,..n...,.,..
ftbloblulomo
Doomol:l ftlllrAnirlcoll Tr1oolny 8 .. 20: .... "'Sci Soma11c C'INNIJlor APC
mwtlona (only In deop
Ill moll)
Doomopl.ult:..,.l """" 101;22Ki>l<%q121 EWSRl-WTl fulbn
ot~lllllnv
Eloo$>ftblolna IQ allno!Tnlillleo UnknOWII
E'lnbi)»rroo L.ooo of ldow<a</Z!fPitt-t.et UnknOWII
rhlbdo-- llpl5; IIIN of 2, 7, 8,
II, 12, 20, 21, 19<121,
20;-ort~
7, ~ 9q22,14q21..:12,
17
~!lllalold hemlll!»- 10;3)(~25)
••do111..,.,.
8~-··-·"'
Epll:t\.ld MR:OI'nlllJ Ah/ilii<N; "'22qll.2
prad'liiii!Jpo h6NFSRN!l
__,,_
ilot1r U/QOOI illp/tnlllwl 101;22Kq24;q121 EWSJ!l-l'llll'wlon
nouro&~;tldaimeltumor 1(21;22)(q221q12) EWSRI-El!G l'wlon
~~~-··-·ot
Elc!nl11110lltiiii!Nilt AIB!Ilbooof22Qil.2
rhlbdold 1llnor hSNFS/IN/1
t(!I;22)(Q22;ql2) EWSR 1-NRIASfu&loll
chondi'Oiiii'CI:Ifnl 1(!1;17)(Q22;q11.2) TAF2N-NR4A3,...,
1(!1; IS)(Q22;q21) TCFl2-NR4113-
Gllntot~lllllnvof T~tnolol:1/l!omiiMIMIW CSF'lflllbl~ -k\,t
ollorlllld~ &1oM ll>l~.lnwdiiW CSFI-C014113
""lllllnva t(l;2)(pl~l3Sl
~m.-...y n.no-.r..IIM>Mre
mya11btol>lullet:Lmor 21>2$
a..~.46· Sottr-.e. I .,.,
I'1.1 II( !-f\1 ~:~c.r:~nuet a-c~e~~auc ot'llllo<a Soft
-n.orlJIII C)loalo,.~ ••~... •...u.-..
SW<:tu11111allol1!tiomof I,
7, 10. 13. 14
Reu..rwo-ofll<ll2
RMinll\lll-
....
R001t1181nlfllt <>I I'I.Ml
HMSA2andHIIIGA1
12q14-ql5 and ilslona; unlcncMTI
6p2142;d-!l
13ql2-14
i.oW-1111dotb~ 117;16l<Q33:Plll
u""""" l(ll;l£)(~11;pll)
Malpnt .-llf\e<OI""""'
t.t'IMth tumor
"'"""'ldlrwncl<d IU2:16J(ql3:pll) RJS.CDJT3futb!
llpolll""""" 1{12:22] (q 13;ql2) Ew.SRl.OOlT3,....,
Nooo!>hiMlliiOI (loN II IP, 7q, lOa, 12<1, ~m...._,l'l
lllllo4\b~ 1611. !tiel. 17q, 19~. CTNI{9J
21lq, 22'1
Softlloul-11111111 Tnonllio<>ltlorotl.-...lflna Ew.SRlfW>no\O!Ih •
tumor 22ql2 WJIIItyof cthlr•n•
SSKlii-SSlfJ, .s:sx:!B-
~~--
10(;18)(pll:qll)
S$1o2 $$XIB-SSJI4
ilolono
'JC2 I SECTION .... SOFT TISSUE AND BONE
UJIISI!t-
lollp
u-
Uoomow
Upo~•llotooll of,....
Upol>lu1rma.41po-
An&loll-
M)'aii-
Chonct'oldlpoma
E'lc!nl-r1flal•*m:ldi-
Elc!nHdiWIITI'.IOitil-
Spindle oollp"""'OI!JIIIc 1-..m•
Hlllmorn.t
-lllttloaiP.rqpoooho)
Al)lbii_,.INIIDustumOf--lpooa"""'o
......It
o.dllo!WoliDdll--
u--
~ llpauu:ou"""'"nd a!llpo~~,..,.,,
Ploom,.plllc llpoouama
M!Jal~l-mo
~~-....,.,......-1:1
IIIIP
...
110t OChenol8o C>dod
-~~~fuellila
Prolf--
Prolf-m)Ottil
~oalllcoM
Flbtt>-.a
lld>omlc-
""""'""'mor II dil!lt&
EIIoloftlmnl
Flb!out lllmoltomo II Infancy
M)'ofli>fon'6'rn:lollb.... -
F1-coll
JIJYM!Io ...,...... ftbllli'IIO-
~ bodytbll>lrt-
Fl-. aflon6on sh-
Ocomoplllll: f l b - •
Mlm . .!Y'lype m)IOII-•
COic&'JinUb'"IOIJIOCSc ~blr.lno
Anatou~brobiMtM'B
COIIJ~r IJl!jallbloma
Nudl.tlqpo flb"""1
Gardner flbroma
CllcfJina flbn>llstum"'
Glln!Q!IIInaloflbloml
I IIIIi lOII IIIt Ooall.r IJIJIIIII'I)
f>upelfte~lftbla- (polmulp~rml
Ocomolii~P"ft
Upoft-
a..~.46· Sottr-.e. I .,.a
'' f.t:] I (!-Ill WHO C&Mik&tJon of Soft 11at n.nca (fcrM*Mf)
-Crnl.r·-·"tl
Soaalyftlln>uotumor and llcml'*~
lntllmnWoly ll'l)"ftbn>blullc tumo<
Low-indo ml'lf\brobloo1ic ""'""""
tot..>l.-mt:IO!yftbrol>l&ade arcol!\1
•-nb......""'
lllllloool
Aclu~ n-,...,.
~-,..,.,.
Low-indoftb~ 01/~lrM\.c~lo col tumOf
.......
.... _loti,.._
~·ptilelklidft.....,_
-Crnl.r·-·"tl
DMc> b&n\111 ftbrouo. hlllloqtomo
Ploodfonn n-•llcqllt lllmar
Glontc:lllll tumor of tollllauao
lllllloool
Pleom01plile MFHAindlll,_ ~hie ..,..,.
Glint c:lllll MFHAindlllerwrtilllll plao"""'>hlc •ramo ..til pnl Clllls
·-·--...
lnftamii'IIIDiy MFHiund- plaornorplllc..,...... ..til pnrnlft01111nlllm1Ntt>n
AnaloWonTIOIM
DMc> loiom)ooiNI
Gtnlltl i&b'nJr10ml
Ltioii1)'0LI,..,.
..... ~
Olcmuo !UmOf
...,.lftO
Malinont alotrrlla 1llnor
·-·-"11-
....
""-"""""'"
Rhobdo"""'""' Ct4/l""". fiiiBIIYI>O.IIIIitel typo)
lllllloool
EmbljiOnal ltlahd..,_..INI tmblll& '!>Indio oel, lmyokl, •napl&ade)
A - rhlbdrlri)'OOircotnl
....
PlaomorpNc rhllxbli)'OII/~'1111
-.~~r ....
HomarWOmuda~b<Wn-p...rttllau•
C.plltry
eo.omo.a
Ario-n..,.
Ve.....
lntom~~J~C~Jittr
~·
Epltlotidhlma'*ma
An&m-ILA
Ljmpl\l ......
,.... I SECTION .... SOFT TISSUE AND BONE
--IIIIo
Rtd'onn INtiJ - - -
hOIIIIII\I.bllidollieilorno
Popl~ry liltrll.wTn~ ~lorn·
Com..,... h.,.nfllaonddl,...••
IC:Ifl(lll ..........
- ......
llllp.tlt
~!lllolold heml/liSOGIII:Iallellol!lll
Alijjou""""" o l d - ·
SilftU...O<hon<*'ome
Mooml:!iymll oliond"""'"""""
......... _ ..... 11111 . . .
-.ra
-~~~-
Ji.iidu_,., m-
o...,. (lui""'IIO) ...... _ .
Pleonlotlllilc ll';all'illln..~111n«
~h-uo11tJrnomo
........,.
~.,...., poilvuclllar apllid~ odl • - (PEtonwil
Clolr cdlli)'Om'*"""')tic tiiTIOf
~,.,
(hlllo!Qii tpootllc). n1lollc ~. 1nd -of
niUC -111. The FNCL<:C.,..S. ~~-- llythrM ponune!Ms: dll'lera1tlltlon
-.0111. ~ peramdllf ls...,.od:
dllar-n (1-3). mllollc adMif (1-3). end ,_..Ia (N).
~-1)1do.
Tho...,,..
uo !lllmmod to
Gro&XGruloOMinotb&e_...
Gro&l2tr$
Grldo24or5
Grldoa&-a
.......,..., TLmOr <II!IIUI- b """""'ljiOdllc end bl'notll~ ""'""'at -.•,
Sootlll S.l'tOI"'''M doistJly r81&1embllrw nomw, metuhl ~\altlllilu•
Scora 2 s........ of_.. -lo0>1!!1>6
Sccf'a 3 s,onc:MII ~ tmtr)a1.11.u.rmmu, undlft'•ilt'tllled urc:om~~, Jnd u~ of
unknownldoullti\11we
.lllllllft-ln 11M> moot mbCI!:otJ . . - . ,.. of tho"""""'., !811-M> hlat>-
fWda {HPft) m •••ud "*'£ • 4fit ob}ed:W.
Scora 11)..0 m-10 HPF&
Scora 210.1; m~O HPFs
Scora a20"' mora miDMif!O HPFs
,..,_,a rrAt EW~Iuat&d on ITOiltEU.Illll'liltkJn tnd w.ldltiDdwth hM'"'* w:t1on.s.
Score 0 ~tum« noe<*
Sootlll ~~tum«~
Sootll2 >~ t.rmor rtl!la'osk
746 I SECTION XI: SOFT TISSUE AND BONE
"'All e-figures are available online via the Solution Site Image Bank.
Chapter 46 • Soft Tissue I 74 7
visceral organs, and the central nervous system. Individual tumors range
from < 1 to over 7 em in greatest dimension and have a cut surface that
ranges from firm and fibrous, to cystic and hemorrhagic. Immature plump
to spindled cells are arranged in whorls and fascicles within a fibromyxoid
stroma; smaller nodules at the periphery of the mass may be associated
with a vessel(s) to suggest an angiocentric origin (e-Fig. 46.13). Hyper-
cellular spindle cell foci are present in some cases with a resemblance to
congenital infantile fibrosarcoma, but the tumor cells lack the t(12;15)
translocation characteristic of the latter tumor. Other findings include a
hemangiopericytoma-like pattern centrally, often with ischemic or hem-
orrhagic regions, dystrophic calcifications, and hyalinization. The spindle
cells express vimentin and SMA, whereas the hemangiopericytoma-like
foci are variably CD34 positive. The differential diagnosis of myofibroma
includes nodular fasciitis; the distinction can be problematic since the two
lesions have some overlapping morphologic and immunophenotypic fea-
tures (j Clin Pathol. 2009;62:236).
8. Angiomyofibroblastoma, a rare tumot; occurs in women of reproductive age,
where it arises in the rises in the pelviperineal region (vulva and vagina)
as a painless, well-circumscribed, slowly enlarging mass (IntI Gynecol
Pathol. 2005;24:26). In men, the neoplasm usually involves the parates-
ticular soft tissues or scrotum. Round to plump spindled myofibroblasts
tend to cluster around blood vessels (e-Fig. 46.14); a subset of cases has
a mature fatty component (IntI Gynecol Pathol. 2005;24:196). Binucle-
ate and multinucleate cells are common in the absence of mitotic activity.
These tumors are immunoreactive for desmin and estrogen and proges-
terone receptors. Some cases have an overlap with cellular angiofibroma
and deep aggressive angiomyxoma (see below).
9. Cellular angiofibroma involves the superficial soft tissues of the vulva or
inguinoscrotal region as well as the perineum, retroperitoneum, and sub-
cutis of the chest U Cutan Pathol. 2003;30:405). Grossly, the tumor is
a well-circumscribed mass that usually measures <3 em in diameter in
women and <10 em in men. The cut surface of the tumor has a yellow to
tan-brown, soft to rubbery appearance. The tumor is composed of plump
spindled cells with minimal eosinophilic cytoplasm, little cytologic atypia,
and few mitotic figures; a background of delicate collagen fibers is present
in tumors in females. The vascular component of the tumor is composed of
small- to medium-sized vessels, with or without prominent hyaline walls,
and is usually present throughout the entire lesion. Regressive/degenerative
changes, including extravasated erythrocytes, hemosiderin deposition, cys-
tic change, and intravascular thrombi are also present.
10. Giant cell angiofibroma, a slowly growing occasional painful tumor, has a
predilection for the eyelids and orbital region of adults, although it has
been identified in a number of other anatomic sites. The tumor is usually
about 3 em in greatest dimension, well-circumscribed, variably encapsu-
lated, with cystic and/or hemorrhagic areas. Microscopically, cellular areas
of round to spindled cytologically bland cells and multinucleated stromal
cells (which often line pseudovascular spaces as is common in giant cell
fibroblastoma) are set in a background of myxoid to collagenous stroma
with small- to medium-sized blood vessels. Both the mononuclear and
multinucleated cells are immunoreactive for CD34 and CD99, and occa-
sionally also for BCL2.
11. Nuchal-type fibroma and Gardner-associated fibroma are virtually identical
in terms of the microscopic features of dense, paucicellular collagenous
bundles, which overgrow and occupy the dermis, subcutis, and deep soft
tissues (Cancer. 1999;85:156). These tumors do not have the infiltrative
752 I SECTION XI: SOFT TISSUE AND BONE
3. Fibrohistiocytic lesions that typically occur in the skin (see Chap. 38) are
dermal dendrocytic proliferations like the reticulohistiocytoma, Langer-
hans cell histiocytosis, and juvenile xanthogranuloma (Am] Surg Pathol.
2003;27:579; Histopathology. 201 0;56:148).
B. Intermediate (rarely metastasizing)
1. Plexiform fibrohistiocytic tumor (PFH) primarily occurs in the first three
decades of life with a preference for the extremities and head and neck
(Ann Diagn Pathol. 2007;11:313). Grossly, it is a firm, multinodular and
poorly circumscribed mass that usually measures <3 em in greatest dimen-
sion in the dermis and subcutis. Microscopically, the architectural pattern is
that of nodules or elongated groups of cells with a plexiform arrangement.
Three cell types are present in the nodules: central multinucleated giant
cells, mononuclear histiocyte-like cells, and spindled fibroblast-like cells
(e-Fig. 46.27). Since one of the cell types may dominate, an appreciation
of the overall architecture is important for diagnosis since the nodules can
have few, if any, multinucleated cells and the spindle cell foci can resemble
fibromatosis. The tumor cells express SMA and vimentin; CD68 reactivity
is also present but is confined to the giant cells and mononuclear histiocyte-
like cells. Approximately 30% to 35% of cases locally recur and <5%
metastasize to regional lymph nodes and beyond (Arch Pathol Lab Med.
2007;131:1135; Am] Dermatopathol. 2004;26:141). A histogenetic rela-
tionship between PFH and cellular neurothekeoma has been proposed since
there are overlapping microscopic and immunophenotypic features (Am]
Surg Pathol. 2009;33:905; Am] Surg Pathol. 2007;31:329).
2. Giant cell tumor of soft tissue occurs in the superficial soft tissue of the
lower and upper extremities, and occasionally at other sites. It is a cir-
cumscribed, nodular mass that has a soft, fleshy, gray to red-brown cut
surface. Individual nodules of the mass measure up to 1.5 em and are
separated by fibrous septa that contain hemosiderin-laden macrophages.
Microscopically, admixed mononuclear round to oval cells and multinucle-
ated osteoclast-like giant cells are set in a richly vascular stroma. Mitotic
activity can be brisk (up to 30 mitotic figures per 10 HPFs are often present),
but cellular pleomorphism and atypia are absent. Metaplastic bone is noted
in up to 50% of cases, and definitive foci of vascular invasion are identi-
fied in 30% of cases. The multinucleated giant cells are strongly CD68
immunopositive, whereas the mononuclear cells express SMA and vimentin
but show only focal CD68 immunoreactivity.
c. Malignant
1. Pleomorphic malignant fibrous histiocytoma (MFH)Jundifferentiated high-grade
pleomorphic sarcoma is the diagnostic term reserved for those obvious high-
grade sarcomas typically presenting in the deep soft tissues, which lack
a lineage-specific immunophenotype. Considerable reassessment and revi-
sionism have been directed toward the question of whether MFH represents
a specific tumor type or a final common pathway of high-grade sarcomas
(Am] Surg Pathol. 1992;16:213; Am] Surg Pathol. 1996;20:131; Am]
Surg Pathol. 2001;25:1030). These neoplasms are characterized by com-
plex cytogenetic rearrangements that can involve over 30% of the genome
(Virchows Arch. 2010;456:201).
The tumor preferentially arises in the extremity and trunk, including the
retroperitoneum. Individuals over 40 years of age present with a rapidly
enlarging mass, and about 5% have metastatic disease at diagnosis, usually
in the lung. The tumor measures 15 to 20 em, if not greater, and has a
white to tan-white, fleshy to fibrous cut surface, with areas of necrosis and
hemorrhage. The microscopic pattern is often complex (e-Fig. 46.28), with
field to field variation in terms of spindle cells, rounded to ovoid cells, and
Chapter 46 • Soft Tissue I 7 59
and the abdominal cavity (typically in the omentum and mesentery). This
tumor can measure in excess of 30 em. It is composed of bland appearing
highly differentiated smooth muscle cells that have minimal atypia and a very
low mitotic rate (for tumors in the extremities and intra-abdominal tumors in
males, the mitotic rate is < 1 mitosis per 50 HPFs; for peritoneal/retroperitoneal
tumors in females, the mitotic rate is ::::;5 per 50 HPFs). While degenerative
changes such as fibrosis, myxoid change, hyalinization, and calcification may
be present, necrosis is absent. The diagnosis of a deep leiomyoma should be
approached with caution and should rely on the absence of virtually any mitotic
activity and atypia (Adv Anat Pathol. 2002;9:351; Ann Diagn Pathol. 2003;
7:60).
Pilar leiomyoma and uterine smooth muscle tumors are discussed elsewhere
(see Chaps. 39 and 33, respectively). There is a substantial literature on the
cytogenetics of uterine leiomyomas, but considerably less on the extrauterine
counterparts (Cancer Genet Cytogenet. 2005;158:1).
C. Malignant
1. Leiomyosarcoma (LMS} is typically a malignancy presenting in mid-life and
beyond. There are four clinicopathologic settings: a retroperitoneal mass;
a tumor arising from a large blood vessel with a preference for veins of the
lower extremity and inferior vena cava; a subcutaneous or intramuscular
tumor of the extremities; and a dermal-based neoplasm. Uterine and intesti-
nal LMSs are not considered in this chapter, although the uterus is the most
common primary site overall for LMS (Adv Anat Pathol. 2011;18:60).
Except for cutaneous LMS, these tumors usually measure in excess of
6 em and have a trabeculated or smooth, glistening, white-gray cut surface.
High-grade tumors are accompanied by necrosis and hemorrhage. Archi-
tecturally, the tumors are composed of intersecting bundles of eosinophilic
spindle cells with elongated nuclei with blunted ends (e-Fig. 46.29). Nuclear
pleomorphism and an increased mitotic rate with atypical mitotic forms
are characteristic. Poorly differentiated LMS is a pleomorphic high-grade
sarcoma whose smooth muscle differentiation is highlighted by immunore-
activity for SMA, desmin, and h-caldesmon. Focal expression of keratin,
EMA, CD34, and S100 may be present. These tumors typically have
complex karyotypes with gains and losses across multiple chromosomes
(Virchows Arch. 2010;456:201).
A distinct clinicopathologic group of smooth muscle neoplasms are rec-
ognized in the immunocompromised setting; these tumors are associated
with genomic integration of Epstein-Barr virus (EBV) (] Clin Pathol. 2007;
60:1358; Am] Surg Pathol. 2006;30:75;] Cutan Pathol. 2011;38:731) and
have been described in both visceral, soft tissue and cutaneous sites. These
tumors have low-grade spindle cell features to the extent that the traditional
criteria for the diagnosis of LMS are not met in all cases, and are designated
as EBV-associated smooth muscle tumors in some cases.
VII. PERICYTIC (PERIVASCULAR} TUMORS. Tumors in this category show evidence of
myoid/contractile perivascular cell differentiation. Morphologically, they have a
tendency to grow in a circumferential perivascular pattern.
A. Benign
1. Glomus tumor usually occurs in the subungual region of the hand, wrist, and
foot, but not to the exclusion of other sites including the bone, lung, and
intestinal tract (Arch Pathol Lab Med. 2008;132:1448). Those tumors are
< 1 em in greatest dimension in most cases, and are painful with minimal
tactile stimulation or exposure to cold. Microscopically, these tumors con-
sists of a mixture of glomus cells (characterized as uniform, small, rounded
cells with eosinophilic to amphophilic cytoplasm and a central nucleus),
smooth muscle cells, and central vascular space (e-Fig. 46.30). On the basis
Chapter 46 • Soft Tissue I 7 61
of the relative proportion of these three elements, there are three subtypes
of glomus tumor: solid, composed of nests of glomus cells surrounding
capillary-sized vessels; glomangioma, composed of small dusters of glomus
cells surrounding dilated vessels; and glomangiomyoma, in which there is a
transition from typical round glomus cells to elongated cells that resemble
mature smooth muscle. Glomangiomas may be multiple or familial, and are
likely vascular malformations (Arch Dermatol. 2004;140:971).
2. Myopericytoma, previously interpreted as a hemangiopericytoma, usually
arises in the subcutis of the distal extremities U Clin Pathol. 2006;59:67).
Less than 2 em in greatest dimension, this tumor is unencapsulated
but well circumscribed. Microscopically, the tumor is composed of a
densely cellular population of oval to spindle-shaped cells with eosinophilic
to amphophilic cytoplasm arranged in multilayered concentric profiles
around compressed blood vessels. Mitotic figures are inconspicuous. There
is diffuse immunopositivity for SMA and focal immunopositivity for
CD34.
B. Malignant. Malignant glomus tumor (glomangiosarcoma) is rare and seemingly
arises from a benign-appearing glomus tumor. The characteristic findings are a
visceral or subfascial origin, a size >2 em, marked nuclear atypia, and atypical
mitotic figures. However, a subset of tumors does not have all of these findings;
these cases are designated as tumors of uncertain malignant potential (Am]
Surg Pathol. 2001;25:1). The round cell type is composed of poorly differen-
tiated round cells, so SMA and pericellular type IV collagen are required for
diagnosis. A spindle cell variant has features of LMS or fibrosarcoma.
VIII. SKELETAL MUSCLE
A. Nonneoplastic disorders
1. Preparation of skeletal muscle biopsies. Histopathologic examination of
skeletal muscle biopsies, whether obtained through an open biopsy or
through a needle biopsy, continues to have a critical role in the evaluation of
patients with suspected myopathy (Curr Neuro Neurosci Rep. 2004;4:81).
One portion of the biopsy should be formalin fixed and paraffin embed-
ded according to standard laboratory protocols; another should be frozen
for enzyme histochemistry studies; and the remainder should be fixed in
glutaraldehyde for electron microscopic studies. While the histopathologic
evaluation of skeletal muscle biopsies is a key component in the evaluation
of neuromuscular disorders, and can be used to diagnose a variety of inher-
ited, inflammatory, and toxic myopathies, it should only be performed in
the context of a thorough history and clinical examination that has included
appropriate laboratory studies (including measurement of serum creatine
kinase) and electromyography. A number of inflammatory, toxic, and axial
myopathies have characteristic histopathologic findings, and immunohis-
tochemical studies can be utilized to characterize inflammatory infiltrates
when present (Autoimmunity. 2006;39:161).
2. Muscular dystrophies and other congenital myopathies are diagnosed pri-
marily on the basis of clinical and electromyographic features. Increasingly,
the histopathologic evaluation of muscle biopsies in these settings has been
supplanted by genetic testing as an increasing number of diseases has been
characterized at a genetic level (Pediatr Dev Pathol. 2006;9:427; Brain
Pathol. 2001;11:206).
3. Mitochondrial myopathies, which traditionally have been diagnosed on
the basis of the demonstration of so-called ragged-red fibers by Gomori
trichrome stain, are increasingly diagnosed on the basis of genetic testing
since the genetic abnormalities in mitochondria that underlie these disease
have recently been characterized (Submicrose Cytol Pathol. 2006;38:201;
Biosci Rep. 2007;27:23).
762 I SECTION XI: SOFT TISSUE AND BONE
B. Benign
1. Rhabdomyoma by definition shows skeletal muscle differentiation, and
includes cardiac rhabdomyoma and extra-cardiac rhabdomyoma (which
has two subtypes: fetal and adult). Both cardiac and fetal rhabdomyomas
may be hamartomas rather than true neoplasms; tuberous sclerosis and
nevoid basal cell carcinoma syndrome accompany these tumors, respec-
tively (Ear Nose Throat]. 2004;83:716; Pediatrics. 2006;118:e1146; Con-
genital Heart Dis. 2011;6:183).
a. Fetal rhabdomyoma occurs almost exclusively in children under the age
of 10 years, with a predilection for the postauricular region and chest
wall (Am] Surg Pathol. 2008;32:485). The tumor forms a nodule in the
subcutis or deeper soft tissues that has a circumscribed noninfiltrative
pattern and is composed of bundles of fetal myotubes with interspersed
small immature-appearing mesenchymal cells. The classic or myxoid
pattern must be differentiated from ERMS, the latter of which has a
less well-organized pattern, hyperchromatic nuclei, and mitotic figures.
The so-called intermediate or cellular pattern (juvenile rhabdomyoma) is
characterized by skeletal muscle differentiation beyond the fetal myotube
stage.
b. Adult rhabdomyoma is solitary in 75% to 80% of cases, but in about
25% of cases is multinodular, even multicentric. There is an overwhelm-
ing predilection for the head and neck region (especially the larynx and
pharynx) (Am] Otolaryngol. 2011;32:240). Microscopically, lobules of
large uniform polygonal cells with abundant granular, vacuolated, or
eosinophilic cytoplasm; well-defined cell borders; and round nuclei with
a prominent nucleolus are present. Cytoplasmic cross striations and rod-
like inclusions are also present, in addition to abundant glycogen. Com-
plete excision is recommended, since recurrence occurs in >40% of cases
that have been incompletely excised.
c. Genital rhabdomyoma presents almost exclusively in the vagina in women
between the ages of 35 and 50 years as a solitary 1 to 3 em polyp that may
have been present for several years. Microscopically, bland, interlacing,
haphazardly arranged rounded to strap-like cells that have abundant
eosinophilic cytoplasm with cross striations, cytoplasmic glycogen, and
a centrally located round nucleus with a prominent nucleolus are present.
The tumor cells are embedded in a fibrous stroma with dilated vessels.
Local excision is curative.
c. Malignant
1. Embryonal rhabdomyosarcoma (ERMS} is the most common soft tissue sar-
coma of childhood (50% to 60% of cases), typically presents before
10 years of age, and accounts for 75% to 85% of all rhabdomyosarcomas
in children. The other types of rhabdomyosarcomas, alveolar and pleomor-
phic types, comprise 20% to 30% and 5% or less, respectively. ERMS has
a predilection for the head and neck (25% to 35% of cases) and pelvis-
genitourinary tract (30% to 40% of cases). Unlike alveolar rhabdomyosar-
coma (ARMS), a minority of ERMS presents in the peripheral soft tissues.
The three basic microscopic patterns of ERMS are botryoid, spindle, and not
otherwise specified (NOS), which proportionately account for 5%, 10%,
and 85% of cases, respectively. The botryoid and spindle subtypes have a
"superior" outcome, whereas the NOS subtype has an "intermediate" to
"poor" prognosis (Pediatr Dev Pathol. 1998;1:550).
The preoperative-pretreatment staging of childhood RMS is a multitier
system, which includes the status of the tumor before treatment utilizing a
TMN-based system (Table 46.6). The prognostically favorable sites of RMS
in children include the orbit, nonparameningeal head and neck, common
a..~.46· Sottr-.e. I JU
.... ...
,, f.t:) I ( !-!-11
Olbl
Hood end IICICk"
Tl « T2
Tl «T2 ..,b
......... ...........
Pl'etltllmtnllllll hllf.. ol ClllkiiNod Rlllll>cb•,.--•
llnuiMI- tu
..,b NON! OfNX
NON! OfNX
M~
M~
Gon!mu~•IIY' n .. T2 .a orb NONI0<NX MO
II Blllllcloo'proololl n .. T2 • NO"NX MO
Ellnlrnly n .. T2 • NO"NX MO
Cranial TlorT2
• NOarNX M~
D0/11116i'iftliOI T10fT2
• NOarNX MO
Ollie""'
111 Bloddelillto>AII"
Ellnlrnly
Tl «T2
n .. T2 •b Nl
NONI0<NX
M~
MO
Cranial n .. T2 b NONI0<NX MO
PIJ'IIl'MI('Inpal T1orT2 b NONiorNX M~
Ollie,..,
IV All T10fT2 eorb NOar Nl Ml
764 I SECTION XI: SOFT TISSUE AND BONE
S-100 positivity (only in 50% of cases), but the tumors are more commonly
positive for CD57 (Leu 7), collagen type N, and myelin basic protein (Arch
Pathol Lab Med. 2009;133:1370). Chromosomal losses are more common
than amplifications (Virchows Arch. 2010;456:201).
Variant patterns of MPNST show rhabdomyoblastic differentiation (malig-
nant triton tumor), glandular formation, and heterotopic bone and/or cartilage
formation. These variants are encountered with some frequency in the setting
of NF1 (the setting in which 40% to 50% of all MPNSTs are seen) (Neurosurg
Clin N Am. 2008;19:533).
XII. TUMORS OF UNCERTAIN DIFFERENTIATION. For many of the tumors in this category,
either the cell type that produces the tumor or the normal cell type that the tumor
is recapitulating, or both, is unknown.
A. Benign
1. Intramuscular myxoma occurs primarily in adults between 40 and 60 years
as a solitary, painless, slowly growing mass within the muscle of the thigh,
buttocks, or limb girdle. Most tumors measure 5 em in maximal dimen-
sion, and are well circumscribed with a pale gelatinous or myxoid cut sur-
face. The tumor cells are small, bland, stellate to spindle in shape, have
a very low mitotic rate, and are present in an abundant myxoid matrix
that contains an inconspicuous vascular network (e-Fig. 46.49). Focal areas
of increased cellularity and vascularity with collagenous background are
present in some cases whose presence may cause concern (Am I Surg Pathol.
1998;22:1222). Simple excision is followed by a recurrence rate of <5%
of cases (Histopathology. 2001;39:287). The differential diagnosis of myx-
oma includes many other myxoid tumors of the soft tissues (Histopathology.
1999;35:291; Ann Diagn Pathol. 2000;4:99). One or more myxomas of the
soft tissues with fibrous dysplasia defines Mazabraud syndrome. Multiple
cutaneous and mucosal myxomas are manifestations of Carney complex
(Lancet Oneal. 2005;6:501; Neuroendocrinology. 2006;83:189).
2. Juxtaarticular myxoma, as the name suggests, usually arises in the vicinity of
a large joint (primarily the knee). The lesion has a predilection for males
between 20 and 40 years of age, and is usually <5 em maximal dimension.
This tumor is indistinguishable from an intramuscular myxoma, but is prob-
ably a different entity than the latter (Virchows Arch. 2002;440:12). Some
cases may have hemorrhage and hemosiderin deposition, chronic inflam-
mation, and fibrosis.
3. Digital myxoma (digital mucus cyst), usually occurs on a finger, and in adult
women as a painful nodule < 1 em in diameter. Histologically, it resem-
bles intramuscular myxoma. Cutaneous myxoma (also known as superficial
angiomyxoma) is a cutaneous or subcutaneous lesion which occurs primar-
ily in adults between 30 and 50 years of age; it must be differentiated from
nerve sheath myxoma of peripheral nerve sheath origin and neurothekeoma
(Mod Pathol. 2011;24:343).
4. Deep aggressive angiamyxoma is a slowly growing locally infiltrative tumor
that usually occurs in the pelviperineal soft tissue of women, but is also
seen as a scrotal mass (Int I Gynecol Pathol. 2005;24:26; IntI Urol.
2003;10:672; Histopathology. 2009;54:156). It forms a soft poorly cir-
cumscribed mass measuring 10 to 20 em in greatest dimension that has a
gelatinous cut surface. Small stellate to spindled cells with ill-defined cyto-
plasm and bland nuclei are set in a myxoid stroma rich in collagen fibers
(e-Fig. 46.50). Numerous, variably sized, thick or thin walled vascular chan-
nels are present, from which smooth muscle seems to spin off and merge
with the surrounding stroma. The tumor cells express vimentin and desmin,
and also usually show nuclear expression of estrogen and progesterone
receptors.
774 I SECTION XI: SOFT TISSUE AND BONE
MO
Ml
No "'I!<NIIJmJil nccl&
Rcwlonol lymph ncdo mot"t""'
No-rtm-.ol&
Dlalart..........U
- l c ..... ,...Ollie.,..,.
51qo1A no NO MO Gl,GX"
Tlb NO MQ Gl,G.X
SlqalB 'T2a NO MO Gl,GX
T2b NO MO Gl,GX
Slqo t!A n. NO MO G2,G3
Tlb NO MO G2,G3
5lqo t!B 'T2a NO MO G2
T2b NO MQ G2
$QII!I n... T2b NO MO c:B
AI!JT Nl MO AJryG
Slqo IV AnyT AllyN Ml AnyG
780 I SECTION XI: SOFT TISSUE AND BONE
SUGGESTED READINGS
Dehner LP. Soft tissue. In: Stocker T, Dehner LP, Husain AN, eds. Stocker and Dehner Pediatric
Pathology, 3rd ed. Philadelphia: Lippincott, Williams & Wilkins; 2011.
Fletcher CDM, Unni KK, Mertens F. Pathology and Genetics of Tumors of Soft Tissue and Bone.
World Health Organization Classification of Tumors. Lyon: IARC Press; 2002.
Kempson RL, Fletcher CDM, Evans HL, et al. Tumors ofthe Soft Tissues. Atlas of Tumor Pathology,
3rd Series, Fascicle 30. Washington, DC: Armed Forces Institute of Pathology; 2001.
Pfeifer JD. Molecular Genetic Testing in Surgical Pathology. Philadelphia: Lippincott, Williams &
Wilkins; 2006.
Weiss SW, Goldblum JR. Soft Tissue Tumors, 5th ed. St. Louis: Mosby; 2008.
Retroperitoneum
Catalina Amador-Ortiz, Louis P. Dehner,
and John D. Pfeifer
I. NORMAL ANATOMY. The retroperitoneal space (in some respects a virtual space) is
located between the posterior parietal peritoneum and the fascia that covers the
muscles of the lumbar region. It extends upward to the diaphragm, downward to
the base of the sacrum and iliac crests, and laterally to the external borders of
the lumbar muscles and the ascending and descending colon. The retroperitoneum
contains loose connective tissue surrounding lymph nodes, the aorta and inferior
vena cava with their vascular branches, the adrenal glands, the kidneys and ureters,
the pancreas, and portions of the duodenum. This chapter will focus on the entities
that arise from the tissues of the retroperitoneal space; disorders arising from the
organs that are completely or partially retroperitoneal are covered in the respective
chapters.
II. NONNEOPLASTIC DISEASES
A. Inflammatory
1. Idiopathic fibroinflammatory lesions constitute a morphologically similar cat-
egory of tumefactive processes, and a variety of diagnostic terms such as
sclerosing mesenteritis and/or panniculitis have been applied. The usual pre-
sentation is a mass in the retroperitoneum and/or root of the mesentery; scle-
rosing changes may extend into the inferior retroperitoneum with similar fea-
tures, if not identical, to idiopathic retroperitoneal fibrosis. A biopsy reveals
a variably intense mixed lymphocytic and plasmacellular infiltrate in a dense,
relatively hypocellular collagenous background. Residual adipose tissue may
have the features of fat necrosis with dystrophic calcification and panniculi-
tis. In the presence of lymphadenopathy, the possibility of lymphoma should
be excluded. Metastatic disease including poorly differentiated adenocarci-
noma of the pancreas, carcinoid, lobular carcinoma of the breast, and acinar
carcinoma of the prostate may be a accompanied by disproportionate fibroin-
flammatory reaction to the actual volume of tumor, and thus also need to be
excluded. If the biopsy has a prominently spindle cell component, inflamma-
tory pseudotumo.r, inflammatory myofibroblastic tumor (IMT), sarcomatoid
carcinoma, pleomorphic spindle cell sarcoma, and IgG4-related sclerosing
disorder are also in the differential diagnosis (Nat Rev Gastroenterol Hepa-
tol. 2010;7:401;] Clin Pathol. 2008;61:1093). Since the histologic findings
of idiopathic fibroinflammatory lesions are nonspecific, even after the other
possibilities have been excluded, the diagnostic line in the report may simply
read "chronic inflammation and fibrosis."
2. Malakoplakia occasionally affects the retroperitoneal soft tissues. The pro-
cess is a response to infection and grossly appears as a yellow plaque-like
lesion. Microscopically, it is composed of abundant inflammatory cells and
sheets of granular and vacuolated histiocytes that contain lamellated, PAS-
positive diastase-resistant inclusions known as Michaelis-Gutmann bodies.
These distinctive inclusions (e-Fig. 47.1)* are thought to represent the rem-
nants of bacteria within phagosomes that have been mineralized by calcium
and iron.
*All e-figures are available online via the Solution Site Image Bank.
781
782 I SECTION XI: SOFT TISSUE AND BONE
4. Fibroblastic tumors
a. Benign fibroblastic tumors other than desmoid fibromatosis are rare in the
retroperitoneum. It may be difficult to differentiate between a presumed
fibrous tumor and a nonneoplastic fibroinflammatory process (e.g., an
IgG4 fibrosclerosing process).
b. Desmoid fibromatosis, IMT, extrapleural solitary fibrous tumor/hemangio-
pericytoma, and gastrointestinal stromal tumors {GISTs) are several examples
of intermediate behavior tumors that may involve the retroperitoneum.
Desmoid fibromatosis usually arises in the mesentery-omentum with
extension into the retroperitoneum, but occasionally is limited to the
retroperitoneum. Those desmoid tumors arising in the proximal lower
extremity can, in the course of several local recurrences, extend through
sciatic notch into the retroperitoneum as well; the presence of familial ade-
nomatous polyposis in these patients should be addressed. Desmoid fibro-
matosis is not encapsulated but is well circumscribed, except in these cases,
which represent a local recurrence. Its cut surface shows a firm, trabecu-
lated pattern resembling a smooth muscle tumot: Microscopically, dense
interlacing bundles of uniform spindle-shaped fibroblasts are present in a
collagenous, keloid-like, or myxoid stroma with no evidence of nuclear
atypia. Scattered mitotic figures may be identified and should not be a
source of concern unless they are atypical (e-Fig. 47.7). The tumor is
immunopositive for vimentin, SMA, and nuclear ,8-catenin staining. If the
tumor is positive for CD34 and CD117 (c-kit), GIST is the favored diag-
nosis as either a local recurrence or a rare primary extraintestinal GIST
(] Gastrointest Surg. 2009;13:1094).
Primary retroperitoneal extrapleural solitary fibrous tumor/hemangio-
pericytoma is uncommon, but documented (]Am Coli Surg. 2004;198:
322; Hum Pathol. 2000;31:1108). These CD34-positive (but CD117-
negative) neoplasms may be quite cellular and mitotically active, in which
case the most appropriate diagnosis is malignant solitary fibrous tumor.
Although grouped together as related (if not identical) tumors in the cur-
rent World Health Organization (WHO) scheme (see Chap. 46), the diag-
nosis of hemangiopericytoma is reserved for tumors that resemble the
cellular areas of solitary fibrous tumor with the characteristic network of
vascular profiles.
IMT, like desmoid fibromatosis, may present in the mesentery-
omentum with extension into the retroperitoneum. IMT typically presents
in children and young adults. Grossly, it is a circumscribed firm, gray-white
mass that measures 5to 10 em. Microscopically, it has a varied micro-
scopic pattern of spindle cells with accompanying plasma cells, myxoid
foci resembling nodular fasciitis, and dense hyalinized collagen. In addi-
tion to immunopositivity for vimentin and SMA, 50% or more of cases
are anaplastic lymphoma kinase (ALK-1) positive (Am] Surg Pathol.
2007;31:509). The presence of IgG4 plasma cells in IMT is seen in some
cases (Mod Pathol. 2011;24:606).
c. Malignant fibroblastic tumors are very rare in the retroperitoneum. Adult
fibrosarcoma is currently a diagnosis of exclusion and, according to recent
series, accounts for <5% of all retroperitoneal sarcomas; sarcomatous
transformation of a solitary fibrous tumor should be considered in the
differential diagnosis (Am] Surg Pathol. 2009;33:1314). Microscopically,
fibrosarcoma is composed of spindle-shaped cells with large elongated
nuclei and scant cytoplasm, arranged in a classic herringbone pattern.
Infantile fibrosarcoma is known to occur in the retroperitoneum, but
most commonly presents in the extremities; the identical neoplasm
arising in the kidney in infancy, cellular mesoblastic nephroma, has the
Chapter 47 • Retroperitoneum I 7 87
I. NORMAL MICROSCOPIC ANATOMY. The bones are composed of compact bone, which
is derived from intramembranous ossification, and coarse cancellous bone, which
is the osseous remnant of endochondral ossification. Compact bone makes up the
cortices of long bones and constitutes their diaphyses and the surface portion of
their metaphyses, as well as the compacta of the flat and irregular bones. Cancellous
bone is present in the medullary cavity and is abundant at the ends of the long
bones. In bone, form follows function (Wolff's law). In the shafts of bones, most of
the forces act upon the surface. Here, the compact bone, which is 90% solid and
only 10% space, bears the compression, tension, shear, and torsional forces. The
medulla, shielded from forces, contains practically no bone at all. The ends of the
bones are supported by the vertical plates and horizontal struts of the cancellous
bone, yet cancellous bone is only 25% bone and 75% marrow by volume; here the
cortex is very thin.
Bone matrix is classified as woven or lamellar depending on the predominant
fiber arrangement of its collagen. In woven bone, the collagen fiber pattern is ran-
dom. This type of bone is found in the fetal skeleton and in processes in which
there is very rapid bone production. In lamellar bone, the bone collagen fibers are
arranged in stacks of tightly packed fibers that are parallel in the same stack. In the
next layet; the collagen fibers are also parallel to one another, but their direction is
different from the collagen in the previous stack so that the bone appears to be lay-
ered. Both compact bone and cancellous bone consist of lamellar bone after the age
of 3 years. After this age, woven bone is almost always pathologic, although the
etiology is often not discernible without imaging studies (Bullough PG. Orthopedic
Pathology. 5th ed. St. Louis: CV Mosby; 2010). In compact bone, the lamellae are
arranged concentrically around central vascular canals termed Haversian canals;
each vascular canal and its associated lamellae are referred to an osteon or Haver-
sian system. In cancellous bone, the lamellae are arranged in linear, parallel plates
(e-Fig. 48.1).* Adjacent osteons are separated from each other and from intersti-
tial lamellae (see the section on circulatory diseases) and circumferential lamellae
(which encircle the inner or outer cortex and are remnants of periosteal intramem-
branous ossification) by basophilic staining cement lines. Cement lines are sliding
planes that are richer in calcium than surrounding bone matrix but the exact com-
position of which is unknown; they are produced by osteoblasts when bone is
synthesized following osteoclast resorption (reversal cement lines) or after a period
of inactivity (arrest cement lines). In the former type, the lamellae are discontinuous
on either side of the cement line and in the latter the lamellae are continuous on
either side (e-Fig. 48.2).
II. SPECIMEN PROCESSING
A. Gross handling and selection of sections. The approach to specimen handling
is largely one of common sense. Small biopsy specimens should be submit-
ted for sectioning in their entirety. If there is any doubt about whether they
*All e-figures are available online via the Solution site image bank.
790
Chapter 48 • Bone Neoplasms and Other Nonmetabolic Disorders I 7 91
contain bone, they should be fixed, briefly decalcified, and rinsed. Most bone
biopsies performed with needles are sufficiently thin for adequate fixation and
decalcification, whereas curettings may sometimes need to be sliced into thin-
ner fragments. The amount of curettings to submit for sectioning depends
on their volume and the uniformity of the curettings. When it is feasible, all
curettings should be submitted. If the lesion curetted is a hyaline cartilage
tumor, as much of the histology as possible should be reviewed to identify
atypical chondrocytes as well as any subtle interface with normal surrounding
bone.
Other large specimens such as total joint replacements and bone resections
also need to be sliced into thinner fragments. Although this may be accomplished
with large band saws or other power-type saws, motorized saws are dangerous
and somewhat time-consuming to maintain properly, especially in a laboratory
that receives a limited number of bone specimens. Vibrating or oscillating saws,
which are usually available in autopsy suites, should be avoided if possible,
because they do not section uniformly and their oscillating movement creates
tension and compression artifacts that often make bone sections impossible to
interpret properly. A very easy approach is to hold bone specimens steadily in
a tabletop vise or clamp and to cut them with a hacksaw in which two fine-
tooth blades are separated by 2- to 3-mm-thick washers. Such an apparatus is
easily and cheaply made, although there are commercial instruments available
for the same purpose. It must be emphasized that double-bladed instruments
should be scrupulously cleaned between every specimen to avoid tissue cross-
contamination between different cases.
The handling and disposition of larger resection specimens depends on the
reason for the procedure. For malignant tumors in which patients have not
received neoadjuvant chemotherapy (after biopsy but prior to resection), grad-
ing, staging, and adequacy of resection are the major clinical issues. Amputations
from these patients should include sections from the soft tissue and vascular mar-
gins as well as those from the tumor itself. Tumor sections should be taken in
such a way as to document the pertinent tumor histology, whether the tumor
involves the medulla and/or cortex, and how far the tumor extends into soft
tissues. The specimen should be cut in such a way as to disclose the greatest
extent of tumor; review of the imaging studies can guide the selection process.
Careful attention should be paid to taking sections from any areas that are
grossly disparate from the appearance of the majority of the tumor. Radical
resections for malignant tumors that are not amputations need the same sec-
tioning methods, but any area of the resection constituting a margin must be
sectioned and appropriately designated. This includes the bone resection mar-
gin, overlying soft tissue dissection margins, and the margins of any skin and
soft tissue encompassing a prior biopsy site.
Specimens resected from patients who have received neoadjuvant chemother-
apy (currently used in osteosarcoma and in Ewing sarcoma/primitive neuroec-
todermal tumor [EWS/PNET]) need more extensive sampling to estimate the
extent of treatment-associated necrosis. This means that one or more thin slabs
should be cut through the entire extent of the bone and tumor, and that the
entire slab or slabs should be fixed, decalcified, mapped, and examined not
only for tumor stage, but also for the extent of necrosis. The slabs should be
photographed so as to produce a section map; if a specimen x-ray machine
is available, specimen radiographs can be used both as section maps and as
controls for adequate specimen decalcification (e-Fig. 48.3). Additional sections
may be taken if there are areas not in the slab selected that appear as though
they might be viable; the pathologist's task in this enterprise is to find viable
tumor if any is present. It is worthwhile to remember that to extrapolate the
degree of necrosis in a single slab into necrosis of the tumor as a whole makes
792 I SECTION XI: SOFT TISSUE AND BONE
tissue. Symptoms and signs are fairly similar in orthopedic diseases; these consist
of pain, loss of function, deformities, and (in the case of tumors) sometimes a
mass or a sense of fullness. Pain is the most common symptom, and although it
may vary considerably, pain severe enough to wake a patient from sleep is the
type suspicious for neoplastic diseases.
D. Importance of radiologic findings. The surgical pathology of orthopedic diseases
most often consists of defining the nature of bone lesions that are space occupy-
ing on imaging studies, advising the clinician if an infection may be present, and
histologically documenting miscellaneous bone diseases that are not diagnosable
by imaging studies alone. Because surgical pathologists usually render biopsy
diagnoses with the assumption that a biopsy is representative of the pathologic
process, it is natural to assume the same parameters in bone biopsies. This
is a potentially dangerous assumption, because most orthopedic diseases are
invisible without imaging studies. This means that to assure that a biopsy is rep-
resentative of a process, the smaller the biopsy specimen, the greater the need to
review the imaging studies defining that process. Because bones are deep seated,
imaging studies are required to grasp the extent and behavior of bone lesions.
For a surgical pathologist, correlating imaging studies with histologic findings
depends on knowledge of normal bone and joint anatomy, normal anatomy as
represented in radiographic images or other imaging studies, and the rudimen-
tary alterations in imaging studies produced by pathologic processes (not only
what a process does to normal bone, but also how normal bone alters the pro-
cess) (Adv Anat Pathol. 2005;12:155). The majority of the radiographic image
produced by long bones is due to beam attenuation by cortical bone in the shafts
and by cancellous bone in the ends (e-Fig. 48.4). The attenuation produced by
flat bones is primarily due to cortical bone, and that of irregular bones depends
on the proportion of bone elements in any given part of the bone.
Space-occupying lesions within bone usually cause bone destruction, bone
production, or some combination of the two. Destructive lesions are not seen
in a single radiographic view until at least 40% of the bone in the path of
the x-ray beam is destroyed. This means that almost an entire thickness of
cortex must be destroyed to see the lesion if an intact and a destroyed cortex
are superimposed in one view, or that at least 40% of cancellous bone must be
destroyed in a bone end. It is partly for this reason that orthogonal views of bones
are taken (e.g., posteroanterior and lateral views) so that destructive lesions may
be isolated in routine radiographs. Radiodense lesions superimpose on the extant
bone, causing more attenuation and easier visibility on a routine radiograph. In
contrast, a lesion that is less dense than bone may fill the entire medullary cavity
of a bone, but if it does not destroy the cortex it will be invisible regardless of the
view because the dominant attenuator of the x-ray beam is the cortex, not the
medullary fat or marrow. It is for these reasons that other imaging studies such
as computed tomography (CT) scans and magnetic resonance imaging (MRI)
are performed. These studies yield information that is complementary to that
derived from routine radiographs. While they may be more sensitive in yielding
information, a particular type of imaging modality should be used in concert
with routine radiographs to answer a particular clinical question not answered
by the radiographs.
Ill. DIAGNOSTIC FEATURES OF BONE LESIONS. There are very few general categories of
bone disease (Table 48.1 ), although there are many individual diseases (McCarthy
EF, Frassica FJ. Pathology of Bone and joint Disorders with Clinical and Radio-
graphic Correlation. Philadelphia: WB Saunders; 1998). Most patients can be
separated into general diagnostic categories on the basis of their imaging stud-
ies. For example, traumatic diseases, which are among the commonest problems,
will demonstrate fractures (with or without bone displacement) or dislocations
on routine radiographs. Metabolic bone diseases (discussed in Chap. 49), which
,.... I SECTION .... SOFT TISSUE AND BONE
,,/,t!i(!!\1 8-IX-bJC._,
Conpnbl
il<M!Iopnonl&Waoqul'ad
Trou.-
Cir<~~~&~o~y
-··
--11\df'lleetd.....
Iaii-t:
Noojlloll1: and lumrr~lk6
--bll
T~llln
-~otol
liJCillltn
Dlft'u.la~ ltr'l'lllrtfmM
....l!wd;UIUIIIf
<I() -~~ .. 101>1101'/Nitf
-.,""""'"' Vecybnlld
nnlllllk ~-·
Any porto! Oil)' bono A1rt Froctura lriOI.; <llloc«b\1 Ho/TIOIJhap: .,..,_,,
~"""
W'!Mn tale and ehondrold .-.....,.,.
mati11t
CmiiDJ
_ .., •••, ..!& C........endoofi.Ba S-40 Wedfll'-wped ,__Illy;
•-111ln;collpsool
"""''* 1!\III'CW and bo""'
Sllbeiiii:IJiu pllla - ..
None
01t!l:1*" eortlap
lclopollllo lnllr- M111Lllll of IBa ~ liazydomly... -.. Noel#:"'"""" and bono: Endaldroma•
r&oombl!11omotb
.,_hrt
Clllcll\c$1!oft and 001111~
-~
"""'
.. --
>5() EM~-~~~
-.,..shlplcf ....
lJitef': et&II'M trabte:ul.ltlon;
loa of -•lldulllll)'
d..,..t:I'Uo,.
,,_....
I'OIICI'IIIIM +
Oe- illlf><:
-INslnmlldltiD
and
~o,.,
....•
Dlnad Ani; open trl,..,. «
inflammation, and edema in the closed confines of the cortex compromises the
medullary nutrient arteries and sinusoids due to resulting increased pressure.
The innermost cortical circulation may be similarly compromised by increased
intramedullary pressure. If the pressurized exudate finds its way into empty
Haversian and Volkmann canals, it may push its way through these intracor-
tical spaces and eventually dissect the periosteum, and its perforating arteries,
from the cortex. If the cortex is deprived of its dual circulation, then it in turn
becomes necrotic; this necrotic bone is called sequestrum. The combination
of necrotic bone sequestrum, marrow fibrosis and/or fat necrosis, and mixed
inflammatory infiltrates (usually including neutrophils and plasma cells) pro-
vides good histologic corroboration of osteomyelitis, but the demonstration of
organisms is the gold standard for the diagnosis of infections (e-Fig. 48.11).
F. Iatrogenic disorders. Treatment-related disorders are seldom a major problem in
the pathologic diagnosis of orthopedic disease, provided that an accurate clinical
history is communicated to the surgical pathologist. For example, the diagnosis
of osteosarcoma would be very unusual in a patient of the sixth decade with-
out prior radiation of the site, or without some other underlying premalignant
bone lesion. Administration of various therapeutic regimens may lead to sec-
ondary alterations in bones; perhaps the most notable of these is the amyloidosis
of bones, tendon sheaths, and ligaments that develops from ,82-microglobulin
accumulation in long-term hemodialysis patients. Substances that have been
given parenterally but that are not metabolized may also be deposited in bones
or joints; without prior knowledge of therapeutic treatment, it may be difficult
to make an accurate diagnosis (e-Fig. 48.12).
G. Neoplastic and tumor-like lesions. Primary tumors of bone are quite rare,
accounting for only 0.2% of all malignancies, or an incidence of 1 per 100,000
individuals per year (Fletcher, CDM, Unni K, Mertens K, eds. Tumors of Soft
Tissues and Bone. Lyon, France: IARC Press; 2002). There is a bimodal age
distribution, with one peak in adolescence and a smaller one in patients older
than 60 years. Among other characteristics, each bone tumor has its own age
predilection, which is very useful from a differential diagnostic standpoint. Pri-
mary benign bone tumors are probably less common than primary malignant
tumors if the very common nonossifying fibroma, osteochondroma, and enchon-
dromas of the hands are excluded. In addition to benign and malignant bone
neoplasms, there are a number of nonneoplastic lesions that can present in a
manner similar to neoplastic conditions (Table 48.3 ); all of these lesions are
discussed below, and their main features are presented in Tables 48.4 and 48.5.
Pathologic stage is among the findings that are recommended to be reported for
bone tumors (Hum Pathol. 2004;35:1173) and the American Joint Commit-
tee on Cancer (AJCC) Tumor, Node, Metastasis (TNM) staging scheme (Table
48.6) and/or the simpler Musculoskeletal Tumor Society scheme (Table 48. 7)
can be used for this purpose.
1. Cartilage-forming tumors
a. Osteochondroma is a cartilage-capped bony protrusion (e-Fig. 48.13) that
arises from the surface of any bone that models or grows by endochon-
dral ossification. On imaging, osteochondromas demonstrate a marrow
cavity and a cortex continuous with those of the host bone. Although
classified as bone neoplasms, osteochondromas may also result from
displacements of the cartilaginous grown plate. This is consistent with
their metaphyseal location and the fact that they cease to grow after
skeletal maturation. Most osteochondromas are sporadic and solitary;
however, multiple lesions are present in osteochondromatosis, which is
an autosomal dominant hereditary condition. The presence of EXT-1
mutations in the germline of these patients has been used as evidence
of the classification of osteochondroma as a neoplasm U Clin Invest.
~i(J:]If(!:f:JI WHOC&Mik&tJon ofBont TumoN
C.l'Ap . . . . rwiii_IOiilllf .....
OD:dtohdrotna ntBIIItll •ara.tl C..
ChOIIdrCinll Eloln,..,.,ma
=..:":!;::.,._ lllllll""'lllr:-
f'loorno Qllll\¥lllomo
MLI!plo c:llond...,IIDIIs Moli&Mirt M'nr>homo, noto1ilei'Mio 6llOdllod
Chondroblolloml
ChOIIdrCinj!JOOidtbrotno tolti-
Chondr...rcomo Gionl ... blmo<
Ooml, ptMy,•nd~
Po"'hnl
Oodll!'nnt!Uod
-K:hyn\11
··--...
Mol~ In pnl ... blmo<
a.wdoiNI
..... _._
c::loorQII
o-.........
-,...,.
Ottedd uh lma
Oolaobluloml
Ool\..1111ohol
c::I\OIIdroblu1le
....... iyome
~ltiOIII·
L.._..._,..
Fbrol>lulle Upoml
Os\ooblullr: u_....me
-
T~
Smollodl
-·1:11--
N-.,rll!lmMOI"M
- ...... ......
J.ow.il*l•-1
SeoorldiJY
AGornoMfnoma
P -..J Mataa111C1c rnoli1.1'1on<Y
H~...r-
......l e _ ~~--~~~~
-..yomel bono cylt
llwnoploolle ft brotM Slmpb<)'ll
Fb-• Flbrou>dl'OIIIWI
Cla~L~ofibl'OI& ~l&.!lo
fllnllolllr\'lk-
....,.,11\onaoolh~
Ban1&1'1 tbroo10 t~
Mal.,..,nt tbroua hll1ioeylorno Erdholn>-(!h_d_
a.&li ""'' hiiiWIDIN
IDO I SECTION X.lt SOFT TISSUE AND BONE
...
P.t11oolotk Ftotllfts of Benwa lioN> Tu- ilJid T\lmOIJ.lllle ....,_
,... riiMiill Ueollfllft IaIIIII Jlll*llat.ft tiiOIII.(t
Mlq!Maa
Oo!oociiOIIdrr.ma ~!lr*ofl.llo lo-30 Col11ioeKIII>I*I bottJ -uoion
Ch<IM'o11111 HONI>II'eot; modulo Arrt Vbllo~callulor h)lalho <111111a8>
oll.ll&
ChoM'o-m.t Eplpllylll/lJlOpll:;sk lo-30 ChOIIdrald·lt.o mi!Jix;
olllll S.l~c:olk""'
.,...,..S nU<Iol
Choncto~ Mat&ptp>of 1.811 lo-30 li.IJ>cool~~~ -"""""""
~-
....
lobuleo ouiTOIIIdod 1W more
ooli ...l'!>indlo<>t!ll•-
......
Flbr<W <tiol>lulll
olllll
-
n.,_,.A>Ctlrlno
......
"""',..,.... Vtrll-;~1hnd
1.811
lo-30 Blood-1\lod • - ,.~Ill'
ftl>nlu& sopl.teo allnl c:olk
"""
QnploO)'IIII Mat&ptp>of 1.811 lo-30 Fll\clofllbd 'eystr/" lnod tot
CD'I~ tb:lva
HlernllliSorno \U1ebloo:~ot•nd 20-60 Colllol)' end/of CIIIIIITIOUO ol.!od
I& -I&
l..&,qblNIIi.
~~ !.' 1• J:! u1 ~mo.:-•~.ocllloftc.}.
r.tlfts of
Plllloktk;
u~~~.~~.1oM,-_.
Dflti11Mlttlft. llldl
llllii!Wrt
S.llelrt
c:r,r...,ltlonal
Flit-
"""~· lnd
"'>l~dl8&
"'"""1\)olo 20-80 V.IW!¥co11*11)011110 co1111a.!Jo
(N()S) 001!11811111W bono
Dod!ll'ooooillotod Malc>ll)olo :oliO ConwntfQ:\11 cb:lnrtOIIAXJrna +
hlltt1IIWCI&oPndlo<:OIIa,_
MMOIIdi)"'TTII ~1\)'11$ 20-8Q Und!11'...,,_...,.... colllllmor +
hyalnoe.tlop
C6Nr0111 E)>I~ '2!>-10 eo-tiona! tumorw!lh •bl.lldJnllorp
doorlllnorcolll
Gnu•
o• :w1Xln"16 Malc>ll)olo of I.So; 1~
low >-10
eor-tiollll Modl,jiiiY Olloold formed d!n>OIIyllf mollp!ft
.-..- ModiA~IY
<dis
l.lldtf~l~--p<'OI-
+ thl::i! - hbo<ull&
T~ ModiA~IY l!.liio64lllod""""" +........ ...,... +
hllttf molts~t•nt-
""-'
....-.,
C.rtel-lcl&
porl_,m
C.Mo<lnslila
Mldti~l~bmb-II'Oilf&ldon
+ thl::i! - tallocll-•
Allundlnt <OI1!1.,. ~ wtlh .,,_
- -
por!OIIoum ITIIII,Vllnt-
~
flbfoun::tf'l!a ~ll)oloc(IBI; l.lo!lpwrt iiplndlo '*In • _....,
""'f-.llD
l!fldofbono
MoiiiJ..rt~bro"' Malc>ll)olo of I.So; 20-8Q loloi~~Jm\ Ol>indlo-ln '*'rlf<llm
~ill rnoy-to - + h~ ...lll (ojhOt
l!fldolbono porUenu l1\lf be ..n)
-""""'
...... ·
~.~
~~
~l; Wllltea"Mj
poMI;Ula
Arty
>4l
Arty
V.<111>1y~l (m"-111) pWml
eollo
~lid<! Pftii(MII!.... IIMIIIrb
""""'"" ,....,.
--
Dlhllloll
Mlll•lii~ CoMo< <II llb!a :2fi-35 ~elll!..,ll>+~lrob!asb+_,or
llrna!llr bono
'tfc Arty
-
Mo.. >4l Mo!lpwrt Oj>lllldlll-; mOIJihol..:;/
IHC ~-'"' 011Jjn
c.rei~YA"na
-
ct..ooma e... o1 Rll: :oliO IRI>I.M ol.......:11111od<dloan-
OO<fUm In II1Y'I:Id lllllllx
~con••nd Anollornosln.J ......,.. ,.,.mall Ihod
"'""........ brt lifallti ~colla OR -.<loW
-~""'·
eollo In rny>dd bldqJJ'ound;
dlal0dlll1olle!HC
IDf I SECTION .... SOFT TISSUE AND BONE
--·ll!llalll 011
MX l b r t - Ql/lnct bo 01" 110<1
MO N~:Jdbant rnl!!lt<Uta
Ml lllllonl rnolo1alls
Mia f..un.l
Mlb Olllot-nt-
• loMJoh--•-wca•
fllqo lA
Tl NO
..........1.10 Gl, 2r.-gnldo, GX
SlqtiB T2 NO 1.10 Gl, 2 r.-~, GX
T3 NO 1.10 Gl, 2 "-17"k GX
T1 NO 1.10 G3, 4 Hilh 11"""
T2. r«l MO ~. 4 Hlah 11ad&
Tll NO 1.10 G3,4
M/T NO Mia AnyG
AnyT Nl AnyM AnyG
AnyT kryN Mlb AnyG
-
Gl
G2
1111
~11.00
Hl,lllpo
...........
Ml
$QIIA
Slqa 18
RtPNiordlsant...........,
01
Gl
T1
T2
MO
MO
Slqa IIA G2 T1 MO
Slqo liB G2 T2 MO
5lqo Ill Nrt /vrt Ml
804 I SECTION XI: SOFT TISSUE AND BONE
direct contact with this osteoid matrix, are the neoplastic tumor cells
which can be quite variable in appearance and include polyhedral cells,
spindle cells with variable nuclear atypia, small blue cells (resembling
EWS, see below), and large markedly atypical cells. The predominant
matrix produced by the tumor can be bone or osteoid, cartilaginous,
or fibrous. Historically, osteosarcomas have been subclassified on the
basis of the predominant matrix production (osteoblastic, chondroblas-
tic, or fibroblastic), but this classification has no prognostic importance.
Instead, as described below, osteosarcomas are best classified on the basis
of radiologic and/or pathologic features that have been shown to have
distinct prognostic implications (Am] Clin Pathol. 2006;125:555).
i. Central osteosarcomas arise within the medullary cavity and include
the following:
(a} Conventional intramedullary osteosarcoma is the prototypical
osteosarcoma, for which most of the above information refers.
Neoplastic tumor cells in conventional osteosarcoma are often
polyhedral or spindle-shaped with unequivocally malignant fea-
tures. Given that preoperative chemotherapy for these tumors is
the current standard of care (and has significantly improved the
5-year survival rate of this tumor from 20% to over 80% ), it is
important to carefully map the tumor in the resection specimen (as
discussed earlier) to determine the extent of tumor necrosis com-
pared to the volume of viable plus nonviable tumor since a favor-
able long-term outcome is associated with > 90% tumor necrosis.
Additional chemotherapy is often offered to patients with less
necrosis as a second-line attempt to further improve survival.
(b) Law-grade central osteosarcoma is a rare type of osteosarcoma (1%
to 2%) composed of a variably cellular spindle celVfibroblastic
proliferation that lacks the degree of cytologic atypia seen in con-
ventional osteosarcoma. In addition, bone production is usually
evident as irregular, somewhat thick, anastomosing, or branching
bony trabeculae. These trabeculae simulate the woven bone of
fibrous dysplasia or the longitudinal seams of bone in parosteal
osteosarcoma (see below), and are separated by a spindle-cell
stroma. Review of the radiologic findings often reveals subtle
signs of malignancy that are useful in making the diagnosis
(e-Fig. 48.34). This tumor has a much more indolent course com-
pared with conventional osteosarcoma; however, there is still a
high recurrence rate if the tumor is inadequately excised, often
with associated grade progression.
(c) Telangiectatic osteosarcoma is characterized by large, blood-filled
spaces separated by highly cellular fibrous septae that contain
markedly pleomorphic cells with a variable amount of osteoid
production (e-Fig. 48.35). Radiologically, this tumor is radiolu-
cent and expansile, and resembles ABC. Compared to other
osteosarcomas, it is more likely to present with a pathologic frac-
ture. Although not necessarily associated with improved survival,
this aggressive osteosarcoma is very sensitive to chemotherapy.
(d) Small cell osteosarcoma is composed of small round blue cells
and histologically resembles EWS except that there is histologic
evidence for osteoid formation (although it is often scant). It is
usually entirely radiolucent. It has a capricious clinical behavi01;
often but not always resistant to usual osteosarcoma chemother-
apy. Although the usual reciprocal chromosomal translocation
described in EWS (see below) has not been generally observed,
Chapter 48 • Bone Neoplasms and Other Nonmetabol ic Disorders I 8 07
this lesion has been shown to have membrane positivity for CD99,
which has led some authors to theorize that it is a variant of EWS
with divergent differentiation.
ii. Surface osteosarcomas. About 1 in 20 osteosarcomas occurs in asso-
ciation with the bone surface rather than in the medullary cavity. The
vast majority of these are low-grade tumors showing radiodensity and
osseous differentiation. They include:
(a) Parosteal osteosarcoma accounts for "'4% of osteosarcomas
and the majority of surface osteosarcomas. It characteristically
involves the posterior distal femur, is associated with the outer
fibrous layer of the periosteum, and tends to wrap around the
bone. Histologically, it consists of well-formed bony trabeculae,
often arranged in parallel streamers separated by a hypocellular
spindle stroma as seen in low-grade central osteosarcoma (e-Fig.
48.36). Cartilaginous differentiation is also common, often seen
as a cartilage cap and sometimes causing confusion with osteo-
chondroma. Radiographically, however, there is no continuity of
the interior of parosteal osteosarcoma and the medullary cav-
ity, the adjacent bony cortex is not continuous with the outside
of parosteal osteosarcoma (e-Fig. 48.37), and the intertrabecu-
lar spaces do not contain fatty or hematopoietic marrow. The
prognosis of parosteal osteosarcoma is similar to that of low-
grade intramedullary osteosarcoma. If inaccurately diagnosed as
benign, or if inadequately excised, these lesions will recur. Recur-
rences may be low grade, but they may also be high grade; low-
grade parosteal sarcoma undergoing high-grade transformation is
termed dedifferentiated parosteal osteosarcoma and has a prog-
nosis similar to conventional osteosarcoma.
(b) Periosteal osteosarcoma arises between the cortex and overlying
periosteum most commonly in the tibial or femoral diaphysis, and
is characterized by abundant cartilaginous matrix and a some-
what greater degree of cytologic atypia than is seen in parosteal
osteosarcoma.
(c) High-grade surface osteosarcoma is histologically identical to con-
ventional intramedullary osteosarcoma, except that it arises on
the bone surface. The prognosis is similar to that of conventional
intramedullary osteosarcoma.
3. Fibrous tumors and tumor-like conditions
a. Fibrous dysplasia. This space-occupying lesion has been classified vari-
ously as developmental, tumorous, or tumor-like. It usually presents as
a solitary lesion, although it may affect multiple bones in a single limb
bud distribution, or multiple bones without limb bud distribution. The
polyostotic form is one of the manifestations of the McCune-Albright
syndrome, which includes pigmented skin lesions and endocrinopathies.
The presence of activating G-protein mutations in both monostatic
lesions and McCune-Albright syndrome suggests that fibrous dysplasia
represents a true neoplasm UPediatr. 1993;123:509). Fibrous dysplasia
may be asymptomatic, but deformities, secondary fractures, and even
pain may be the presenting manifestation. Radiographically, the lesion
is almost always intramedullary, and it tends to affect those portions
of bone formed by endochondral ossification. While secondary corti-
cal atrophy may take place because of intramedullary expansion of the
lesion, fibrous dysplasia usually does not involve the cortex. Fibrous dys-
plasia is often expansile and results in modeling deformities of the host
bone. It is well circumscribed and radiolucent, but less radiolucent than
808 I SECTION XI: SOFT TISSUE AND BONE
the underlying bone that it has replaced; radiologists often refer to its
appearance as having a ground glass quality (e-Fig. 48.38).
Histologically, fibrous dysplasia consists of various combinations of
any tissue present in bone, so fibrous tissue, bone, cartilage, and vascu-
lar tissues are produced in various combinations. The usual microscopic
pattern, however, consists of loosely arranged, vascularized fibrous tis-
sue in which disconnected curved microtrabeculae of bone are disposed.
These trabeculae are not only woven in their collagen fiber pattern, but
when a section is examined under polarized light, the fabric of their col-
lagenous background forms a continuum with the fabric of the fibrous
tissue (e-Fig. 48.39). Cartilageformation is not unusual, and occasionally
cartilage is formed in such excess that lesions may be mistaken radio-
graphically and histologically for cartilaginous neoplasms. Treatment is
usually focused on relief of deformities or other morbid symptomatology.
The prognosis is usually excellent.
b. Osteofibrous dysplasia is a fibro-osseous lesion and is invariably seen in
the tibia, fibula, or both. Its peak incidence is in the first two decades
of life. Radiographically, the lesion is radiolucent and usually based on
the cortex; it may extend to the medullary cavity. The lesion may be
unilocular or multilocular; while it tends to be circumscribed, it may
also diffusely involve the diaphysis and cause secondary bowing defor-
mities (e-Fig. 48.40). Histologically, osteofibrous dysplasia resembles
fibrous dysplasia except that the microtrabeculae of bone tend to be
rimmed by appositional osteoblasts even at their very earliest synthesis
(e-Fig. 48.41). The fibrous stroma tends to be more cellular than in
fibrous dysplasia, and is less contiguous with the trabeculae under polar-
ized light. The lesional bone also tends to mature at the periphery with the
surrounding normal bone. The lesional bone tends to undergo sponta-
neous involution with time, although sometimes it behaves more aggres-
sively.
c. Nonossifying fibroma {fibrous conical defect) is the commonest space-
occupying lesion of bone, estimated to affect one in four individuals.
Even though it is thought to be developmental, in rare cases, the lesion
behaves as a tumor of limited biologic potential. Unless it is associated
with a fracture, patients are generally without symptoms; the lesions are
typically discovered incidentally during the course of evaluation for some
other condition and the routine radiographs are virtually diagnostic. The
lesion is a well-circumscribed radiolucent defect in the metaphyseal cor-
tex with scalloped sclerotic borders, and it is almost always longer in the
cephalocaudal than axial direction (e-Fig. 48.42).
Histologically, the lesion consists of spindle cells arranged in a
distinctly storiform pattern. A fair number of multinucleated giant
cells, histiocytic cells with foamy cytoplasm, and histiocytes contain-
ing hemosiderin pigment (e-Fig. 48.43) may also be present. While bone
formation is not observed (hence the name), lesions that have fractures
or microscopic infarctions are admixed with reactive bone. Because this
lesion may be focally cellular, it is important to review the radiographs
to avoid misdiagnosis. Most nonossifying fibromas are self-healing and
do not require clinical intervention.
d. Desmoplastic fibroma. This rare tumor occurs in adolescents and young
adults, with the mandible being the most commonly affected site. Radi-
ologically, it often expands the involved bone, is entirely radiolucent,
and is usually well circumscribed. Histologically, it is composed of bland
fibroblastic or myofibroblastic cells in a background of collagen iden-
tical to that found in desmoid tumors and/or soft tissue fibromatosis
Chapter 48 • Bone Neoplasms and Other Nonmetabolic Disorders I 809
tumors with a high metastatic rate. Most tumors are radiolucent with
poor margination; howeve.t;, a sclerotic rim is occasionally identified.
While angiosarcomas are often solitary, hemangioendotheliomas tend to
present as multifocallesions in the same bone or in the same limb bud
distribution and may be mistaken for skeletal metastases (e-Fig. 48.58).
Angiosarcomas are usually characterized, at least focally, by the pres-
ence of irregularly anastomosing vascular channels that are lined by
highly atypical endothelial cells, but they may largely consist of solid, pat-
ternless aggregates of polyhedral or spindle cells (e-Fig. 48.59). Poorly
differentiated angiosarcomas may fail to express vascular markers. It
should be noted that angiosarcomas are known to express cytokeratins,
which is an important consideration when metastatic carcinoma is in the
differential diagnosis. Most angiosarcomas are associated with a poor
outcome.
Epithelioid hemangioendothelioma is composed of cords, nests, or
sheets of plump cells that, in their attempt to form vessels, are often vac-
uolated, and some of the vacuoles may contain erythrocytes. An extracel-
lular myxoid or hyalinized stroma is characteristic, although not always
identified. Immunohistochemical reactivity with vascular markers such
as CD31, CD34, and Factor VIII-related antigen can be used to con-
firm endothelial differentiation, and may help to exclude a diagnosis
of metastatic signet-ring cell adenocarcinoma. Epithelioid hemangioen-
dothelioma may have an indolent course.
7. Epithelial tumors
a. Adamantinoma comprises <1% of malignant bone tumors. There is a
wide age distribution, with the median age of patients between 25 and
35 years. The tibia is most frequently involved, followed by the fibula,
and both sites synchronously. Radiologically, an intracortical radiolucent
lesion is evident, which may involve the medullary cavity (e-Fig. 48.60).
Histologically, classic adamantinoma is composed of epithelial cells
having a basaloid, tubular, or squamoid appearance; has a predomi-
nantly spindle cell pattern; or consists of a mixture of the two patterns
(e-Fig. 48.61); a storiform fibroblastic proliferation that contains vari-
able amounts of woven or lamellar bone may also be present. Rarely,
the tumor is predominantly composed of the latter component, with
only rare scattered epithelial cells (single or in small nests, sometimes
only detected by immunohistochemistry); such tumors have been termed
as having an "osteofibrous dysplasia-like pattern" or as "differenti-
ated adamantinomas,, and usually present in patients younger than
20 years. Adamantinomas are invariably immunopositive for various
keratins and epithelial membrane antigen, and often also for vimentin.
Classic adamantinomas are indolent tumors with a high local recur-
rence rate and metastasis in only about 20% of patients; differentiated
adamantinomas almost never metastasize.
b. Metastatic carcinoma is the most common tumor affecting the skeleton,
which is the third most common site to be involved by metastatic carci-
noma after the lungs and liver. Primary carcinomas of the breast, lung,
prostate, kidney, and thyroid gland compose >80% of all bone metas-
tases. Radiologically, metastatic deposits can be radiolucent or radio-
dense or can display a mixed pattern (e-Fig. 48.62). Given its high
incidence, metastatic carcinoma should always be in the differential diag-
nosis of solitary or multiple bone lesions in patients older than 40 years.
The histology usually resembles that of the primary carcinoma, if it
is known. It should be noted that a fibroblastic, osteoblastic, or vascular
response to the metastatic tumor may occasionally be quite prominent
Chapter 48 • Bone Neoplasms and Other Nonmetabolic Disorders I 81 3
and overshadow the tumor cells, which may only be focally evident
(e-Fig. 48.63). Consequently, immunohistochemistry may reveal isolated
subtle tumor cells that are not obvious in small biopsy specimens.
8. Miscellaneous neoplasms
a. EWSJPNET. In addition to its classic presentation in the diaphyses of
long bones, the tumor can involve axial bones such as the pelvis and
ribs, as well as soft tissues (see Chap. 46) and other organs. EWSIPNET
has been described at various ages, but most patients are younger than
20 years. Although patients usually have pain and a mass, they may
also present with fever and leukocytosis suggestive of an infectious pro-
cess. Radiologically, a destructive, permeative lesion is usually evident,
often with an overlying multilayered but discontinuous "onion-skin"
periosteal reaction (e-Fig. 48.64 ). Occasionally, there is a large soft-tissue
mass with no obvious bone destruction on the routine radiographs, but
the intraosseous component becomes evident on aCT scan or MRI.
Histologically, EWS/PNET is the prototype for "small blue round cell
tumors" as it is composed of sheets of such cells, often with glycogen-
containing clear cytoplasm (e-Fig. 48.65). Evidence of neuroectodermal
differentiation may be manifested by extracellular eosinophilic neuropil-
like structures or Homer-Wright rosettes (composed of groups of tumor
cells that surround a central core of eosinophilic extracellular mate-
rial). Immunohistochemically, EWS/PNET shows characteristic strong
cell membrane immunoreactivity for CD99, negativity for CD45, and
variable reactivity for neural markers such as neuron-specific enolase,
synaptophysin, CD57, neurofilament, and S-100 protein. Cytokeratins
may also be positive.
A characteristic feature of this tumor is the presence of a recurrent
balanced reciprocal translocation involving the EWS gene on chromo-
some 22 and a member of the Ets family of genes, the most common of
which (85% of cases) is the FL11 gene on chromosome 11 (Br] Cancer.
1994;70:908). Immunohistochemistry, polymerase chain reaction, and
fluorescent in situ hybridization have all been used to confirm the diag-
nosis by detecting expressed FLI1, the fusion transcript, or the translo-
cation itself, respectively. The prognosis of EWS has greatly improved
with multimodality treatment, and 5-year survival rates now approach
70%.
b. Chordomas are derived from notochordal remnants and account for "'4%
of malignant bone tumors. Most tumors present after 30 years of age
with a peak incidence between 50 and 60 years of age. The midline of
the axial skeleton, particularly the sacrum and the base of the skull, is
usually affected. Radiologically, a lucent lesion is seen with scattered
calcifications; there is often a large associated soft-tissue component
(e-Fig. 48.66). Histologically, chordomas are composed of lobules of
tumor in which sheets, cords, or nests of vacuolated, eosinophilic to
clear cells are embedded in a myxoid matrix (e-Fig. 48.67). Chordo-
mas express cytokeratins, epithelial membrane antigen, and S-100 pro-
tein, an immunohistochemical profile that is helpful in distinguishing
this tumor from chondrosarcoma (which is cytokeratin and epithelial
membrane antigen negative). Chordomas are aggressive tumors that are
most notable for local recurrence when incompletely excised, but also
have a metastatic potential. A controversial entity, "chondroid chor-
doma," characterized by areas of mimicking hyaline cartilage, appears
to have a better prognosis, whereas "dedifferentiated chordoma," with
its high-grade sarcomatous component, is associated with a very poor
outcome.
814 I SECTION XI: SOFT TISSUE AND BONE
9. Cystic/cyst-like lesions
a. Simple bone cyst is an intraosseous space-occupying lesion consisting of
an accumulation of fluid. The lesion is usually lined by a thin mem-
brane composed of flattened cells of unknown type that may be involved
in the production of the fluid, which usually appears serous. The base
of the lesion is usually situated at an active growth plate, and the cyst
is more or less maintained by continuous remodeling of the bone around
the area of fluid pressure. The lesion is circumscribed and radiolucent,
and usually involves the proximal humeral, femoral, or tibial region
(e-Fig. 48.68). It does not expand the bone or cause deformity unless
there has been a fracture with displacement before healing, and it tends
to be symmetric. While fluid may not be obvious radiographically,
the contents are demonstrable by MRI. Histologically, the diagnosis is
one of exclusion and depends upon correlation of the imaging, operative
findings, and lack of any other diagnostic tissue. Because most simple
cysts are treated conservatively by injection of steroids or other scleros-
ing agents, it is unusual to see the lining of a simple cyst histologically
unless there has been repeated fracture.
b. lntraosseous ganglion. This is a subarticular defect in the cancellous bone
filled with mucoid fluid. The bone surrounding the defect is remodeled
and sometimes sclerotic (e-Fig. 48.69). The defect is histologically similar
to the subarticular cysts associated with overlying osteoarthritis (geodes)
except that the subarticular plate and articular cartilage are radiographi-
cally intact in patients with intraosseous ganglia. The lesion is presumed
to arise from a microscopic continuity of the subarticular plate with the
joint space that either cannot be detected by imaging studies or that has
healed; this etiology would allow pressurized synovial fluid to come in
contact with the intertrabecular medullary space. There are no diagnos-
tic features histologically as the concentrated fluid is practically acellular
and resembles the contents of tenosynovial ganglia.
c. Aneurysmal bone cyst. This peculiar lesion derives its name from its
expansile character. It is not a true cyst, but rather a collection of
blood-filled spaces that are separated by fibro-osseous tissue septa con-
taining a varying amount of multinucleated giant cells and immature
bone. Most ABCs occur prior to 20 years of age. The lesion usually
arises as an eccentric radiolucent lesion in metaphyses of long bones
(e-Fig. 48.70). When it is central and symmetric it can often be distin-
guished from a simple cyst because it causes the bone to become wider
than the growth plate. The bone destruction associated with ABC is per-
haps the most rapid associated with any osseous lesion. While its inter-
nal edge tends to be well marginated radiographically, ABC sometimes
extends across adjacent bones, particularly if it arises in the spine. Com-
plementary imaging studies, particularly MRI, reveal peculiar fluid-fluid
levels on T2-weighted axial and sagittal views (e-Fig. 48.71); these levels
are caused by the signal differences in erythrocytes and plasma, reflect
erythrocyte sedimentation, and demonstrate that blood in intact ABC is
both unclotted and stagnant or slow moving.
Histologically, the lesion has vascular spaces that progress from very
small capillary spaces to very large sinusoids separated by fibrous septae
and sometimes by bone (e-Fig. 48.72). Within the septae are fibroblasts,
scattered multinucleated giant cells, and osteoblasts associated with the
bone production. Rarely, the lesion is almost entirely solid, although
sometimes the solid variant may demonstrate fluid levels on imaging. In
about half the cases, there is some other lesion associated with the cyst
and admixed with curetted fragments; this has been termed secondary
Chapter 48 • Bone Neoplasms and Other Nonmetabolic Disorders I 81 5
*All e-figures are available online via the Solution Site Image Bank.
816
Chapter 49 • Metabolic Diseases of Bone I 81 7
bone
0 OC precursor @] 08
eoc £2.:::>
"'-.(
bone lin ing cell
bone. Decalcification required for standard paraffin processing removes the dis-
tinction between newly synthesized osteoid and mature calcified bone. In contrast,
undecalcified plastic sections can be stained in several ways to demonstrate osteoid.
The von Kossa stain, a silver-based stain used with a basic fuchsin counterstain,
shows calcified bone matrix as dark brown or black, whereas the unmineralized
matrix (osteoid) appears pink-red (e-Fig. 49.4A). A trichrome stain, either Goldner
(e-Fig. 49.3) or modified Masson (e-Fig. 49.4B ), also distinguishes mineralized bone
from osteoid. These latter stains allow easier interpretation of cellular morphology
than the von Kossa, and also highlight peritrabecular or marrow fibrosis.
A second critical marker of OB function is tetracycline labeling. Tetracycline
family antibiotics are calcium-chelating fluorochromes that bind to actively min-
eralizing bone surfaces, can be taken orally, and are well-tolerated. They are given
in two courses, separated by 2 weeks (see below). If bone formation is active dur-
ing both intervals, examination of unstained sections by fluorescence microscopy
demonstrates two bright bands of labeling (a double label) (e-Fig. 49.5). Similarly,
active bone formation during only one of the labeling periods yields a single tetra-
cycline label (e-Fig. 49.5). Combination of the extent of labeled trabecular bone
surface and the distance between labels provides the mineral apposition rate and
bone formation rate. In a normal subject, most surfaces with osteoid, as seen on
trichrome or von Kossa stains, have single or double labels.
During normal endochondral bone development, cartilage formed at the growth
plate is replaced by bone in the primary spongiosa through the action of OCs.
Toluidine blue stains cartilage purple, and cartilage may be found within trabeculae
near the growth plate in children (e-Fig. 49.6). However, a finding of entrapped
818 I SECTION XI: SOFT TISSUE AND BONE
cartilage in an iliac crest bone biopsy in an adult, or > 1 em from the growth plate
in a child, is indicative of OC dysfunction such as in osteopetrosis.
II. INDICATIONS FOR BIOPSY, TISSUE SAMPLING, AND PREPARATION
A. Indications for biopsy. The most common indications for metabolic bone biopsy
are end-stage renal disease (ESRD) and unexplained hypercalcemia or hyper-
phosphatemia, osteoporosis unresponsive to therapy, or suspected osteomala-
cia. Patients who have multiple or unexplained fractures (particularly if they
are failing to heal), unexplained bone pain, or an elevation in serum alkaline
phosphatase may also be candidates.
B. Biopsy procedure. A critical component of evaluation of metabolic bone biop-
sies is in vivo fluorochrome labeling of bone via use of a regimen of tetracycline
(250 mg orally four times a day [PO qid]) for 3 days, followed by a 14-day inter-
val, then 3 more days of therapy (250 mg PO qid). Biopsy is performed on the
third day after the last dose (biopsy interpretation may therefore be confounded
by recent therapeutic use of antibiotic drugs in the tetracycline family; similarly,
inadequate labeling can be caused by malabsorption syndromes or by taking
tetracycline with meals, dairy products, iron-containing medications, antacids,
or calcium supplements).
Biopsy is performed as an outpatient procedure; the most accessible site for
biopsy is the anterior iliac crest. When obtained, the specimen should be placed
directly into 70% ethanol.
C. Sample preparation. The specimen should be fixed in 70% ethanol for at least
48 hours, and this solution is suitable for shipping and long-term storage at room
temperature. Following dehydration with xylene, the specimen is mounted in
methyl methacrylate, and the tissue core is sectioned parallel to its long axis at
5 to 7 J.£-m thickness using a tungsten blade. Undecalcified sections are stained
with von Kossa, Goldner or modified Masson trichrome, and toluidine blue.
One section is decalcified and stained with H&E. Thicker 10-J.£-m sections are
coverslipped without staining for examination under fluorescence.
D. Quantitative versus qualitative evaluation. The American Society for Bone and
Mineral Research has described a nomenclature for a basic set of structural
and kinetic features identified by nondecalcified bone biopsy (] Bone Miner
Res. 1987;2:595), and there are two commercially available systems that allow
quantitative analysis based on these standards (OsteoMeasure, OsteoMetries,
Inc. and Bioquant Osteo II, BIOQUANT Image Analysis Corporation). Refer-
ence values, based on somewhat limited populations, have been published (Engl
I Med. 1988;319:1698; I Bone Miner Res. 1988;3:133; Bone. 2000;26:103;
I Bone Miner Res. 2004;19:1628). Although some laboratories perform quan-
titative analysis on all specimens, qualitative assessments are often adequate
for diagnosis of individual patients. Quantitative analysis is most useful in the
setting of research studies.
Ill. DIAGNOSTIC FEATURES OF METABOLIC BONE DISORDERS
A. Osteoporosis. In the setting of osteoporosis or osteopenia, biopsy establishes
the rate of bone remodeling (turnover), degree of mineralization, architectural
integrity, and effects of treatment. lliac crest bone biopsy is a poor indicator
of bone mass, as bone volume/tissue volume (BV!TV) is variable within this
region. However, trabecular connectivity, which describes the intactness of the
trabecular meshwork, correlates with bone mass. In osteoporosis/osteopenia,
trabeculae are very small and often appear as isolated islands (low connectiv-
ity), rather than as an interconnected grid (good connectivity) (e-Fig. 49.7).
In high-turnover osteoporosis, osteoid surface is enhanced (e-Fig. 49.8A), with
normal or increased numbers of OCs and OBs. The extent of double tetracycline-
labeled trabecular bone surface is also increased, although the distance between
the double labels is usually normal (e-Fig. 49.8B). In low-turnover osteoporosis
Chapter 49 • Metabolic Diseases of Bone I 8 19
or osteopenia, there is little osteoid, few OCs or OBs, and rare or absent tra-
becular double tetracycline labels (e-Fig. 49.9). Even in low-turnover states,
double labeling in the cortex is usually present, and is a good positive control
for adequate tetracycline dosing and specimen processing.
Many patients with osteoporosis have been treated with bisphosphonates,
often for several years. Although biopsy studies have shown that normal
turnover is intact in most patients (]AMA. 2006;296:2927), some cases of
severely suppressed bone turnover have been reported and are associated
with increased fractures (] Clin Endocrinol Metab. 2005;90:1294; N Engl]
Med. 2006;355:2048). Bisphosphonates target the OC, decreasing resorption
and enhancing apoptosis, producing changes that can readily be seen in tis-
sue sections with OCs appearing either hyperchromatic with pyknotic nuclei
(e-Fig. 49.10A) or round and unpolarized (e-Fig. 49.10B).
B. Osteomalacia. In osteomalacia, newly formed organic bone matrix fails to min-
eralize normally, and the result is wide osteoid seams, often greatly increased
in extent along the trabecular bone surface (e-Fig. 49.11A). Some osteoid may
be completely unlabeled by tetracycline (e-Fig. 49.11B), whereas other surfaces
may show irregular and diffuse fluorescence (e-Fig. 49.11C); double tetracycline
labels are rare. Florid osteomalacia due to nutritional rickets (vitamin D defi-
ciency) is rare, but milder cases may be found unexpectedly and bone biopsy is
the only definitive diagnostic tool. Osteomalacia is also seen in fluorosis, usually
caused by excessive fluoride in drinking water (e-Fig. 49.110 and E). The degree
of osteomalacia in hypophosphatasia, a rare deficiency of alkaline phosphatase
activity, can be quite severe (e-Fig. 49.11F).
Tumor-induced osteomalacia (110, also known as oncogenic osteomalacia)
is a rare form of osteomalacia that can occur in both adults and children. lliac
crest bone biopsy in TIO shows the typical features of osteomalacia described
above (e-Fig. 49.12A). The bone manifestations of TIO are caused by mes-
enchymal tumors that secrete the phosphatonin FGF-23, leading to hypophos-
phatemia due to renal phosphate wasting. The tumors are typically small and
slow-growing, and can be difficult to detect. If not visible by CT or MRI, ses-
tamibi or octreotide scans may be useful, as well as targeted venous sampling
for FGF-23 (Expert Rev Endocrinol Metab. 2009;4:435). Histologically, the
majority of the tumors in cases of TIO are best characterized as phosphaturic
mesenchymal tumor (mixed connective tissue variant) and have low cellularity,
bland spindled cells, distinctive "grungy" calcified matrix, and an incomplete
rim of membranous ossification and/or formation of an osteoid-like matrix at
least focally (Am J Surg Pathol. 2004;28: 1) (e-Fig. 49.12B). They may be locally
infiltrative into muscle or fat (e-Fig. 49.12C), have prominent vessels reminiscent
of hemangiopericytoma, myxoid change, hemorrhage, or OCs. TIO tumors typ-
ically stain for FGF23 but not S100, CD34, or smooth muscle actin. Resection
of the solitary tumor mass results in complete resolution of hypophosphatemia
and clinical symptoms in the majority of cases.
C. Renal osteodystrophy. Chronic kidney disease-metabolic bone disease (CKD-
MBD) has been defined as a systemic disorder of bone and mineral metabolism
due to CKD manifested by one or more of the following: (i) abnormali-
ties of calcium, phosphorus, PTH, or vitamin D metabolism; (ii) abnormal-
ities of bone turnover, mineralization, volume, linear growth, or strength;
(iii) vascular or other soft tissue calcification (Kidney Int. 2006;69:1945). CKD-
MBD is associated with increased morbidity and mortality. Three predomi-
nant patterns in bone biopsy have been described: high bone turnover with
osteitis fibrosa (hyperparathyroid bone disease), low bone turnover (includ-
ing low-turnover osteomalacia and adynamic bone disease), and mixed uremic
osteodystrophy.
820 I SECTION XI: SOFT TISSUE AND BONE
SUGGESTED READINGS
Monier-Faugere MC, Langub MC, et al. Bone biopsies: a modern approach. In: Avioli LV, Krane
SM, eds. Metabolic Bone Disease and Clinically Related Disorders. San Diego, CA: Academic
Press; 1998:237-273.
Recker RR, Barger-Lux MJ. Bone biopsy and histomorphometry in clinical practice. In: Favus M,
ed. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism. Washington,
DC: American Society for Bone and Mineral Research; 2006:161-169.
Joints and Synovium
Peter A. Humphrey
I. NORMAL ANATOMY. Joints are composed of the ends of contiguous bones and the
associated soft tissue elements, including cartilage, ligaments, tendons, and syn-
ovium. Diarthrodial movable joints, which are the most common type, are usually
covered by hyaline cartilage. Histologically, this articular cartilage is hypocellular
with a glassy extracellular matrix composed mainly of collagen, proteoglycan, and
water. Embedded within the matrix are chondrocytes within surrounding spaces
(lacunae). There are four zones in articular cartilage-superficial, intermediate,
deep, and calcified (e-Fig. 50.1)"' and chondrocytes have a different appearance
depending on their location. Those near the surface of the articular cartilage are
small and flattened; in the middle zones, the chondrocytes are more rounded and
arranged in columns. The deep and calcified layers of articular cartilage are sepa-
rated by a thin, basophilic line known as the tidemark, which represents the miner-
alized front. The calcified cartilage base interdigitates with underlying subchondral
bone.
Ligaments, which join two adjacent bones, are formed mainly of collagen. At the
insertion site onto bone the ligamentous tissue is calcified. Tendons are connective
tissue structures connecting muscle to bone. Microscopically, scant fibroblasts are
found within parallel collagen bundles.
Synovium is a glistening white membrane with delicate villous projections
that lines the inner surface of the joint capsule (Am j Clin Pathol. 2000;14:773)
(e-Fig. 50.2). The inner lining surface is created by synovial cells, including fibrob-
lastlike cells and histiocytes, which are arranged as a thin two- to three-cell layer
of closely packed cells with elliptical nuclei and abundant cytoplasm. A fibrous or
fibroadipose supporting layer lies beneath the synovial celllayet: Synovium also
lines the flexor tendons of the hand and bursae (subcutaneous and subtendinous
sacs).
II. GROSS EXAMINATION AND TISSUE SAMPLING. Joint or synovial soft tissue is received
as needle core tissue, as fragments from arthroscopic or open synovectomy, as
fragments from revised total joint arthroplasty, or as excisions of soft tissue ten-
don sheath or extra-articular masses. If fragments are received, the numbe.t; colo.t;
shape, and aggregate size of the fragments should be recorded. If meniscus tissue
is submitted, fibrillations or tears should be noted. If chalky white deposits are
identified, some of the tissue should be placed into absolute (100%) alcohol to
preserve the crystals. Decalcification may be required for calcified cartilage, bone,
or soft tissue. Fragments that appear different from normal should be selected for
embedding, along with representative unremarkable-appearing fragments.
A. Revision arthroplasties. The presence or absence of necrosis, purulent exudate,
and foreign material should be recorded. The explanted prosthesis should be
described, including any identification numbers or defects.
B. Soft tissue tumors. For excised soft tissue tendon sheath or juxta-articular masses
size, color, consistency, shape, and nodularity (single vs. multiple) should be
provided. The outer surface of the specimen should then be inked, and cut
sections should be characterized as to color, presence or absence of hemorrhage
and necrosis, and distance of tumor to inked margin. One section per centimeter
"'All e-figures are available online via the Solution Site Image Bank.
822
Chapter 50 • Joints and Synovium I 8 23
and incomplete excision. Accordingly, margin status and a high mitotic rate
should be reported.
2. Giant cell tumor of tendon sheath, diffuse type (pigmented villonodular synovi-
tis) presents as an intra-articular or extra-articular tumor. The gene expres-
sion profile of the neoplasm resembles that of activated macrophages (Arthri-
tis Rheum. 2006;54:1009), and structural rearrangements of 1p11-12 have
been reported in the lesion; however, these special studies are not needed to
establish the diagnosis.
a. The intra-articular form typically involves the joint space of the knee of
young adults, although ankle, hip, and shoulder involvement have also
been reported. Involvement of the synovium can be localized with the
formation of either nodular or pedunculated masses; the more common
diffuse form affects virtually the entire synovium. Grossly, tissue from the
diffuse form removed by open synovectomy is spongy, diffusely thickened,
and brownish-yellow. Microscopically, subsynovial fibrohistiocytic cells
with uniform round to oval nuclei are seen. Foam cells and iron pigment
are always present, and giant cells are common and tend to be arranged in
groups. Aside from the villous structures, the overall appearance is similar
to nodular tenosynovitis (e-Fig. 50.12).
The localized form has an excellent prognosis and a low recurrence rate
when managed surgically, whereas the diffuse form has a reported recur-
rence rate of up to 46%. Radiation treatment has shown mixed results.
Combined surgical and nonsurgical approaches may be necessary, and in
some patients, total joint arthroplasty may be the only effective treatment
(jAm Acad Orthop Surg. 2006;14:376).
b. The extra-articular form is much less common; the knee, ankle, and foot
are predominant sites of occurrence. Some, but not all, cases are exten-
sions of the intra-articular form; the rare exclusively extra-articular cases
likely arise from the synovium of bursa or tendon sheaths. Grossly, the
tumor is a large, multinodular, and white to yellow-brown mass, with-
out villous projections. Microscopically, there are sheets of rounded to
polygonal mononuclear cells with the formation of clefts and pseudog-
landular spaces. Giant cells are fewer in number compared with the giant
cell tumor of tendor sheath, localized type. Xanthoma cells, spindle cells,
and chronic inflammatory cells are present. Scant data exist on outcome
for these patients, although recurrence rates appear high, at 40% to 50%.
3. Malignant giant cell tumor of tendon sheath is rare type of sarcoma and can
be diagnosed when a histologically benign giant cell tumor of tendon sheath
is admixed with overtly malignant areas, or when malignancy is seen in a
recurrence of the benign tumor. Histologic indicators of malignancy include
diffuse infiltrative growth, scant giant cells, cytologic atypia, necrosis, and a
mitotic count of> 10 per 10 HPFs.
4. Synovial chondromatosis is a benign nodular cartilaginous proliferation aris-
ing in the synovium of joints, bursae, or tendon sheaths. It is usually monoar-
ticular in distribution, involving the knee or hip in adults. Grossly, there
are multiple osteocartilaginous nodules, each measuring <1 mm to several
millimeters in diameter, which may be embedded within synovium and/or
mobile as loose bodies (note that loose bodies can also be seen in epiphyseal
osteonecrosis, osteochondral fracture, and osteochondritis dissecans, so their
presence is not diagnostic). Microscopically, the nodules of hyaline cartilage
show dusters of chondrocytes that can show nuclear atypia with nuclear
enlargement, hyperchromasia, and binucleation (e-Fig. 50.13), findings that
should not be viewed as evidence of malignancy. Clonal chromosomal abnor-
malities have been cited as evidence for a neoplastic process, but cytogenetics
is not used for diagnosis. Local recurrence after excision occurs in ""15% of
Chapter 50 • Joints and Synovium I 8 27
General Principles of
Cytopathology
Brian Collins
IV. FNA. One of the most challenging and dynamic areas of cytopathology is FNA.
Virtually any organ or abnormality (either palpable or localized by imaging tech-
niques) can be subjected to FNA. The technique essentially provides a microbiopsy
of cellular material (cells and tissue) for microscopic examination and a wide vari-
ety of ancillary tests. The method permits immediate interpretive evaluation which
makes possible real-time adjustments of the biopsy procedure, appropriate speci-
men triage, and directed ancillary testing.
A. Principles. The procedure is minimally invasive and accurate. At its core, FNA
involves the use of a thin bore needle with a cutting end moved in a piston motion
to obtain cells and tissue. The cellular elements present within the needle are
then processed for diagnosis (e-Fig. 51.1).*
B. Technique. While simple, proficiency and adequacy require an understanding
of the procedure and experience in its use. It is the cutting, bevelled end of
the needle and capillary action of a repetitive "piston-like" motion movement
which provides the cellular elements necessary for diagnosis. The appropri-
ate application of needle movement is critical since insufficient movement will
not adequately sample the tissue, and prolonged movement can lead to hemodi-
lution and entrapment of tissue in clot. The appropriate number of tissue passes
and appropriate negative pressure will vary for each individual case and clinical
presentation.
Negative pressure during the procedure (typically applied by an empty 10- or
20-cc plastic syringe attached to the needle) can be useful but is not always nec-
essary. There are many solid cellular lesions/neoplasms that can be adequately
sampled without aspiration, including lymph nodes and the thyroid. Vascular
organs and neoplasms benefit from a nonaspiration FNA technique since aspi-
ration can lead to bleeding and specimen hemodilution.
Needles utilized can range from 22 to 27 gauge. Larger gauge needles tend to
cause more bleeding and paradoxically tend to be less diagnostic. Needle length
can vary significantly (5/Sth in. up to 8 in.) and will depend on what is required
to reach the lesion.
C. Procedure. In broad terms, the needle is directed to the area of interest by either
palpation or under image guidance.
1. Palpable. This method involves any lesion that can be localized or identified
by manual palpation. It must be stressed that FNA of every location requires
an understanding of the associated local anatomy, especially for areas in
the head and neck, thyroid, chest wall, and various soft tissue locations.
Superficial FNA of palpable lesions is well tolerated, minimally invasive, and
has a very low complication rate.
2. Image guidance. This method involves utilizing an imaging modality to direct
the needle, most commonly, ultrasound and CT imaging. The principles of
the FNA biopsy itself remain the same. In some cases, a stylet within the
needle is helpful; it is used to prevent sampling of organs as the needle moves
toward the lesion, and then removed once the needle tip is in the desired
location. An immediate assessment of the FNA sample makes it possible for
the pathologist to guide the clinician to ensure that a diagnostic sample is
collected and that the appropriate ancillary studies are initiated at the time
of the procedure.
D. Materials
1. Palpable. The materials required are simple and readily available. They can
be conveniently organized in a small basket or box, which can be easily
transported from the laboratory (e-Fig 51.2).
*All e-figures are available online via the Solution Site Image Bank.
Chapter 51 • General Principles of Cytopathology I 8 31
2. Image guidance. The appropriate type of needle and the FNA approach are
determined by the clinical setting. On-site presence of the cytopathology
team with glass slides, fixative (alcohol), needle rinse tube (normal saline
or RPM!), stains (Diff-QuikTM ), and a microscope, will help optimize the
likelihood that a diagnostic sample is obtained and that appropriate ancillary
studies are initiated.
E. Clinical application
1. Immediate evaluation. Immediate evaluation is the standard of care because it
provides several advantages. First, it guides the procedure and thus ensures
a maximum effort to obtain a diagnosis. By examining the slides during the
procedure, immediate evaluation can be used to help decide if more FNAs
are necessary; guide the needle position placement by determining if lesion
or nonlesional material is present; and if nondiagnostic, support a decision
to move to a potential second site. Second, with immediate evaluation, the
trade-off between the length of the procedure and number of biopsies can be
managed to balance the requirement for diagnostic tissue without unnecessar-
ily extending the length of the procedure. By assuring a diagnostic procedure,
the number of nondiagnostic samples can be minimized and repeat proce-
dures and delays in diagnosis can be avoided. Third, the cytopathologist can
identify those cases where tissue for appropriate ancillary studies needs to be
collected to enhance diagnostic accuracy. These situations include identifying
a lymphoproliferative process where rinse material is sent for flow cytometry
analysis, abscess/granulomatous inflammation where microbiologic cultures
are indicated, poorly differentiated neoplasms where a cell block can provide
material for immunohistochemical evaluation, and lesions where cytogenet-
ics/molecular diagnostics can contribute to diagnosis and treatment.
F. Specimen processing
1. Direct smears are stained by the Papanicolaou and Diff-Quik™ methods.
2. Liquid medium. FNA samples that are collected in a liquid medium provide
a wide variety of options for further analysis. While it is helpful to perform
needle washes during the FNA procedure, the best chance for a sufficient
cell block involves direct (no slides prepared) dedicated FNA samples placed
in the liquid medium (saline or RPM!). Cell block preparation affords a
variety of options and advantages; sections of the cell block contribute to
the morphologic evaluation of the lesion, and also provide cellular material
for immunohistochemical analysis, interphase FISH, and for a wide range of
PCR-based molecular tests.
SUGGESTED READINGS
Crothers BA, Tench WD, Schwartz MR, et al. Guidelines for the reporting of nongynecologic
cytopathology specimens. Arch Pathol Lab Med. 1999;133:1743-1756.
Gupta PK. University of Pennsylvania aspiration cart (Penn-A-Cart): an innovative journey in fine
needle aspiration service. Acta Cytol. 2010a;54:165-168.
Gupta PK. Progression from on-site to point-of-care fine needle aspiration service: opportunities
and challenges. Cyto;ournal. 2010b;7:6.
Wright TC, Massad LS, Dunton C], et al. 2006 consensus guidelines for the management of women
with abnormal cervical cancer screening tests. Am] Obstet Gynecol. 2007;197:346-355.
SECTION XIII
Ancillary Methods
and eosin (H&E). Small specimens may be completely utilized for frozen section
slide preparation, but if possible, a portion of the tissue should be preserved for
routine handling to avoid freezing artifacts that can compromise interpretation
of the permanent sections (e.g., in the case of brain biopsies). For larger tissue
samples, judgment must be exercised in gross sampling so that the area(s) of
highest diagnostic yield is selected for frozen section. Cytological imprints from
tissues can be an important adjunct in diagnosis, especially for hematolymphoid
abnormalities, lymph node biopsies, and thyroid lesions.
C. Interpretation. The interpretation of frozen sections requires integration of the
histologic morphology in the H&E-stained sections; the gross features of the
specimen; information from the surgeon regarding the origin of the tissue,
the indication for the consultation (including the clinical history, radiological
findings, and intraoperative observations); and the ways in which the frozen sec-
tion diagnosis will affect the operative strategy. In some difficult cases, it may be
necessary to request additional tissue for frozen section analysis. If additional
tissue cannot be obtained, deferral of a definitive diagnosis pending examination
of formalin-fixed paraffin-embedded sections is acceptable. In routine clinical
practice, such deferrals are employed in <5% of frozen section diagnoses.
D. Communication of findings. Clear communication of a concise diagnosis to the
surgeon is the last step of the intraoperative consultation. Usually, there is a
narrowly defined clinical problem that frozen section is to solve; this should
be specifically and unambiguously addressed in the diagnosis. The diagnosis is
written and signed, with the date and time, by all interpreting pathologists and
made part of the final pathology report. If the written diagnosis is verbally trans-
mitted, the pathologist should first confirm with the recipient of the information
the identity of the patient (using two identifiers) and the surgeon. The operating
room staff member taking the diagnosis should repeat it back to the pathologist
to confirm accurate communication.
Certain phraseologies tend to be resistant to misinterpretation. For instance,
"negative for malignancy" or "positive for malignancy" are generally under-
stood well. Since some diagnosis can easily be incompletely heard or misunder-
stood in a busy operating room, the diagnosis should always be repeated back
as a safeguard.
E. Accuracy of frozen sections. The accuracy of frozen sections will vary from insti-
tution to institution on the basis of the types of surgical cases evaluated and the
experience of the involved pathologists. Table 52.1 highlights the fact that the
accuracy of frozen section diagnosis is dependent on the anatomic site. Regular
self-audits of the frozen section service are desirable so that surgeons and pathol-
ogists are aware of the performance characteristics of the modality in their own
hands. Such audits of single institutions, and pooled data across hundreds of
institutions, show that accurate diagnoses are made overall in >95% of cases,
while discordance with the final diagnosis occurs in 1% to 2% of cases. Deferral
of the diagnosis until permanent section diagnosis occurs in 1% to 4% of cases.
F. Sources of error in frozen sections. Errors can be divided into errors of interpre-
tation and errors of sampling; both usually result in false-negative diagnoses.
False-positive diagnoses are rarer, likely because experienced pathologists tend
to appropriately defer to permanent section rather than making an incorrect
diagnosis on substandard material.
Misinterpretation accounts for about 40% of errors overall (Arch Pathol
Lab Med. 1996;120:804; Arch Pathol Lab Med. 1996;120:19). Interpretations
of frozen sections are more prone to this error than interpretation of permanent
sections due to the presence of artifacts that are not encountered in routinely
fixed, paraffin-embedded sections. Some tissues are more likely to show signifi-
cant artifact, especially those with high fat content, such as most pelvic lymph
nodes.
: I 'I i.t:! I J.fiS (Ell.mpM of,..,.. 8lc1loft EntU1111oft
~ ... ,...,.It
111111 •Ill• ~111110111 t;allllllfl*ln C.• liM..,
Br- Umllod <-tall
OoM< 73'!!; f<lf-1-of~ -f<lf -ldo•<tf~
Qllbll.ddor 95" Mloln UOild'> ~e l.leoM""' r«Jol-" wlll:ll ~ 1o re<~~~hd
m~.n~lb*Jn
lMt
I.
~dlem-thdlll--k;
Kam- ... -,.,.....,1-
2. R"'"...-.1Minod.-,.,h.-11Aarc;ol<tlomo
3. - ·... hlj>ol-*col<h>me
w"
-IY - 1 tlfnpl!
99'!!;
!I0"-&6'V ts•-a~
lholll: dofoml reb> of only 3-<l'l!;
Possibly -fill c- 1M!)
nodi& ftrft'lo:!!' •·tse
broaot~r
I.Jmflll -lor liq!r\1 -uxrs• 21>-4011'.' UmiiDd IU1d lloponclontupon h> lymflll , _ - (,..IB<I)
o..y L'lltll; em>rlUI dbpmpoltioN!oty rep!a.nlld amo~ miiC:h>us buno"
!mil
98'l!; Mloln ueod f<lf d ~ .,_,.
S'l(;
l'l!. AtyiJbll duetll otllltiU,..., e<~~" ~he 0$111ns or P«nC-.
may
"'""'""" ., pBnal)l-;-
loa% for m1111no
mlmlo """'""""'' dofomll reb> of 6-7ll>
Po1l1111y!DU ~ (t>r poooUI)'r'Did ... L111o11J f<lf ~~- d poodip-olci ..MIUS nonparo!llyroid tlr.wili;
...,....lll,.oU lbsue) lrlllloqume 1o< d\ii,Crmb Ill pomtl'floU """'""""· ... 1o< ""'...,-.
- . . . . ~m 1\ypooplosll
Sktl, Mil'-,..... Uptl> 50ir. Slrol\it1d""""rapd <-lull
Sltl.nomwn<mO Alatt 2l'. buod c n l.leoM f<lf 1fll) ..-ruoe~cn <If me!D'Ia " ~he ree ' <don cftumcre Mil
Inil......,
- ......
"'''"'"""' """"follow!" bon! Oil and tumOfloftho "'"" -~ly- oaro. ond
Mollo'"*"""~
nr.,.'*l IM. lnodeqllll& t>rt>llbl~r 1MIons, .. ttt..amplnJ reqlftd Is not pnodlool
<- tallU: lloUot t>r lodcl1b wllh I>'lilhJY erchbdur&; dofiml M1D I& ~'l!.
«When f'll:ldM. with t&bnklon'lfltl..._ (cO.2: mm) n tcn~ik~Med ·~ltht&.'
~ ftOdM • wbmlenltl,.ttm•ft~ ·~'
~ poMe trmpl!- .. --.....
'Datil on ..,.,..tlonwtll _,,...,._II""""' ast>llo••up _,,...,._ or&nolf>•tlm\l!d.
Chapter 52 • Frozen Sections and Other Intraoperative Consultations I 8 35
Sampling errors occur in two ways. The first is sectioning error. Diagnos-
tic tissue may be present in the frozen block, but the block may not be faced
sufficiently for the lesion to be present on the actual frozen section slides;
the diagnostic material may then be found in routine permanent sections of the
residual frozen block, a scenario that accounts for 10% to 15% of errors. The
second type of sampling error is gross sampling error. This accounts for about
45% of errors overall, and is seen when the diagnostic tissue is not in the portion
of the specimen sampled by the frozen section; good gross pathology skills will
minimize, but never eliminate, this problem. Furthermore, it is not feasible to
completely sample larger lesions by frozen section, and so sampling errors will
always exist for large lesions with heterogeneous composition.
Discrepancies between a frozen section and the final (permanent section)
diagnosis should be documented in the final surgical pathology report, along
with the reason for the discrepancy. If the discrepancy is of clinical significance,
the pathologist should immediately alert the surgeon to the change in diagnosis.
G. Anatomic sites deserving special mention. The anatomic sites with the most dis-
crepancies between frozen and permanent sections are skin, breast, lymph nodes
for metastatic disease, the female genital tract, and thyroid. Frozen sections do
have a role in the surgical management of disease in these sites, but since loss of
diagnostic material during the performance of frozen sections is unavoidable,
each case must be critically evaluated as to whether the frozen section diagnosis
will add enough value to warrant this loss.
1. Skin. Frozen sections for margin assessment in the excisions of large non-
melanoma skin cancers such as basal cell carcinoma and squamous cell car-
cinoma are indicated if the lesion has vague infiltrative borders or is in a
location in which wide excision is not possible, as in the case of tumors
of the eyelid, nose, or ear (Arch Pathol Lab Med. 2005;129:1536). If the
borders of a lesion are well defined, frozen sections are not as necessary.
It is widely agreed that the intraoperative primary diagnosis of pigmented
lesions is ill-advised. The determination of prognosis and subsequent man-
agement of melanoma requires an accurate assessment of Breslow thickness,
and frozen artifact may cause so much specimen distortion as to make a
depth of invasion assessment meaningless in both permanent and frozen sec-
tions. Freezing artifact compounded by actinic damage, frequent in patients
with pigmented lesions, also conspires to obscure histology. Subtle changes,
such as intraepidermal spread by single melanocytes, are very difficult to
appreciate in frozen sections, making the method a poor choice for margin
assessment as well. For example, the false-negative rate for lentiginous spread
of melanoma by frozen section evaluation may be as high as 50%.
2. Breast. Once commonplace, the initial pathologic diagnosis of breast tumors
is now very rarely made during surgery, since methods for detection of smaller
tumors and more sophisticated treatment algorithms (as opposed to mastec-
tomy only) now usually lead to diagnosis on the basis of needle core biopsy or
aspiration cytology. Margin evaluation is sometimes requested during breast-
conserving procedures, but the high fat content of breast tissue makes frozen
specimens technically difficult to section and prone to freezing artifact. Eval-
uation of margins by imprint histology is an alternative to frozen section,
but it is insensitive unless the margins are grossly involved, in which case the
examination is unnecessary (Arch Pathol Lab Med. 2005;129:1565). As a
rule, an intraoperative gross examination of a lumpectomy specimen is used
to determine whether or not there will be additional excisions.
3. Lymph nodes for metastatic disease (including axillary sentinel lymph nodes for
breast carcinoma)
a. Sentinel lymph nodes. Although current diagnostic modalities have reduced
the need for breast frozen sections, requests for frozen sections of breast
836 I SECTION XIII: ANCILLARY METHODS
sentinel lymph nodes have increased (e-Fig. 52.1A and B).* Unfortunately,
frozen sections on sentinel nodes consume considerable tissue; if no metas-
tases are identified, further evaluation of the node has therefore been sig-
nificantly hampered (e-Fig. 52.2).
Numerous studies have indicated that the sensitivity of frozen section
evaluation of sentinel nodes is about 60%, although the specificity is near
100% (Mod Path. 2005; 18:58). It has been argued that the metastases
that are typically missed by frozen section are submicroscopic (<0.2 mm in
greatest dimension) or are detected by cytokeratin immunohistochemistry
alone, and that since the significance of these classes of metastases is not yet
cleat; the low sensitivity of the approach underestimates its clinical utility.
A sensible middle ground seems to be to perform frozen sections when the
clinical history and gross examination of the node give a high degree of
suspicion for metastasis. Imprint cytology, as a routine measure, is another
possible approach to intraoperative sentinel lymph node evaluation.
b. Other lymph node frozen sections. The intraoperative evaluation of lymph
nodes can be performed by frozen section or by imprint cytology, and if
indicated, tissue should be allocated for flow cytometric analysis, routine
histology, and molecular diagnostics.
4. Thyroid. The widespread use of fine needle aspiration has altered the approach
to frozen section evaluation of the thyroid. Frozen section evaluation of
thyroid excision specimens is best performed in conjunction with imprint
cytology, the latter of which is more sensitive for the nuclear grooves, inclu-
sions, and chromatin-clearing characteristic of papillary carcinoma. Since
the distinction between follicular adenoma and carcinoma by frozen section
(e-Fig. 52.3) would require thorough sampling of the tumor and capsule for
invasion and angioinvasion, frozen section is poorly suited to this application
(Can I Surg. 2004;47:29).
5. Female genital tract. Of all the neoplastic processes of the female genital tract,
ovarian neoplasms are probably the best suited to intraoperative frozen sec-
tion diagnosis (Arch Pathol Lab Med. 2005;129:1544; lnt I Gynecol Cancer.
2005;15:192). Even so, borderline mucinous neoplasms of the ovary have a
significant rate of discordance between the frozen section and permanent
section diagnoses, primarily due to sampling error. The diagnosis of cervical
dysplasia is fraught with difficulties due to frozen section artifact and low
concordance with permanent section diagnosis.
Ill. OTHER INTRAOPERATIVE CONSULTATIONS. Intraoperative nonmicroscopic consulta-
tions are often required even though no frozen section is needed. Indications include
gross diagnosis (such as benign simple ovarian cyst or leiomyoma of uterus); gross
confirmation of the presence of a lesion or mass; identification of a margin or
region of interest that requires special sampling for permanent sections; specimen
orientation; triage of tissue for ancillary testing modalities that require special pro-
cessing or fixatives (in which case good judgment is required to ensure a balance
between the need for tissue for routine histopathologic evaluation and the need for
tissue for specialized testing); and collection of tissue for tumor banking or research
studies.
A. Opening of a viscus organ for gross examination and fixation. Gastrointestinal
specimens commonly require opening in the operating room so that the surgeon
can see whether or not lesional tissue has been excised. In partial intestinal
excisions for inflammatory disease, in which the risk for malignancy is increased,
a careful intraoperative examination may inform the surgeon of a previously
undetected tumor, which may have implications for additional surgical therapy.
*All e-figures are available online via the Solution Site Image Bank.
Chapter 52 • Frozen Sections and Other Intraoperative Consultations I 8 37
Similarly, gross examination and opening of the adnexa and/or uterus can
aid in determining the extent of surgery that is required. Such gross examination
often leads to frozen sections when ovarian surface papillary excrescences are
identified, or when solid or complex architecture features are discovered in an
ovarian cyst.
B. Tumor for banking must be chosen so that the remaining lesional tissue material
will still be suitable for a complete diagnostic evaluation. The banked tissue
should not include a surgical margin, or have an important relationship to an
anatomic structure that would impact staging or the need for adjuvant therapy.
In cases of small specimens, it may be necessary to defer banking for the sake
of a thorough diagnostic evaluation.
SUGGESTED READINGS
Lechago J. Frozen section examination of liver, gallbladder, and pancreas. Arch Pathol Lab Med.
2005;129:1610.
Marchevsky, AM, Changsri C, Gupta I, et al. Frozen section diagnoses of small pulmonary nodules:
accuracy and clinical implications. Ann Thorac Surg. 2004;78:1755.
Young MP, Kirby RS, O'Donoghue EP, et al. Accuracy and cost of intraoperative lymph node
frozen sections at radical prostatectomy.] Clin Pathol. 1999;52:925.
Electron Microscopy
Ashima Agarwal and Frances V. White
~~td'etlld Dlillt><IIGn.,._tjmplm\aand
,_..,..
maanch)mlll t.nd carcttoma
norlymplldd Ill"'""
Glondulu 11!11111dl&.m
III1Brcoluir 111111111
Ml:roo.ilolll cotoroo1ielo Glondulu 11111111dlllt1 of kl0111111co11oh ol ~~·~
d-tra<t ol-tSo..rmorn&&
~piMmit 1Dnoftbo11l Squom0U141j)ttt.ollum lden'l:tflatSon d squtmaua
dll!n-ln "f''thollllumm
PIM1 . .1011Cnl0llnd Ma~IU)11t .... ld-ol-mu
maiii'IOIOmn
,_ur~ Philleo ~11l011doct1no•nd kl0111111co11oh ~ nouroondoctlno and
....
~•mal
Lll\pltslna aolk
MU~rmd~'&
l d - ~ Llnprhllll
.....
pA>IIIir_,. tuoll"" Llnpo!t""
~
EB:Inln<lan.,OOjcob
-.0111
Olld/llt OCher
memb10no ablo11ono
840 I SECTION XIII: ANCILLARY METHODS
*All e-figures are available online via the Solution Site Image Bank.
Chapter 53 • EIectron M icroscopy I 84 1
I. RECEIPT, ACCESSIONING, AND GROSS DISSECTION. Most, if not all, biopsies and
large tissue specimens are routed to the pathology laboratory. Under normal cir-
cumstances, the specimens are received in 10% neutral buffered formalin (formalin
begins the fixation process and prevents autolysis and decomposition). The speci-
men is first logged into the surgical pathology computer system and given a unique
identifying number, referred to as an accession number or case number. Once acces-
sioned, the specimen is taken to the gross dissection room; depending on the prac-
tice setting, residents, fellows, pathologist assistants, and/or trained technicians are
responsible for gross processing of the specimen under the supervision of an attend-
ing pathologist. Gross processing entails describing the specimen by its size, shape,
color, and overall general appearance, followed by placing samples of the tissue
in processing cassettes (for biopsies, the entire tissue specimen is placed in a cas-
sette; for larger specimens, regions of tissue are sampled according to established
protocols). Each cassette is labeled with the accession number as well as a part
designator and number; this numbering scheme is designed to allow the location
of a particular section of tissue within the context of the whole specimen.
II. PROCESSING. The loaded cassettes are stored in 10% neutral buffered formalin
until automated tissue processing. The normal processing cycle is ""'8 hours long
and in general is designed to remove the water from the specimen and replace it
with paraffin. Automated, closed-system tissue processors utilize agitation, vac-
uum, and increased temperature to optimize the process. In general terms, the
process is as follows. First, the tissue is subjected to 10% neutral buffered formalin
to ensure complete fixation. Complete fixation aids in the dehydration steps and
prevents tissue shrinkage and other artifacts caused by excessive or rapid dehydra-
tion; chemically, formalin fixation produces methylene cross-links between nucleic
acids and/or proteins. Once the tissue is well fixed, it is subjected to several changes
of graduated alcohols in a gradient starting at 70% and ending at 100%, a process
that removes water from the tissue at a slow controlled rate designed to prevent
excessive shrinkage and disruption of the architecture and cellular components.
After complete dehydration of the tissue has been accomplished, a clearing agent
is used to remove the alcohol and allow tissue infiltration by paraffin; this clearing
agent must therefore be miscible in both alcohol and paraffin. Xylene is most often
used for this purpose, although commercial xylene substitutes are available. In the
next step of processing, heated paraffin infiltrates into the tissue. Paraffin is a solid
at room temperature but has a relatively low melting point, and so is a good choice
as an infiltration and embedding media. While pure paraffin wax was used in the
past, current commercially available paraffins are formulated with various plastic
polymers to allow better infiltration and a more rigid crystalline structure, both of
which aid subsequent microtomy.
Ill. EMBEDDING. Properly fixed and processed tissue sections are embedded in molds to
prepare them for microtomy. The tissue is removed from the cassette and oriented
in the base of a mold that is of a size to allow paraffin to surround the tissue
section. During embedding, the tissue is oriented with the understanding that the
surface placed down in the mold will become the face of the tissue block, and
will thus be the surface cut into first by the microtome blade. Attention must be
842
Chapter 54 • Histology and Histochemical Stains I 843
rinse, the slides are differentiated using acid alcohol, rinsed, and the hematoxylin
is "blued" by immersion in a weak ammonia water solution. The slides are rinsed
again, placed in 80% alcohol, and stained with eosin. Excess eosin is removed by
alcohol rinses, and the slide is prepared for mounting with a coverslip and resinous
media by removal of the alcohol using xylene rinses.
VI. OTHER FREQUENTLY USED HISTOCHEMICAL STAINS. Tissue stains range from very
simple to complex in methodology, and can be used to demonstrate most major
tissue elements relevant to pathologic diagnosis. They are based on the chem-
istry of various dyes and metals, and most were developed prior to the advent of
immunohistochemistry. In general, a histochemical stain consists of the main chem-
ical reaction that demonstrates the specific tissue element of interest, followed by
chemical reactions that provide staining of the background uninvolved tissue ele-
ments, often including nuclear detail. Histochemical stains are usually grouped by
the tissue element they stain.
A. Carbohydrates. In humans, carbohydrates exist as various sugars and polymers
linked to proteins. Simple sugars cannot be detected by standard histochemical
procedures because they are water soluble and thus removed during process-
ing; however, polymers such as glycogen can be detected. Naturally occurring
polysaccharides can be classified into four groups on the basis of their histochem-
ical staining differences: neutral polysaccharides (Group I), acid mucopolysac-
charides (Group II), glycoproteins (Group III), and glycolipids (Group IV).
Amyloid must also be included because, even though it is not a carbohydrate,
its histochemical staining properties are similar to those of polysaccharides. The
histochemical stains most often used to detect carbohydrates and differentiate
various types of carbohydrates are Aldan blue, colloidal iron, mucicarmine, the
periodic acid-Schiff (PAS) reaction, Congo red, and Thioflavin T.
1. Mucicarmine. The mucicarmine method is used to detect tissue mucins and
utilizes the tissue dye carmine. When carmine is reacted with aluminum, it
forms a compound that has a net positive charge and is attracted to the
negative acid groups of epithelial mucins. Metanil Yellow and Weigert's
hematoxylin are used as counterstains and produce yellow staining of the
background tissue elements and blue-black nuclear staining (e-Fig. 54.1).*
2. Alcian blue, a phthalocyanine basic dye, forms salt bridges with the acid
groups in mucopolysaccharides. Staining tissue sections in an Aldan blue
solution at pH 1.0 produces staining of only sulfated mucopolysaccha-
rides, while staining at pH 2.5 produces staining of all mucopolysaccharides.
These two methods make it possible to differentiate sulfated from carboxy-
lated mucopolysaccharides; further differentiation between mucosubstances
of connective tissue origin and that of epithelial origin can be achieved by
the addition of hyaluronidase digestion (e-Fig. 54.2).
3. Colloidal iron. The colloidal iron stain is based on the chemical principle that
at low pH colloidal ferric ions can be absorbed by both carboxylated and
sulfated mucopolysaccharides, as well as other glycoproteins. The absorbed
ferric ions are detected by use of the Prussian blue reaction (see below).
4. Congo red. Congo red reacts with cellulose and amyloid. The dye is a lin-
ear molecule that attaches to amyloid in a sheet-like fashion resulting in
so-called apple green birefringence when subjected to polarized light. This
"apple green" birefringence is considered specific for amyloid in Congo red-
stained tissue sections (e-Fig. 54.3).
5. Thioftavin T. Thioflavin Tis a fluorescent tissue dye that has an affinity for
amyloid. Thioflavin T fluoresces yellow to yellow-green when the tissue
*All e-figures are available online via the Solution Site Image Bank.
Chapter 54 • Histology and Histochemical Stains I 845
possible to halt the differentiation at the point when the elastic fibers are the
only tissue elements still stained. The tissue section is then treated with van
Gieson's solution, which contains the dye acid fuchsin; in a very strong acid
solution the dye selectively stains only collagen. Picric acid, used to maintain
the proper pH during staining, stains the rest of the tissue elements yellow.
The VVG stain demonstrates red-stained collagen, black elastic fibers, and a
yellow background (e-Fig. 54.9).
5. PTAH. The PTAH stain requires mordanting of the tissue section in Zenker's
fixative before staining. Because phosphotungstic acid is present in the stain-
ing solution in excess over hematoxylin, all of the hematoxylin is bound into
a tungsten-hematein lake, which selectively binds to cell nuclei, fibrin, and
cross-striations in muscle fibers. The excess unreacted phosphotungstic acid
stains the remaining tissue elements red to red-brown.
6. Pentachrome. The pentachrome stain is a compound stain that essentially
combines the elastic fiber staining of a modified VVG stain with a modified
trichrome stain. Alcian blue is first used to stain mucosubstances, and then
iron hematoxylin is used to stain elastic fibers. Following the differentiation
of the iron hematoxylin, a combined crocein scarlet and acid fuchsin solution
is used to stain muscle, cellular cytoplasm, amorphous ground substance,
and collagen red. Phosphotungstic acid in solution is then used to decolorize
the collagen and amorphous ground substance, which is then subsequently
stained yellow using a saturated alcoholic safran solution.
7. Oil red 0. This stain is used to demonstrate fat in tissue sections, or lipid
droplets in cell cytoplasm. The dye Oil red 0 is highly soluble in lipids, and
when used in solution with isopropanol is actually more soluble in fat than
in alcohol.
This stain requires the use of frozen section tissues because the alcohol
and xylene steps in standard paraffin processing remove virtually all lipids
from the tissue. The staining itself is fairly straightforward: frozen tissue
sections are cut, fixed with formaldehyde, and then stained in the Oil red 0
solution. The sections are next rinsed free from any excess stain and then
counterstained using hematoxylin. The stain results in blue cell nuclei and
bright red staining of fat droplets.
C. Microorganisms. There are many different stains that can be performed to
demonstrate microorganisms, specifically bacteria and fungi, in tissue sections.
The most commonly used stains are the acid-fast bacteria (AFB), the Fite modi-
fication of the AFB, Gram, Grocott methenamine silver (GMS), Warthin-Starry,
and PAS.
1. Gram stain. The tissue Gram stain is not much different from the standard
Gram stain performed in the microbiology lab. In the tissue Gram stain, how-
evet; after the use of crystal violet to demonstrate gram-positive bacteria by
a blue colot; basic fuchsin (a red dye) is used to demonstrate gram-negative
organisms as well as cell nuclei. Differentiation of gram-positive and gram-
negative bacteria is still a critical step; overdifferentiation is a common stain-
ing error. The final step in the Brown-Hopps Gram stain involves treating
the tissue with a picric acid solution that renders the background yellow.
In addition to identification of bacteria, the stains can be used to demon-
strate some cases of actinomyces, Nocardia infections, coccidioidomycosis,
blastomycosis, cryptococcosis, aspergillosis, rhinosporidiosis, and amebiasis
(e-Fig. 54.10).
2. AFB. The tissue AFB stain is simply a modification of the standard Ziehl-
Neelsen and Kinyoun stains that are based on the fact that carbol-fuchsin,
a solution created by reacting basic fuchsin with phenol in alcohol, is sol-
uble in lipids. Tissue sections are first treated with the carbol-fuchsin solu-
tion and then differentiated using acid alcohol; bacteria that have waxy,
Chapter 54 • Histology and Histochemical Stains I 847
lipid-containing cell walls resist decolorization with acid alcohol and are said
to be "acid fast." A methylene blue counterstain is used to highlight other
tissue elements and provide a background to highlight the red microorgan-
isms (e-Fig. 54.11). A slight modification of this procedure can be used to
specifically stain for Nocardia species in tissue sections. Another modifica-
tion of this stain, known as the Fite AFB stain, is used when Mycobacterium
leprae is suspected (e-Fig. 54.12).
3. GMS. This stain utilizes most of the same chemical reactions and principles as
the JMS stain. In the GMS stain, however, a stronger oxidizer, chromic acid,
is used instead of the weaker periodic acid. Since the cellular walls of fungi are
very thick and contain much more carbohydrate than basement membranes
and reticulin fibers of the surrounding tissue, the stronger oxidizer allows for
creation of dialdehyde groups from the carbohydrates of the fungi cell walls
with overoxidation and subsequent destruction in basement membranes and
other carbohydrate structures in the tissue section. The GMS stain utilizes a
light green counterstain, resulting in fungus cell walls that are various shades
of black to taupe in a light green background (e-Fig. 54.13).
4. Warthin-Starry. The Warthin-Starry method is used primarily for the demon-
stration of spirochetes, but other bacteria are also stained. The procedure is
based on the principle that bacteria in general and spirochetes in particular
have the ability to bind silver ions.
The staining procedure therefore involves impregnation of the spirochetes
in the tissue with silver ions, with subsequent reduction of these ions to metal-
lic silver using a developer containing hydroquinone. The stain demonstrates
black spirochetes against a yellow to pale brown background. The spiral mor-
phology of this form of bacteria can be fully appreciated by the use of this
method (e-Figs. 54.14 and 54.15).
5. PAS. This stain is often used for the demonstration of fungi in tissue, but is
most helpful when a counterstain of light green is applied. The method used in
stains for microorganisms is no different than when used for carbohydrates.
D. Nervous system. Most of the stains used on tissues from the nervous system are
for demonstration of either nerve fibers or myelin sheath. Two commonly used
stains for central nervous system tissues are the Bielschowsky and the Luxol fast
blue.
1. Bielschowsky. The Bielschowsky and all of its modifications are silver stains
that follow the principles and general steps of the reticulin stain. In the
Bielschowsky technique, the tissue sections are impregnated with a 20%
silver nitrate solution and then treated with an ammoniacal silver solution to
which formaldehyde has been added. Nerve endings, neurofibrils, neurofib-
rillary tangles, and neuritic plaques are all stained black.
2. Luxol fast blue. Luxol fast blue, a phthalocyanine dye that is soluble in alcohol,
is attracted to bases found in the lipoproteins of the myelin sheath. For this
stain, tissue sections are treated with Luxol fast blue over an extended period
of time (usually overnight) and then differentiated with a lithium carbonate
solution. Since Luxol fast blue has a strong affinity for the lipoproteins of the
myelin sheath, it remains bound to these lipoproteins even after removal from
other tissue elements. The myelin sheath is stained blue against a colorless
background.
E. Pigments and minerals. Pigments are substances deposited in the interstitium of
tissues, or as inclusions or granules in the cytoplasm of cells. Pigments can be
derived from minerals such as iron and calcium, or can be endogenous such as
melanin. The following staining techniques are used for the demonstration of
the most commonly encountered pigments.
1. Iron. The Prussian blue reaction is the most common staining technique for
the demonstration of iron in tissue sections. Prussian blue stains only weakly
848 I SECTION XIII: ANCILLARY METHODS
bound iron. Strongly bound iron, such as iron in hemoglobin, will not stain.
The principle of this stain is simple: when treated with potassium ferro-
cyanide in an acidic solution, ferrous ions in tissue react to form an insoluble
blue pigment. A nuclear fast red counterstain is usually applied to demon-
strate the background tissue morphology (e-Fig. 54.16).
2. Urates. A modified GMS stain can be used to demonstrate uric acid crystals.
No oxidation of the tissue sections is performed; instead, the sections are
reacted in the methenamine solution for an extended period at an elevated
temperature. Silver ions deposit on the uric acid crystals which in turn reduce
the silver ions to metallic silvet; so no toning is necessary. A light green
counterstain is usually applied, and the resulting stained section demonstrates
black uric acid crystals in a green background.
This stain requires the use of alcohol-fixed tissues because uric acid is
soluble in water.
3. Calcium (von Kassa method). The von Kossa method to stain for calcium is
very simple. Tissue sections are incubated in a 5% silver nitrate solution
under a very strong light source. The silver ions deposit on the calcium and
are reduced to metallic silver by the strong light, in much the same pro-
cess as occurs in photographic film. The stained section is rinsed free of
any unreacted silver ions by a sodium thiosulfate wash, and then counter-
stained with nuclear fast red to highlight the background tissue morphology
(e-Fig. 54.17).
4. Copper (Rhodanine method}. Copper can be demonstrated in tissues using
several methods, but the most sensitive method employs rhodanine. Tis-
sue sections are subjected to a saturated solution of 5-(p-dimethylamino-
benzylidene) rhodanine in aqueous solution. The rhodanine reacts with
proteins that have bound copper rather than directly with the copper itself.
The excess stain is rinsed from the sections, and the sections are counter-
stained with Mayer's hematoxylin, an aqueous hematoxylin that will not
overstain the rhodanine reaction. This tissue stain demonstrates bound cop-
per as a granular red pigment with pale blue cell nuclei (e-Fig. 54.18).
5. Argyrophil staining. Argyrophil substances within a cell bind silver ions. They
are "silver loving" but do not reduce silver to its visible metallic form. There
are several different techniques for the demonstration of argyrophil sub-
stances, all of which are chemically similar to the Warthin-Starry technique.
A solution of silver nitrate is used to impregnate the argyrophilic substances
in the tissue, and a reducing solution containing hydroquinone is then used
to reduce the bound silver ions to metallic silver. Nuclear fast red is often
used as a counterstain. By this approach, argyrophilic substances are stained
black.
6. Argentaffin staining (Fontana-Masson method). Argentaffin substances not only
bind silver ions like argyrophilic substances, but also reduce bound ionic sil-
ver to metallic silver without the use of a developer or other reducing agent.
This property of argentaffin substances, which include melanin, underlies
the Fontana-Masson stain. An ammoniacal silver solution is used to treat
tissue sections, and the argentaffin substances within the tissue not only
bind the silver ions in the solution, but also reduce them to metallic silver.
Gold chloride is used as in the reticulin stain to tone the metallic silver from
brown to black. Nuclear fast red is the counterstain of choice for this stain
(e-Fig. 54.19).
7. Bile pigments. Bile pigment stains are used on liver sections to distinguish
bile pigments from lipofuchsin. Fouchet's reaction demonstrates biliverdin,
bilirubin, and most other bile pigments. The tissue sections are treated with an
aqueous solution of trichloroacetic acid and ferric chloride, which renders an
emerald green precipitate, and von Gieson's solution is used as a counterstain
..,.
,,[1:)1 J%!'1 Common HlltrKMmlr:~l Stalftl
Al<lon bU. pH 2.5
Al<lonbu pH 1.0
n.. •...... d•lllilllllllll
II. addle""'""~
or~y ...- - m-~-
!lid
Bluo
Bluo
Cdlollllllron 8oth colta\)lllod and suffdld m~ Bluo
-OIIdn and Ill! li)'al~
MuciconnN E1i1H11oJ MII'*'O: ~C<I/OCOIOIUio Red
PAS N-1 MUOOI>Ot.-ehOIIdao, !ltiooiBII ~
-
tw · nwnt mfll'l'lbra.M&, .and fur«!
0<1\§>red AmjtJid Al>llio>-
Thlallr.t1T AmjtJid
R-.lh R!llk:Uin flbo"' Ble<:t
Trlch"""" Noolol,......,, m.- Ble<:t. bbl. nld
JMS Booomont mtllnb!OIIoo 81e<:t
PTAH Fltm,-m--. Deop purplo
Pti!rtttd'f'om• Nudl!ll1 ~·,..n, m~ .-Wstle~bdira Bl&d<, )Oli<Nt, rod
Fltn\, mwdt, muct1 B!tct, r4!d, tiue
...
OlradO lilt Red
VVG Elllllcflbo"' Ble<:t
AFII Add faot baclll Red
Fa& ..VB ~ Red
Gram Dl'll~~lram Q,. n>-1""lllwl
~bllcll -;Ill»
G,. n>-noplllo
bllcll: ral
-·-
OM$
W.r111~
G-
Fuhll
s~
- r l l , PIJ1i<>Jiarlf
Splrocholasand~
Bacllrll, prlmorllf H. ~
--fljlorl TtUIIO to ble<:t
81e<:t
Bl&d<
Black
Bluo
B-k;( N..,.onclhp. """""' flbf!lt, lllnposand Ble<:t
rigors of formalin fixation and paraffin processing but not acid decalcification;
therefore, application of the stain to bone marrow specimens requires the use of
nonacid decalcification). In the Leder method, tissue is treated with a solution
of naphthol-chloroacetate and pararosaniline, and reaction with the cellular
chloroacetate esterase forms a red precipitate. Hematoxylin is used as the coun-
terstain to demonstrate nuclear detail (e-Fig. 54.20).
G. Staining. Table 54.1 presents tissue stains listed by their most common name,
the tissue element that they demonstrate, and the resulting coloration or specific
result.
SUGGESTED READINGS
Bancroft JD, Gamble M, eds. Theory and Practice of Histological Techniques. 5th ed. Edinburgh,
UK: Churchill Livingston; 2002.
Carson FL. Histotechnology: A Self-Instructional Text. 2nd ed. Chicago: ASCP Press; 1997.
Sheehan DC, Hrapchak BB, eds. Theory and Practice of Histotechnology. 2nd ed. Columbus, OH:
Battelle Press; 1980.
Immuno histochemistry
Peter A. Humphrey
B 00 00 00
00 00 00
00 00 00
00 00 00
00 00 00
00 00 00
00 00 00
00 00 00
00 00 00
00 00 00
~ gg ~ gg ~ gg
~ ~ ~
)~ J~ J~
~~ ~~ ~~
)~ ~~it~~~y \~~~~~~
~ Secondary o Peroxidase
Q antibody
2. Adl'tlialtllll....,oslt)>lll¥,.mmun-n - · ohould
3. Tho flllOr\ _,ld alto ln<blo tho-.,
bo,..,
1. AI mmunostain resub lhcukl be repotted, wtllllllhlr paa.l:trrfltJ MPU\16, or nOI"'CCnb1bu'tay
*·
• Tile 1111111re ctlhe OCIOCim..., tooled, 1l'o:lofl oodlon. 110111111h ooc11on. <YIOit&l>.
• Tht p!JII'.fftfl1 bloc::k number uk'ld toobtU\ !:l!ld:bnt tlcr IMMUI"'IS&ddt'l!,.
• Tile anUIIodiM """"· ln<bllna'<ltln ·~·tilt dono ""mber
• Tile roW! ct .Uinll\ll 1«- antibody, lndudlrc colullr lcaiii!JIIon \It! on"'-
4. An ln1DIJ>- ct lilt flndlnp In the ..nlltt! ol the d...,.... IOIII! '"''"..,..to tho
.,., - !IIQ)I:tl pillloqJf!lpoot llmm"""'lilodlimli:d '"'""'""' I!JI!Orl!!d ..perldl!ty
S. EXoct ~.11111.1on recrleWI rnolll«l...nd """'Iori COIIdlbi>IIOOd 110t llo 1>1\~o!Cho
ropoot bill must bo..,.~llle In llhornorym.a...,IIINI ""'"'&
oncilJuy lall.t (.-..:h u mol ....Jar IPD "lio: - ) . Afu:r mUll&lion of poci:ljve aad
...,..U... """1ZVII, the- U..... oeaiDao"""""' l>e --d for P"""""" of d>e
aaJ, aad numbocr of immu-ea.:Ooc <>ellt or foci. In tomt;-
ate&(a) of la!ctcot, lcctl!i:otiOil of lWZniiiO!drtoch=icol tipal, lrltaulty of ol,it
limittd ....t .....n tiolnt; .amplcf, che uea of~~ mt.J -be p-i». d>e
~tor
*All e-figures are available online via the Solution Site Image Bank.
856
Chapter 56 • Immunofluorescence I 8 57
dish with a balanced salt solution while it is examined with the dissecting micro-
scope. Because glomerular diseases are a common indication for renal biopsy,
the tissue is distributed in such a way that approximately two or more glomeruli
are examined by electron microscopy, two or more by immunofluorescence, and
10 or more by light microscopy. The tissue assigned to electron microscopy is
placed in glutaraldehyde; tissue for immunofluorescence is frozen, and tissue
for light microscopy is placed in formalin. When a biopsy needs to be trans-
ported to the laboratory from a remote site, it should be placed in a container
with transport medium such as Michel's medium. Although it has been shown
that immunofluorescence can be performed on FFPE tissue for the evaluation
of renal biopsies (Kidney Int. 2006;70:2148), FFPE tissue is not routinely used
for immunofluorescence evaluation.
Light microscopic evaluation of the renal biopsy is performed using hema-
toxylin and eosin, periodic acid-Schiff (PAS), trichrome, and methenamine
silver-stained sections. The PAS and silver stains facilitate examination of the
basement membranes, and the trichrome stain highlights areas of interstitial
fibrosis. Evaluation with fluorescein isothiocyanate (FITC)--conjugated antibod-
ies against IgG, IgM, IgA, C'3, C1 q, fibrinogen, albumin, K. and }.. are performed
by direct immunofluorescence. Transplant kidney biopsies are also stained for
C4d using an indirect immunofluorescence technique. C4d is used to assess for
humoral rejection, usually seen as immunopositivity of the peritubular capillar-
ies. Addition of a 3% Evans Blue counterstain (Health Scientific, Saint Louis,
MO) is helpful to decrease the FITC background and to enhance visualization
of the tissue (Am] Transplant. 2009;9:812) (e-Fig. 56.3).
C. Lung. In the event of suspected humoral rejection after pulmonary transplan-
tation, biopsy of pulmonary parenchymal tissue acquired via transbronchial
biopsy can be submitted in Michel's medium for evaluation of complement
(C4d) by direct immunofluorescence. It is worth noting that evaluation for the
presence of C4d can also be accomplished using an immunoperoxidase method
on formalin-fixed tissue.
Ill. INDIRECT IMMUNOFLUORESCENCE. Blood (5 to 10 mL) drawn into a tube without
anticoagulant is required for all indirect immunofluorescence studies. The serum
is removed and is applied to an epithelial substrate. The substrate varies with the
clinical diagnosis, and the clinical diagnosis should guide the decision to pursue the
appropriate indirect immunofluorescence study. For indirect immunofluorescence,
serial dilutions (1:10 to 1:1280) of serum are inoculated onto the tissue substrate
together with fluorescein-labeled anti-lgG.
A. Pemphigus vulgaris. The primary utility for indirect immunofluorescence is for
the diagnosis of pemphigus vulgaris and to follow response to therapy. Serial
dilutions are performed and the end point of positivity of intercellular IgG is
reported (e-Fig. 56.2). Commercially prepared slides using guinea pig or mon-
key esophagus are used. As in other serologic tests, it is possible to get a pro-
zone effect in patients with pemphigus vulgaris, so additional dilutions may be
required to avoid a false-negative result.
B. Paraneoplastic pemphigus. The substrate for evaluation of paraneoplastic pem-
phigus is murine/rat bladder epithelium. Because this test is not commonly
ordered, it is usually only performed at reference laboratories.
C. Bullous and/or cicatricial pemphigoid. Circulating antibodies that produce
a linear basement membrane zone positivity (e-Fig. 56.1) are detected in
fewer than half of the patients with documented pemphigoid, making ancil-
lary testing by indirect immunofluorescence minimally useful for this disease
process.
D. Dermatitis herpetiformis. Circulating antibodies are not detectable in the serum of
patients with dermatitis herpetiformis; therefore, indirect immunofluorescence
is not indicated.
858 I SECTION XIII: ANCILLARY METHODS
• All e-figures are available online via the Solution Site Image Bank.
859
880 I SECTION XIII: ANCILLARY METHODS
Electronic System
..---
1
I Analog to Digital
1 Converter
I : I
I LG).,.---'> ~
I t
Orange detector
I········~ -<·······~
E
Q)
......
en
I f
Red detector Flow System
l_~ ..-~
>.
rJ)
cu
f
(.)
:;::;
a.
0 Green detector
~
Slide angle detector
Q Laser
Figure 57.1 The How cytometer is composed of a How system, optical system, and electronic
system. The flow system transports cells in a stream to the laser beam for analysis. The optical
system consists of lasers to illuminate the cells in the sample stream and optical filters to direct
the resulting light signals to the appropriate dete<:tors. The electronic system converts light signals
into electronic signals that are processed by the computer.
DNA Histogram
G1
DNA Content
of a cell will differentiate normal cells from aneuploid malignant cells. The
histograms from malignant tumors will show abnormal peaks corresponding to
more (hyperdiploid) or less (hypodiploid) DNA than nonnal cells.
C. Cell cycle analysis. The cell cycle is composed of four phases. Cells in Gl are
recovering from division or are preparing for division; cells in S phase are in
the process of synthesizing new DNA. Cells in G2 phase have finished DNA
synthesis and therefore have double the normal amount of DNA (tetraploid).
M phase encompasses division into two similar daughter cells. (Note that cells
in GO phase are not cycling at all). The cell cycle can be plotted as a histogram
with the number of cells per channel on the y-axis and the fluorescence intensity
of cells stained for DNA content on the x-axis (Fig. 57.2).
SUGGESTED READINGS
Keren DFt McCoy 1Pt Carey JLt eds. Plow Cytometry in Clinical Diagnosis. 3rd ed. Chicago: ASCP
Press; 2001:31-65.
Givan AL. Flow Cytometry, First Principles. New York: Wtley-Liss; 1.9.92:75-102.
Ormerod MG, ed. Flow Cytometry. 3rd ed. Oxford: Oxford University Press; 2005:23-33.
Cytogenetics
Shashikant Kulkarni, Hussam AI-Kateb,
and Catherine Cottrell
draw blood-
mix with
anticoagulant
Blood in culture
medium with mitogen Add colcemid to
for 72 hrs at 37"C arrest cell division
at metaphase
Figure 58.1 Overall scheme for the production of metaphase chromosomes for traditional cyto~
genetic analysis.
1. Culture initiation. Different specimen types have different sample and han~
dling requirements (Table 58.1). Inappropriate handling, as well as delay
between specimen collection and culture initiation, can markedly decrease
the likelihood that the sample will grow in vitro, so communication and
coordination with the cytogenetics laboratory are essential.
In vitro culture relies on a sterile microenvironment, and so specimens
should be collected under sterile conditions. In practice, sterility is most
difficult to achieve when sampling solid tissues; in this setting, clean instru~
ments and a clean cutting surface, together with transport of the specimen
in medium supplemented with broad~spectrum antibiotics, can be used to
minimize contamination.
Bone marrow and solid tissue neoplasms consist of cell types that pro-
liferate spontaneously in culture, although often at a low rate. Lymph
nodes are composed of cells that have a low intrinsic proliferative rate
but that can be induced to divide much more rapidly by the addi-
tion of mitogens. Phytohemagglutinin (PHA) stimulates proliferation of
T-lymphocytes. Lipopolysaccharide (LPS), protein A, 12-0-tetradecanoly-
phorbol-13-acetate (TPA), Epstein-Barr virus, synthetic oligonucleotides,
and pokeweed mitogen induce proliferation of B-lymphocytes, and are also
required for successful culture of some leukemias and lymphomas of B-cell
origin.
2. Culture maintenance. The length of in vitro culture depends on cell type.
Since bone marrow cultures contain spontaneously proliferating cells, they
can be harvested after only a 24- to 48-hour culture interval, if not
directly after specimen collection. Peripheral blood cultures usually require a
72-hour culture interval. The growth rate of solid tissue specimens is difficult
to predict; some solid tumors require culture periods of 2 weeks or longer.
3. Cell harvest. Colcemid, a synthetic analogue of colchicine (an alkaloid from
the bulb of the Mediterranean plant Colchicum), prevents separation of
sister chromatids and is used to block the proliferating cells in metaphase,
thus allowing an accumulation of cells at metaphase stage. A hypotonic
solution is then used to swell the cells so that, after fixation, the chromosomes
are adequately spread for microscopic analysis.
Since cells in culture do not proceed through the cell cycle in synchrony,
chemical synchronization of cell division is often required to obtain an
acceptable mitotic index. A common chemical approach involves addition
of excess thymidine, which stalls cells at the S-phase of the cell cycle by
decreasing the amount of dCTP available for DNA synthesis. When the
excess thymidine is removed (or the effect of excess thymidine is elimi-
nated by the addition of deoxycytidine), normal DNA replication resumes,
and the collective release of the cells from S-phase produces a transiently
high mitotic index. Alternatively, 5-fluorodeoxyuridine (which inhibits the
enzyme thymidylate synthetase) can be used to stall cells at the G liS bound-
ary; in this method, addition of thymidine releases the block.
4. Banding. The different techniques that can be used to stain metaphase chro-
mosomes can be divided into two general categories: methods that produce
specific alternating white and dark regions (bands) along the length of each
chromosome and methods that stain only a defined region of specific chro-
mosomes (Table 58.2). In general, the dark bands are gene-poor AT-rich
regions, whereas the light bands are composed of gene-rich GC-rich regions.
The quality of staining depends on several technical factors, including suffi-
cient separation of the chromosomes in the metaphase spread to allow clear
visualization. Although there are no internationally accepted standards for
banding resolution, ideograms are used as reference points (e-Fig. 58.1).*
Many countries, including the United States, Canada, UK, France, Japan,
and Australia have established standards that specify the minimum require-
ments for the number and quality of cells that must be processed for chro-
mosome analysis depending on sample type, although many cases require
even more detailed analysis.
5. Microscopic analysis. The method used to stain the chromosomes dictates
whether bright-field microscopy or fluorescence microscopy is used to visu-
alize the chromosomes. Conventional photography has traditionally been
used to produce high-resolution prints of the stained chromosomes, but
*All e-figures are available online via the Solution Site Image Bank.
••• I SECTION X.lllt ANCILLARY t.IETHOD5
-
TlciiiiiUMta.ltllf'I.IOIIII'tc-...-o....- - -
Gicmlolllndlrc(G-b<ndl\l)
_,,,_,
Darl;-an>JU riot>; IWrtbendon
CO!t:l!
Ql/hoemo t>ol'odiiJJ: BiWI! l''l!»l'oU"'JU rk:ll
CQ-Midln&)
_ , . hond~(R·-nal AT<"Ich ....nailllll'o illm!l' (hll't dill
nuo-), CC1.!t:l! repna
-~ dlllily"""" bflllll
nuo-
4,6-0ioml:l!IIO-:Z.~
(l)APO mt~lrc
-··•taatlllt>•-·11·
Oon-IMIIaA><IIn>mllt*l
--.. (C.-t>JI)
...... DAI't btl<i>IO'·It:l! llll.lotrs; Died~•
- .tnltutb Q.-ng
II. -
e!Jo:t<oai< i""'!Pag ..,....,. ha-... !lOW n:plaoed ..,......,,;.,.) ~
(.,.V. . $8.2.).
tbc bui$ of . . l~ti01!, ll1<l b41eding p.t!W'a
QCni!OJXl(:re
D. ~fill ln. M""f ai the c:omma.a c:a~~~t~ <>I Nihue to obt.&in a qtogeo<tX;
.....Jt (Tlohlo: 58.3) ""' he .met..~ by carefW ,.w.;,., of Yiahle- .mh
p:rom,p< liP,.;""" """"l/'lt to the c:ytagro•ctiaa labcmamry ill the approp:lim
mcd!•nn Noorotllcl..., .....ro1 .,..... of - r fcllutc ..,.. lal!.t:t=.t to lD. ftlro
C1l!tatc lJ1<I CIIIIIOI he cltm!neted by""""- theiii<>Ot III.Cdcolcua labotato:ty led!.·
cUqiJC.
0.0...-
O..~l>y--~ ...1&
l>yo ftOI'oJ'IIIIWtii!Mc:!ont of!J.mor-
P,Ut:tobiiJOI't...-
Poll <UIIIIIa Ill~"'
T6CIInlcoletro10 lll\IOMill collie~ Gll:lt>l!deDtllotlon, orRtlllll
Mlod.,.. CAn ob!lo!TniiiY lo-.oliad. .- lllebno!Troolltl h ln0011'0<1tJ l'tterl!lotedl
Chapter 58 • Cytogenetics I 8 67
For both multiplex FISH and SKY, a cocktail consisting of labeled probes for
each of the 24 chromosomes is hybridized to metaphase chromosome spreads,
and the fluorescent emissions are measured by computerized imaging systems.
Specialized software is used to determine the combination of fluorophores present
along the length of each chromosome, which makes it possible to assemble a
karyotype.
A. Advantages. Multiplex FISH and SKY are used to detect aneuploidy, detect
interchromosomal rearrangements, and identify marker chromosomes (extra-
chromsomal material of unknown origin). In many cases, multiplex-FISH or
SKY make it possible to establish the chromosomal origin of rearrangements
that cannot be defined on the basis of routine cytogenetic analysis. A web-
based database has been developed to facilitate identification of chromosomal
aberrations detected by multiplex FISH (http://www.ncbi.nlm.nih.gov/sky/), a
database that contains links to other websites that can be used to integrate the
cytogenetic map with physical and sequence maps.
B. Disadvantages. The lower limit of the size of individual DNA chromosomal frag-
ments that can be visualized by either technique is in the range of 1 to 2Mb,
although neither technique provides direct information on the involved chro-
mosomal bands. Similarly, multiplex FISH and SKY will only reveal intrachro-
mosomal deletions and duplications that are large enough to result in a change
in size of the affected chromosome; neither technique is designed to detect intra-
chromosomal rearrangements such as inversions, and neither is informative in
regions with repetitive DNA.
C. Modifications of multiplex FISH and SKY. Mixtures of so-called partial chromo-
some paints, each of which hybridizes to only a band or subband of an indi-
vidual chromosome, can be used to produce a pseudocolor-banded karyotype
at a resolution of about 550 bands (Cytogenet Cell Genet. 84:156, 1999). The
use of partial chromosome paints makes it possible to employ multiplex FISH
and SKY methodology to identify translocation breakpoints and to detect inter-
chromosomal rearrangements.
V. COMPARATIVE GENOMIC HYBRIDIZATION (CGH). While CGH often has a higher sen-
sitivity than conventional cytogenetic analysis, of even greater significance is the
fact that CGH can be performed using DNA extracted from fixed as well as fresh
tumor samples. The technique therefore makes it possible to perform a genome-
wide scan for structural alterations even on those cases for which conventional
cytogenetic analysis is not feasible or is unsuccessful. CGH essentially opens the
entire formalin-fixed tissue archive to at least limited cytogenetic analysis.
For a typical CGH test, genomic DNA from a tumor sample is labeled with a
red fluorophore, and genomic DNA from a paired normal tissue sample is labeled
with a green fluorophore. The green and red probes are mixed and used in a single
hybridization.
A. Metaphase CGH. This technique is basically a variation of metaphase FISH used
to survey the entire genome for chromosomal deletions and amplifications (Sci-
ence. 1992;258:818; Trends Genet. 1997;13:405). The labeled probe mixture
is used in a hybridization to metaphase chromosomes prepared from normal
cells, and the ratio of the green to red fluorescent signals is measured along the
length of each chromosome. Areas where the ratio deviates significantly from
the expected one-to-one relationship indicate a change in DNA copy number in
the tumor; areas where the red to green ratio is significantly> 1 are areas of chro-
mosomal gain (usually amplifications), and areas where the red to green ratio is
significantly <1 are areas of chromosomal loss (deletions). The smallest chro-
mosomal alterations that can be reproducibly detected are about 3Mb long.
B. Array CGH (aCGH). This approach utilizes a microarray consisting of an ordered
arrangement of DNA molecules (features) linked to a solid matrix support. The
labeled probe mixture is hybridized to the micro array, and the ratio of the green
870 I SECTION XIII: ANCILLARY METHODS
000000
000000
~··,<;,~<W &fl:ffi],ruJ
Figure 58.2 Array CGH methodology. Top: Method. A patienfs DNA is labeled
with a red dye and a control genomic DNA preparation is labeled with green
dye. The DNA preparations are mixed and co hybridized to an array of BACs or
oligonucleotides on a glass slide. The DNA bound to each spot (known as a fea-
ture) of the array is quantified using a laser scanner. In the patient's DNA, normal
regions will be indicated by a yellow balanced color; regions of duplication will be
identified as red, and regions of deletion will be identified as green. Bottom: Data
presentation. The data from each feature of the array (represented by circles) are
plotted in relation to the features' positions along the chromosome and a balanced
copy number (horizontal line). In this illustratiofit a duster of adjacent features
that falls significandy below a balanced copy number result (oval) indicates the
presence of a deletion. The resolution of array CGH is in theory limited only
by the number of features in the array; commercially available arrays currently
provide a resolution of <10 kb. (Adapted from Beaudet et al. Annu Rev Med.
2008;59:113.)
to red fluorescent signals is measured for each feature (Fig. 58.2). Because each
DNA feature has been mapped to a specific region of the genome, the ratio of
the green to red fluorescent signal for each feature provides information on the
gain or loss of the corresponding chromosomal region. The resolution of aCGH
is in theory limited only by the number of features in the array; commercially
available arrays currently provide a resolution of< 10 kb. Genomic microarrays
(aCGH and related microarray-based methods) are currently clinically applied
to detect genomic copy number changes as well as copy neutral changes (uni-
parental disomy [UPD]Iloss of heterozygosity [LOH]).
VI. MICROARRAY ANALYSIS. Since the advent of the use of genomic microarrays in the
clinical laboratory, the technology has rapidly become the standard of care to eval-
uate patients for genomic imbalance, especially in diagnostic testing for patients
Arrq ott111Ml1eo snp. nuc- 01~ .,.,.
I>O!IIn«<>hlorn llll6'0
Numbof ~ mlllon 1,000,000 Mo!Wo <ll)O,OOI) Ma!Wo
DNA lnPIII~&r~Jinlrrwt 200-500 ftll 1-S!'«
P-IYPM SNP •nd olio p,_ OII.Jp probes
--·UlH
Umb~,..,. < 10.201111 Aalaw•lG-201<11
Th-fof-nof Mlybaulawu!>'l(, 20.'10\1:.
AbltY10-<WJ
AbltY.,_. r...
unlpor•nbl<loorlly
AbltY!o- y.,.
c:onwl,lllnJy
Mechod of -mom d l'tobo .,olllrtallotilo Pll1iflllt-Piod~eclti
ldll!wd ~ 111&1101i<frt
"'i!t numbof mbolan"'
..mplo) ""'""'rod.,
an In ~-·~~
contrd ONAlod-
llllmrof.-.,.,..,_, -pinlllld-
No rw
872 I SECTION XIII: ANCILLARY METHODS
Affymetrix I!lumina
a. Genomic DNA (250 ng) a. Genomic DNA (750 ng)
Nspl N
_,.......___
b. Digestion b. Amplification
~
~ ~
~
~
d. Precipitation, resuspension ,
hybridization
~ A
J ------- ~.
~~- go
Figure 58.3 SNP array procedures. In the Affymetri:x platform (Affymetrix; Santa Clara, CA),
genomic DNA is digesred with the Nspl restriction enzyme and the resulred DNA fragments
are ligated to adaptors and subsequently amplified; the amplification products are fragmented,
end-labeled, and hybridized to the array. In the Illumina platform (IDumina, Inc., San Diego,
CA), the entire genome is amplified and then hybridized to a bead array; allelic discrimination is
achieved by a single base extension reaction. In both platforms, the probe inrensity is measured and
compared with an in silico reference to evaluate DNA copy number. (Adapred from Schoumans
and Ruivenkamp. Methods Mol Bioi. 2010;628:53.)
MlnNI!aa •s,.t~al Dlla!Jifan
odd Addlllonal mo11!!1ol of 111la'Jowtlo"'*'
~reb!IICialtoD Nlll1botof .... lh..cil<lorlo
OM\ Cer'l'trttnler&
"-""ion(,) Brak
OO.blo- ~ ,) Brakandrounlon
o;mma (.) SepiiRI:l~anumbot'. M:l~hromotetl"'Ilend
abno!Tnon.o
del Oaf~
drJt OerNt!Midl-
dm., Ooublo m!MIOO
dup lluplation
I ~h~~
ld01n lclei1looloblocnuollllouo., ~dono
lnv ..........,
fft& INMtlon
Mdrbf dUM'diOI"''llll
"'"
mhn sip\(-) Loa
m..,~..., h<l MLl!lplo Clllllot, abo dool,lnoloo GIRt numbon•IIIISH
pW...,(+) GU1
a-molt(?) u,_.,~ d.~...... ldell1lb1lon orabllonnol!y
r R)t•m......,.,.
"'P Radpnm
sluh(l) S.rrletcoll-orclonD
--·~) S.rrletdlronmomosandb~Nlijlolrbln
lnwhlfna mao then CI'IO ~tu·omo.IIOO"lG
""""'_,-
t 11anolotallon
. . .N .........
a tt .,-.,.::tn•
C1 Crtlliiilllll.:r•a,J•t. . .illll•
45.X Tumor aynd""""
47:1X'f I() hal'- "'"d"""•
MINIINI
47,XV,+21
clll•- -pi.WIN ,.,.. _ llto>my 21 (ll<Nm ~l"tlnO)
48,XY,+21e,+21!20l
bf-
tl\& lore liM Cq) of dlr0ft..,._,6 22 .8t
rop,n I, bond 2. Th6ftlltnt.mt.ln
dlsf&n- til& numbor of.,...
w«<lll>o olllaMid abf>ol•nolll!ot and til&
lilt 111m bot~ 11>o- nil!lbof of
t \1 .,.....,_NH-
47,l(Y,-r.bh dorQ)(~)
<Oih111ot ....... liotJiod.
*All e-figures are available online via the Solution Site Image Bank.
876
c~t~pt~r sg • Fluee~ it~Sib.l Hybritri:trtiet~ I .,,
,,(,I ]lj.f!\1 EamplluiDIIIIJ!MikTutlbff'IAI
,_IIIII_..
Trisomy 13. 18, 21
xr__,..,.
.!Mil..........
~ ...a. (Sj>)
Pr-·-~ (l!iq)
01 c.o,..
synd- (22q)
flwlllll'll ......
xr FISti on 061 rrl,.,_,bw1r!lllloetd:d1Kid ...., ~n> IJido lit
-nolo""' I.OinJl kn_,&,.,flii,,.IILI_,. In !>Ifmary bJmor
011....,
Ql_nol (11..-..
__ ... .. -~~--
-~~~~~ -bn)
G_u,_,.
-··
00110 11111DIIIcdono
-~~.
••..-cn..
<a.e,
u~no)
Boctj Fluid&
lnt~-~~
Cell cuture r-·..-Jtk)na
hllllll• -1101111'11 ............
-·-lnod-
l!lln ...:dollo!U J,llt\l
D..._,"""' nuelol
to the traditional whole slide approaches (Adv Anat Pathol. 2001;8:14), and
although complications due to tumor heterogeneity can be problematic, ade-
quate sampling can be optimized by incorporating multiple cores from each
specimen. TMA-FISH is an excellent method for new probe validation, profi-
ciency testing, interlaboratory comparisons, and quality assurance/quality con-
trol(] Histochem Cytochem. 2004;52:501).
D. Disadvantages and pitfalls of FISH. Although recent technical advances have
greatly enhanced the clinical applicability of FISH, a number of limitations
remain. Signal fading is one of the main disadvantages. Clinical labs typically
circumvent this pitfall by capturing digital images as a permanent record of
each case; a permanent record is not otherwise possible unless chromogenic
detection (CISH) is used. Unfortunately, multicolor CISH is not as simple as
multicolor FISH; currently available chromogens lack the spectral versatility,
sensitivity, and spatial resolution attainable with fluorochromes. Some com-
mercial CISH applications bypass this problem by providing the test and ref-
erence probes separately, so that in place of one dual-color FISH assay, two
single-color CISH experiments are performed. Recently developed photostable
quantum dots offer a potential alternative for permanent fluorescent signals
(] Histochem Cytochem. 2003;51:981). Other limitations include a variety of
artifacts, particularly common in paraffin sections, that make correct interpre-
tation of FISH results dependent on significant experience.
1. Truncation artifact. This artifact is due to the underestimation of copy number
because of an incomplete DNA complement within transected nuclei, and it
is therefore important to assess controls cut at the same thickness.
2. Aneuploidy and polyploidy. Artifacts due to aneuploidy and polyploidy can
result in confusing signal counts and are a particularly common finding in
malignant and even some benign neoplasms. Although the simplest approach
is to interpret absolute losses ( <2 copies) and gains (>2 copies), "relative"
losses and gains can also be delineated on the basis of a reference ploidy,
obtained either by flow cytometry or the assessment of multiple chromosomes
by FISH. For example, cells with four chromosomes, nine centromeres, and
two copies of the p16 gene region on 9p21 would be interpreted as having
polysomy 9 and a hemizygous p16 deletion (e-Fig. 59.7A); a similar tumor
with no p 16 signals would be interpreted as polysomy 9 with homozygous
p16 deletion (e-Fig. 59.7B).
3. Autofluorescence. This is a particularly common problem in FFPE tissue sec-
tions. Although autofluorescent tissue fragments are usually larger and more
irregular than true signals, some fragments have just the right size to mimic
true nuclear signals. The use of multiple filters is helpful, since autofluores-
cence will often appear at several wavelengths of light, whereas true signals
only fluoresce at one wavelength.
4. Partial hybridization failure. This issue is most problematic when combining a
highly robust probe (e.g., centromere) with a comparatively weak probe (e.g.,
small locus specific probe). This artifact can be minimized by counting only
in regions where the majority of cells have discernible signals. Signals from
both probes should be seen in normal cells (e.g., endothelial cells) within the
region for the counts to be considered reliable.
E. Additional technical considerations. Many different FISH protocols are available;
they vary depending on individual preferences and specimen type. Simple pro-
tocols are generally better, requiring less "hands on" time, fewer opportunities
for error, and fewer troubleshooting requirements. Automated instruments are
now available to minimize hands on time, though they are expensive. In general,
the basic steps of the protocols are similar to those of immunohistochemistry
and include deparaffinization, pretreatment/target retrieval, probe and target
Chapter 59 • Fluorescence in Situ Hybridization I 879
as part of the human genome project have made it possible to rapidly iden-
tify vectors that contain sequences of interest, gene names, or physical maps
of individual chromosomes (http://www.genome.ucsc.edu). Similarly, mapped
BAC clones spread throughout the human genome at 1-Mb intervals have
also become available (http://mp.invitrogen.com). However, regardless of how
a probe is obtained, it is important to verify its identity, either by screen-
ing for the DNA sequence of interest by PCR or by performing metaphase
FISH to determine that the probe localizes to the appropriate cytogenetic band
(e-Fig. 59.8).
Ill. CLINICAL APPLICATIONS. FISH testing is clinically useful when a cytogenetic alter-
ation (deletion, gain, amplification, and translocation) is sensitive and specific for
a single tumor type, either as a diagnostic biomarker (as in many hematopoi-
etic malignancies) or as a prognostic biomarker helping to predict which tumors
will be aggressive or indolent (HER-2/neu amplification testing in breast cancer).
Some translocation and deletions detected by FISH are also helpful in predicting
response to a specific therapy; examples include detection of t(11;18) in a sub-
set of mucosa-associated lymphoid tissue (MALT) lymphomas (which tend to be
resistant to conventional therapy), detection of FIP1L1-PDGFRA fusion formed
as a cryptic deletion at 4q12 in chronic eosinophilic leukemia which is sensitive
to imatinib therapy, and detection of del(13q) and/or t(4;14) in a subset of cases
of multiple myeloma (which tend to have a worse prognosis). The most common
clinical applications of FISH testing currently include HER-2/neu amplification
testing for breast cancer, UroVysion testing in urine cytology specimens, lp/19q
deletion testing in gliomas, and testing for signature translocations associated with
specific hematologic, soft tissue, and/or pediatric malignancies. Clinically relevant
examples for each alteration type detectable by FISH are summarized in Table 59.3.
A. Aneusomies and deletions. Aneusomies represent gains and losses of whole chro-
mosomes. Deletions are losses of distinct chromosomal regions, varying in size
from loss of a specific gene or portion of a gene to an entire chromosomal arm.
Aneusomies and deletions are amongst the most common alterations detected in
neoplasms by FISH (Table 59.3), although it is sometimes difficult to distinguish
specific tumor-associated polysomies and monosomies from nonspecific gains
and losses that are secondary changes due to the genomic instability that is char-
acteristic of many malignancies. The use of reference probes helps to distinguish
such chromosomal gains or high-level polysomies from true gene amplification
(e-Fig. 59.3).
One of the most common FISH applications is testing for deletions of 1 p
and 19q in diffuse gliomas. The presence of lp and 19q codeletion (typically,
loss of the entire arm of each) has diagnostic, prognostic, and predictive value in
that this genetic signature (e-Fig. 59.2A and B) is associated predominantly with
pure oligodendrogliomas with enhanced therapeutic responsiveness and overall
survival time(/ Neuropathol Exp Neurol. 2003;62:111). There is no current
consensus for the optimal way to enumerate signals for chromosomal losses and
gains, though in clinical cases a common approach is to use two individuals who
count signals in 100 cells each; when the counts are concordant, they are simply
added for a total enumeration of 200 cells, but when there is a discrepancy or the
counts are borderline for an alteration, then either the same individuals count
additional cells or a third enumerator is utilized. Despite the clinical utility of 1 p
and 19q testing, the precise gene targets on these chromosomes remain unclear.
In other tumor types, a specific tumor suppressor is known to be targeted
by various chromosomal deletions (Table 59.3). Notable examples include the
INI1/hSNF5 gene on 22q11.2 in malignant rhabdoid tumors and atypical ter-
atoid/rhabdoid tumors, the NF2 gene at 22q12 in meningiomas, the RB1 gene
at 13q14, the TP 53 gene at 17p13.1 in multiple myeloma, and the NF1 gene at
17q11.2 in malignant peripheral nerve sheath tumors.
..,
__ .........,...,,. -- I
Cltlptlr sg • Fluet~ il'l Sib.l Hybritli:trtiet~
....__
.,,.
.., ....,.,,
''I.I IIJUII ~tid Mlratlofu ComlftOfliJ DeleeiM bf' Alit
Ollpierdr<'-'loN 1~- w!thllllt- Dlo~~
plOd-
~llol Cord- +3, 7, or 17:91>- Dll.,-
Lu~Con:lnoma +7!>.- S~>.or6 Dll.,-
cu.
..._,
13a-.11Q-.17Q- Dlo-~
OBN +7,-10 Dlo!.)IOOIIe
__.
PVoetatt tet'Ch:l'l\a 111>-.lkt+
171>-. I~; (117q)
p~
DIIJl'O'!IO
l.eUolmiMIM!ll; +B. +12; --S, -7 Dlo-~
Dll.,-
_..,...
MRT• .m!!T !NIIA>SNRH 224-l
Dlo.,_
""lli>lo II...,.. IIFZWQ-1
RBl U3a-l, IJI53D7~) p~
...........
-
MPKST llfl D7Q-) Dlo!.)IOOIIe
8tti~MtOIJtiM"nl HE/I;- p...-_p!Od-
GBN
IHr1j<
EGFII Dlo-~
Dll.,-
1111:4Aicl>ll-• NYCN, MlJC
Dlo-~
~-£'I'IIS.8A
~CIII'CII'IQTIO P~O<'<Id-
• ,,, 1 ,, EWSJI'tjEf £W$.FIJJ, Dlo!.)IOOIIe
~llaln:.:tnl SYT.sg; SVT-BA Dlo!.)IOOIIe
AMolarRMS f'KIG.F1IHk, FXHR·BA DIIJl'O'!IO
0!1\CT EWS-WTl.~ Dll.,-
MIRC ll_ll,>n'll CHOP.SA Dll.,-
Dlo.,_
Claorc:elltai"'I))I'nll ~1n.,f.WS-8A
IMT AUHPIU AlJHI'IIH. Dlo-~
AUC-<:ARS, AlX-BA
Bultlil ~llano N~M- DIIJl'O'!IO
MAlT tlmlil.,., AP1241AJIJ, JGH.MALTI, DIIJl'O'!IO
IIWJ'l·BA
Folllwla- tlml>ll<><"'
AlA
/6H-Ba2
ALK·BA
Dlo-
Dlo-~
~ted-
Mlr'ol!o . . ljmJWnl H:H-«NNJ1 DIIJl'O'!IO
""Iii>",.,..,.... H:H-«NNJJ, ~ P..,-
IGH~
CML BCIMBl DII..-,WRD,
plOd-
AML Allll-atl, CSFB-BA, Dlo~~
A\11.-RAR!I, AAAA-BA, ~ted-
IIIJ.-<lA, ~..A8l
AU. n:t..Wll. ~..AB~ MU..BA
Dlo-~
p!Odlc!Nt
BCL...-w!th l6lf-Ba2: llfYC-<Git 111'()-BA Dlo-~
- ... HoriTIOdlill8
bet _. OLBCL•nd
- tlmli'""" PDt;FTIA, PtiiOFII8, l"'ifffl;
Mjolklhr>d ljmJidl
._~o~m ... RPJU-I'OOF/IA Dlo-~
p!Odlc!Nt
...lOIII,II\1
882 I SECTION XIII: ANCILLARY METHODS
~·d-
Gon . .mpllllcltson
TrtnJiocdon
.... * I ... - - DIIA problt
Oommlt<lal
"""""' l!podlle !>4!. BAC doneol
-~ltllfOIIrollao•
IJiilp1oollc aid
P~•ld
Prodk* reo-..-laclt ol-oeto I>OIIent11101al1!'
...... ' . , eptlta 'fF ,bzn .-.rt1»111tn ~lillllllllli)llefl'll ......
Arc:IINIJ p.ondll.,..mbtdllod U.U. ,.,... f>'olllllrcz!ln , . _ CJI'*'I:I
~-·~
Mo<pldop:ollf"""""I>Pna!Lmora- to-~
nne.-.
d ...... ~."""""""'· <ll'ldlw !>l1ilftt doatll
f'lllont..,.orotilerdei!IOITAI>IblttreloWnt~,.._.
- o f .....,.lonlllu,...l rna~_,.
-ryp. at .o4u,.nl thnpy a.dm-.bW:od
---to-otu<~tq-1
SIIUIUc:ol _ , 1nol)oll.: numbar of opoctnano n - for _l..,lt\l:1i,...
I' (.W Jtl11 !llmplu of s,.bok and Alllnolollo"" u..tl• lolo/pi'IIA rrsH
' - Nom .......
QlnNII. . . .,.., l*a!JII•
Phatlrln(+) PIMOnlonupocllc<lln>mosoM6
Ml'olu\111(-l Abtonlonupodllc:ctu..,_o
of+ Duplcationona.,.lllcctuomoiOOno
X """''*"of
PI1ICIIdeo oi&Mio _,
S&tnloalon ~) Sec>a- p-on dll'lll'<IK doi-O~IOMOOtmOt
f'<rbl (J Sec>a- ~node rMUb 1\'rm isll ......
amp Ampl~od II&NI
em ComecllriO< ad)lalnt sl,inlll
btl Wllan usod l'ifba~. rafm to hybfld-n li>ChiQ!lOIOU ...
nuc bill Nudoar or .,.,rphooolluau h'ltlldlzii1Soll
1><0 P•r1111 clr-o pellll
oop Sec>al'llll!d 1\!J101a (o!lk:tlm uaualy adjlolnt In ""'""II~
~*I ~~~
wcp Wllalo ·~-pOrt
II& I SECTION X.lllt ANCILLARY t.IETHOD5
llltjl,_.fda,tOV llllldk:al-21CFRIIOII.'!O
In Vlln> DloJp!OIIIc """*'<:<s lor
Hum1111ha
• Ana~ '-lito <ASII4)
llltj!J-.omp.<>rg """""lor l'olllolofl.¥ P -
- - ~ lll'•odal,
'"""""'~·
dbouo, lnf-.s
.,1111 bunorol
CHAMP 1-MIIorAMP mombot>
Teotdln>:!Oitoo
• Solid tumoro
• H..-pot~~.qy
• fftfed:loutdi!MM
Chapter 60 • Direct and Indirect Methods for DNA Sequence Analysis I 889
o.- TP
"""""M''- lP + f'H
TH
O\qlaolle~=FP+TH
- . ,.. . , _ ... _lP~f'P
.....,...
IMI
All~nlool
DIIIJI-
Opotl!l:nl
Dlqnoltic•IOII>t:Y, d\q!lmtk"'*'ll'd:y, Y-1-
T..,knl:ll...,llit.ty, mdmlool-ll'd:y, p-lan,~. pl<llldency
5
3
1
I
r-- target
region --;
II I 11111111111111111_:11 111111111 'I' Ill 1111111 II II 5 3 1
5' 3'
31 ======================= 51
Step 2 .J Cool to allow primer annealing
51== == 31
Primer 1
3 ·.@.!!!!!.s•
Primer 2
31 5 ',.,.. 3' 51
51 === 31
3 • , ~ nm q@p n g u gqJrp n np m 5 .
31 5' iiiQJblbiiOJIIIIII]ijiliUiillli Wi 3'
=== 51
' End of 1st cycle
3 I 5 j !IQJ!)II!illilllillJMIIIIII)III]Illllllill!IIIUIIII!Dilillldll35 I
r lll iiliiillllilllllllllllllllllllllllllllllllllll
,
~ : greater than 10 copies of
target region
7
Figure 60.1 Schematic diagram of PCR. Each cycle consists of three sreps: The reaction mix
is heated to denature the double-stranded DNA template, the reaction mix is cooled to permit
annealing of oligonucleotide primers to sequences that flank the targetregioD.t and then the reaction
mix is warmed to permit the heat~stable polymerase to synthesize new DNA strands. Each newly
synthesized DNA strand then acts as a template in subsequent three~step cycles of denaturation,
annealing, and DNA synthesis, producing exponential amplification of the target region.
b. PCR has hi&h sensitivity and specificity. When optimized, PCR can detect
one abnormal cell in a background of 105 normal cells, and can even be
used to analyze single copy genes from individual cells (Methods Enzymol.
2002;356:295, 334). PCR can also be used to detect a broad range of
generic abnormalities ranging from gross structural alterations such as
translocations to single base-pair changes.
892 I SECTION XIII: ANCILLARY METHODS
c. PCR products are easily labeled for detection. For primer-mediated labeling,
a labeled chemical group (usually a fluorophore) is attached to the 5' end of
either or both oligonucleotide primers. Alternatively, the PCR product can
be directly labeled by including one or more labeled nucleotide precursors
into the PCR mix.
d. Phenotype-genotype correlations are possible. When performed on tissue
sections, PCR provides only an indirect correlation of morphology with
underlying genetic abnormalities. Microdissection, in which the region of
interest is carved out of the FFPE tissue block, scraped from tissue sections
or cytology slides, or collected more precisely with a micromanipulator
apparatus, provides some enrichment for morphologic-genetic correla-
tions. More precise phenotypic-genotypic analysis is achieved by collect-
ing individual cells by laser capture microdissection, by flow cytometry, or
even by immunomagnetic methods. In situ PCR performed on histologic
tissue sections themselves is perhaps the ultimate method for providing
morphologic localization of genotypic expression; however, the technique
is so technically demanding that it has limited use in clinical laboratories.
2. Limitations of PCR
a. PCR only analyzes the target region. Testing only provides information on
the target segment amplified by the specific primer set employed.
b. PCR only amplifies intact target regions. Mutations that damage a primer
binding site (including insertions, deletions, and even point mutations)
preclude amplification of the target region by PCR and can easily lead to
errors in test interpretation. Similarly, mutations that alter the structure
of the target region in ways not accounted for during primer set design
(e.g., large insertions, deletions, inversions, or translocations) may pre-
dude amplification.
c. Amplification bias. PCR bias refers to the fact that some DNA templates
are preferentially amplified versus other templates within the same reac-
tion. PCR bias can be caused by differences in template length, random
variations in template number (especially with very low target abundance,
producing an artifact known as allele dropout), and random variations in
PCR efficiency with each cycle. Amplification bias can even result from dif-
ferences in the target sequence itself as small as a single base substitution.
PCR bias can cause over tenfold differences in amplification efficiency in
some settings, a difference that can influence quantitative PCR (Q-PCR)
test results and loss of heterozygosity analysis. PCR bias can be a partic-
ularly troublesome problem in multiplex PCR.
d. Technical factors. There are several technical factors that can lower the
sensitivity and specificity of PCR in routine clinical practice below that
obtained in optimized research settings. Nonspecific inhibitors ofPCR are
sometimes present in patient samples, including heparin and uncharacter-
ized components of CSF, urine, and sputum. With the extreme sensitivity
of PCR, strict attention to the physical organization and methodologies
of the laboratory are required to avoid cross-contamination of specimens.
However, the most important technical limitations are introduced
when fixed rather than fresh tissue specimens are used for testing due
to the degradation of DNA and mRNA that occurs prior to and dur-
ing fixation, as noted earlier. Test sensitivity and specificity are compro-
mised by degradation since it makes it necessary to amplify shorter target
sequences or employ a nested PCR approach, both of which increase the
risk of amplification of nonspecific sequences and cross-contamination
(Am] Surg Pathol. 2002;26:965).
C. Factors that affect testing on a diagnostic level. The intrinsic biologic variability
of disease has the greatest impact on the diagnostic sensitivity and specificity
a...,«). Oi'«t tftd fl\tlil'ld Ultltledt fer DNA S&qi./8/'W!i& AII•!Jti:a I I ••
"""""'
P-1nd posilooiOO<IIMI~ , _ , _
........blo.
ln\wllf'l,_, of motoc.ar nndii\P lOIII! til&
cll!!l fnlm tho<lllll:ol_,_, encloll>of
pr!l1oqy -
Mo"'QO!IIIforln......, dllooroo mot~l!olila;
n'llllrthodl.l!lr& untt~ ~lfle~rTt
opllor\&••dlar ~......,...""'
11dia111dli)'tll6m_lu_
R""'l"l dlnlr:llna ._t
tilt last u flla&Mab, lrelbnant, . - dl'*"l ou!coma
""'..,,..,,..s
-
-Mtlloli>lk:ltllolr- m<1il- 1'1 VIe addllb1 of""'
rnolocUar l>l1ldoiY r""'blo CCII fit mo<lcal
894 I SECTION XIII: ANCILLARY METHODS
a. The relative merit of the molecular testing should focus on the ability of
the test findings to improve patient care. For clinical utility, the molecular
diagnostic test must provide an improvement in the standard of patient
care by providing new or refined information with the potential for clini-
cal stratification of disease subtypes, prognostic categories, treatment reg-
imens, gene-targeted therapies, survival statistics, or disease progression.
The test results should complement the findings of established tests, such
as cytogenetics, immunohistochemistry, and cell surface marker analysis.
In the context of surgical pathology, the test results must be correlated
with the histopathologic features of the case.
b. Routine clinical use of molecular tests must consider practical aspects
of clinical prevalence (the disease should represent a significant health
problem or diagnostic dilemma), test run frequency, clinically relevant
turn-around-time, sensitivity, and specificity. Testing for diseases which
are common in many populations (e.g., cancer, microbial infections, and
genetic predispositions) and have well-defined molecular markers will be
performed in many laboratories. Molecular genetic testing for rare disor-
ders will be routinely available only at selected laboratories with specific
clinical programs or areas of institutional focus and expertise.
c. Testing must include steps to validate the result, and will include positive
and negative assay controls, definition of the details and limits of interpre-
tation of test results, and provisions for proficiency testing of the analytic
method, competency of the technologists, and interpretive expertise of the
laboratory director.
d. Results must be reported in a context that explains the molecular assay
data and integrates the findings with other pathology results to avoid
seemingly contradictory reports in comparison with other laboratory tests
that possess different levels of resolution or detection.
e. Biosafety, legal, ethical, and privacy issues must consistently be observed.
2. Discordant cases. Cases will arise in which there is a lack of concordance
between the diagnosis suggested by the molecular test results and the mor-
phologic diagnosis. The debate over the best approach to resolve the ambi-
guity presented by these cases reflects the fundament impact of molecular
genetics on the classification of disease as well as the status of morphology
as the historical standard of diagnosis by which new methods are measured.
Rather than arbitrarily assuming that genetic testing or morphology is supe-
rior in all cases, the most reasonable way to handle discordant cases is to
acknowledge the presence of the discrepancy, and then reappraise the clin-
ical data, pathological findings, and therapeutic implications of all the test
results.
For those cases in which the diagnosis suggested by morphology and
genetic testing are different, prospective clinical trials are required to assess
whether stage, prognosis, and response to treatment are more accurately pre-
dicted by the molecular test results than by the morphologic findings on which
most staging and treatment protocols are based. Epidemiologically, there is a
distinction between diagnostic testing and prognostic testing, with different
study designs required to assess the performance of tests in these different
settings(/ Clin Epidemiol. 2002;55:1178; Ann Intern Med. 2003;139:950).
V. VARIATIONS OF PCR
A. Nested PCR. In this technique, two consecutive PCRs are performed on the same
DNA sample; an initial amplification of a longer target sequence followed by a
second amplification of a shorter sequence contained within the first amplicon.
The second PCR may involve two internal primers (fully nested) or one internal
primer and one of the original primers (seminested). Nested PCR provides a
marked increase in sensitivity compared with traditional PCR, and is desirable
Chapter 60 • Direct and Indirect Methods for DNA Sequence Analysis I 895
when the target sequence is present at an extremely low copy number, such
as when the mutation is present in only a small subset of the cell population
under study, when the nucleic acids have been degraded as a result of tissue
fixation, or, for reverse transcriptase-PCR (RT-PCR) as discussed below, when
the target mRNA is expressed at an extremely low level. Since the increased
sensitivity carries an increased risk of cross-contamination, reproducible nested
PCR results require strict attention to laboratory technique, rigorous use of
controls, and confirmation of product identity.
B. RT-PCR. RT-PCR makes it possible to amplify RNA extracted from a tissue
sample; a complementary DNA (eDNA) strand is synthesized from the RNA
template using the enzyme RT, and the eDNA is then amplified by conventional
PCR. Fresh (or fresh frozen) tissue is the preferred source of RNA for RT-
PCR. RNA from fixed tissue is an acceptable substrate for testing (e-Fig. 60.1),*
even though it always suffers some degree of degradation depending on the
prefixation interval, the type of fixative, the length of fixation, and the method
used to isolate the RNA.
1. Advantages of RT-PCR. RT-PCR permits direct amplification of multiexon
sequences by eliminating the intervening introns, and thus greatly simpli-
fies mutation scanning methods. Similarly, RT-PCR makes it much simpler
to demonstrate the presence of translocations that create fusion genes by
making it possible to directly detect the fusion transcripts encoded by the
translocations (e-Fig. 60.2). RT-PCR can also be used to detect changes in
mRNA structure that result from alternative splicing, to demonstrate aber-
rant splicing due to mutations, and to evaluate the level of gene expression
through the quantitative methods discussed below.
2. Limitations of RT-PCR. RNA is a more technically demanding substrate with
less stability than DNA. Tissue samples must be processed rapidly (ideally,
within 20 minutes) to avoid mRNA degradation, especially since many muta-
tions render transcripts more susceptible to cellular mechanisms that clear
abnormal transcripts from the cell and result in unstable mRNA. A nested
PCR approach is often necessary when the target RNA is present at very low
levels, but RT-PCR carries an increased risk of contamination and amplifica-
tion of nonspecific sequences because the transfer of the first PCR product to
a separate tube for the nested PCR entails transmission of a highly amplified
DNA preparation.
C. Q-PCR. An ideal PCR would generate a perfect twofold increase in the number
of copies of the amplicon in each cycle of the reaction. In reality, inhibitors of
the reaction, accumulation of pyrophosphate molecules, decreasing polymerase
activity, and reagent consumption all contribute to a plateau phase in the later
stages of the reaction during which the amplicon is no longer accumulating at an
exponential rate (Clin Chem Lab Med. 2000;38:833). Reliable quantitation of
PCR therefore involves more than simple measurement of the amount of product
DNA present at the end of 30 to 40 cycles of the reaction. Real-time PCR, also
referred to as Q-PCR, employs real-time measurements of DNA accumulation
(usually via fluorescence-based approaches) during the early exponential phases
of PCR progress to provide precise estimates of the initial concentration of the
target sequence(s).
A wide variety of different chemistries for Q-PCR are in routine use, includ-
ing the so-called TaqMan (also known as 5' exonuclease or hydrolysis real-time
PCR), molecular beacon (which can be designed to distinguish targets differing
by only a single nucleotide), scorpion (also known as self-probing amplicons),
*All e-figures are available online via the Solution Site Image Bank.
896 I SECTION XIII: ANCILLARY METHODS
As with all other molecular diagnostic tests, significant attention must be paid
to NGS results to ensure data quality, including the incorporation of con-
trols that allow the identification of sample contaminations, library chimeras,
sample mix-ups, tumor-normal switches, and variable run quality.
B. Indirect DNA sequence analysis. Indirect identification of normal and mutant
alleles at a specific locus, which correlate with the presence of disease can be
of clinical utility and substitute for direct determination of specific nucleotide
sequences. Virtually all of the indirect methods are based on PCR and can be
applied to a broad range of clinical specimens. Examples of indirect methods
include the following.
1. Allelic discrimination by size. Alleles that vary by small insertions or deletions
can be distinguished on the basis of the size of the PCR product after gel
electrophoresis, perhaps the most straightforward method for indirect DNA
sequence analysis (e-Figs. 60.4, 60.7, and 60.8).
2. Allelic discrimination based on susceptibility to a restriction enzyme. Using
the technique known as restriction fragment length polymorphism (RFLP)
analysis, mutations that either create or destroy a restriction endonuclease
site can easily be distinguished by a two-step process that involves DNA
digestion with the restriction endonuclease followed by gel electrophoresis
to size fractionate the digested DNA. Virtually all RFLP analysis is performed
on PCR product DNA (e-Figs. 60.7, 60.9, and 60.10).
3. Allele-specific PCR. Allele-specific PCR (also known as the amplifica-
tion refractory mutation system [ARMS]) employs oligonucleotide primers
designed to discriminate between normal and mutant target DNA sequences
that may differ by a single base (] Mol Diagn. 2007;9:272). Simultane-
ous analysis of multiple loci via a multiplex PCR format is possible if the
amplicons from different loci are of different sizes. PCR conditions must be
optimized with sufficient stringency so that amplification only occurs when
complete DNA sequence complementarity exists between primer and target
molecules.
4. Single-strand conformational polymorphism (SSCP) analysis. SSCP is one of
the most simple and widely used techniques used as a mutation scanning
system. Single-stranded DNA molecules fold into complex three-dimensional
structures, stabilized primarily by intrastrand base-pairing hydrogen bond
formation, that alter the mobility of the molecule during nondenaturing gel
electrophoresis. SSCP has limited resolution of DNA fragment sizes (100 to
400 bp long is optimal), and while a base sequence may be indicated by the
altered mobility, SSCP does not provide information about either the location
of the base change within the DNA fragment or the chemical identity of the
base change. In practice, the DNA fragments evaluated by SSCP are generated
byPCR.
5. Melting curve analysis. Every DNA duplex has a characteristic melting tem-
perature (dependent on sequence and duplex length) and though small, the
differences in melting temperature between the duplexes can be reliably
detected by high-resolution melting analysis. Differences in the melting tran-
sition of PCR products can therefore be used to infer sequence variations
such as SNPs, small deletions, and small insertions. The most straightforward
methods melt unlabeled PCR products in the presence of DNA-binding dyes
such as SYBR Green that differentiate double-stranded from single-stranded
DNA during melting by changes in fluorescence intensity (Nature Protocols.
2007;2:59).
C. Clonality assays. Demonstration that the cells in a lesion share a common genetic
alteration can be used to support classification as a neoplasm rather than as a
polydonal reactive process, although it is important to emphasize that clonal
neoplasms are not necessarily malignant.
Chapter 60 • Direct and Indirect Methods for DNA Sequence Analysis I 899
1. Assays based on immunoglobulin and T-cell receptor genes. Most PCR clon-
ality assays performed clinically are used to assess lymphoid infiltrates on
the basis of evaluation of immunoglobulin gene or T-cell receptor gene rear-
rangements. PCR primer design is an important component of these assays;
since generation of immunoglobulin and T-cell receptors involves deletions,
template-independent nucleotide additions, and single base-pair changes,
consensus primers are designed to bind to conserved sequence regions, and
multiple sets of primers are used in order to insure that a broad range of
rearrangements can be detected. Demonstration of a monoclonal or oligo-
clonal population of cells within an infiltrate is very often, but not always,
indicative of malignancy since oligoclonal or monoclonal gene rearrange-
ments may characterize reactive lymphoid proliferations.
2. Clonality assays based on specific gene mutations. This class of assays focuses
on detection of specific mutations in individual genes, including single base-
pair changes and larger-scale structural changes (such as deletions or inser-
tions of viral genomes). This type of analysis can be useful when attempting to
show that two neoplasms represent independent synchronous tumors rather
than one tumor with metastases.
D. Microsatellite instability (MSI) assays. Defects in the DNA mismatch repair sys-
tem produce a characteristic pattern of mutations known as MSI. Direct anal-
ysis of the genes responsible for mismatch repair is not desirable in routine
clinical practice because it requires complete DNA sequencing of (at least) four
causative genes; there are no specific mutation "hot spots," and the genes may
be inactive as a result of epigenetic silencing rather than mutation. PCR-based
analysis offers a more efficient, though indirect, method to identify defects in the
DNA mismatch repair system via detection of the short increases or decreases
in the length of the short tandem repeat (STR, or microsatellite) sequences that
are the characteristic feature of MSI. Mutations in the genes which normally
monitor the fidelity of DNA replication of these repeated sequences is defective
and allows for the generation of the variable lengths of repeats in the tumor
cells.
Laboratory testing regimens for MSI have only been formally addressed in
the context of colo rectal cancer (Cancer Res. 1998;58:5248 ). For all other tumor
types, MSI testing is not yet standardized in terms of the number and identity
of microsatellite loci that must be analyzed, or in terms of the number of loci
that need to show length alterations to be considered indicative of MSI.
DNA derived from either fresh or FFPE tissue can be analyzed in the MSI
assay. Comparison of the size of the PCR products from the target STR loci in
the neoplasm versus normal tissue is used to detect changes in the length of the
microsatellite sequences indicative of MSI. In most cases, the profile of PCR-
amplified sequences permits straightforward classification as indicative of MSI
or not; however, there are no uniform criteria for interpretation of marginal test
results, although standards have been proposed (Mutat Res. 2001;461:249).
Sources of variation in MSI analysis include the presence of contaminating non-
neoplastic tissue (which can limit test reliability because demonstration of MSI
in even the most sensitive testing regimens requires that neoplastic cells comprise
at least 10% of the total population), the identity of the microsatellite markers
used in the analysis (the susceptibility of a given microsatellite to instability is
highly dependent on both the number of repeats and the length of the repeat
units), and the potential for biased amplification of some alleles.
E. Infectious disease testing. PCR-based molecular genetic approaches frequently
have higher sensitivity than standard special stains, and can often provide
information not typically available from special stains such as species-specific
identification and drug sensitivity. Molecular methods are also a useful way to
detect organisms that cannot be cultured (e.g., human papilloma virus [HPV])
900 I SECTION XIII: ANCILLARY METHODS
or that are notorious for their slow growth in culture (e.g., Mycobacterium
tuberculosis).
1. Bacteria
a. Mycobacteria. In most assays, PCR targets the highly conserved gene that
encodes the 65 kDa heat shock protein, but other target loci include the
genes encoding 16S rRNA or the repetitive insertion element IS611 0. PCR
testing has been successfully applied to FFPE tissue from a wide variety of
sites, including the respiratory tract, GI tract, GU tract, skin, bone, liver,
and lymph nodes, and also to cytology specimens. Alternative isother-
mal amplification approaches originally developed for use in the clinical
microbiology laboratory have also been adapted for use with processed
tissue specimens (Expert Rev Mol Diagn. 2004;4:251).
b. Helicobacter pylori. Although the genome of H. pylori is remarkable for
the polymorphism between different clinical isolates, PCR-based methods
have nonetheless been developed that permit successful detection of vir-
tually all reported forms of H. pylori by PCR from either fresh or FFPE
tissue, including the nonculturable coccoid form. The most common target
loci in PCR assays include the 16S rRNA gene, urease gene, or arbitrary
regions chosen empirically based on their utility. Recently developed real-
time Q-PCR methods that target the 23S rRNA gene permit simultaneous
detection of H. pylori and antibiotic-resistant testing.
c. Other bacterial pathogens. PCR has been used to detect Bacillus anthracis
organisms in patient specimens associated with bioterrorism or acciden-
tal environmental release from bioweapons facilities (] Clin Microbial.
2002;40:4360).
2. Fungi. Most PCR assays target sequences within the fungal rRNA genes.
PCR can be performed using universal primers that bind to highly conserved
sequences in the region, followed by direct or indirect DNA sequence analysis
of the PCR product to identify the specific fungal pathogen. Alternatively,
sequence differences in the 18S rRNA gene can be used to design primers
that are specific for individual fungal pathogens. Sequence polymorphisms
of the mitochondrial large subunit rRNA gene have also been used as a target
in PCR tests to identify fungal pathogens in tissue specimens, and Q-PCR
methods have also been developed for detection of fungal pathogens in tissue
specrmens.
3. Viruses
a. HPV. Most PCR protocols for HPV testing make use of consensus primers
targeted to the viral Lt gene that are potentially capable of detecting
all HPV types that affect the anogenital region. Following amplification
using consensus primers, the HPV type can be determined by either DNA
sequence analysis or membrane hybridization with type-specific probes.
However, both approaches are labor intensive and difficult to automate,
which makes them poorly suited for screening a large volume of patient
specimens. For this reason, HPV testing of cytology specimens is usually
performed using liquid-based methodologies (as is discussed in more detail
in the cytopathology section of Chap. 34 ).
b. Hepatitis C virus (HCV). RT-PCR methods used to detect HCV in liver
biopsy specimens focus on the 5' noncoding region of the virus that is
highly conserved between the (at least) six genotypes and more than 90
subtypes of HCV that have been described worldwide. Maximal RT-PCR
test sensitivity can only be achieved via a nested RT-PCR approach, or
when the PCR products are evaluated by Southern blot hybridization.
c. Epstein-Barr virus (EBV). A Q-PCR methodology has been described that
targets five highly conserved segments of the EBV genome(] Mol Diagn.
Chapter 60 • Direct and Indirect Methods for DNA Sequence Analysis I 90 1
903
C.D
0
~ ·1 z1 : T~ssue RNA
' T1ssue DNA
I
--+---+ Germ Line DNA
A
- *=>
h
9
n I
000000
000000
000000
000000 < ===
-
I
AAAAA.
~¢::l e
wvvv· ~
C
i--i
~ aCGH eDNA MMt • e
;....:
1
B.a. > I
•
< ~ ••• ¥D
.. ACGGTAT..
SNP I Re-Sequencing
•Expression
5' 3'
Figure 61.1 Microarray applications and technology. Microarray technology can be used for many different
applications, depending upon the nucleic acid composition of the array (probes) and the test material (target) that
is hybridized to it. A: For aCGH assays, fragments of genomic DNA (i.e., BACs) are spotted as probes onto the
microarray surface using robotics. Genomic DNA from the patient's tissue and germ line are then cohybridized to
the array to identify regions of chromosomal gain or loss. B: Alternatively, genomic DNA sequence can be used to
computationally design oligonucleotide probes that detect specific alterations in DNA samples (either germ line
or tissue) for either SNP genotyping or DNA resequencing studies. C: eDNA clones generated from cellular RNA
also can be spotted as probes onto the microarray and patient tissue RNA subsequently hybridized to quantify
relative mRNA transcript abundance for gene expression profiling experiments. D: Similarly, oligonucleotides can
be designed based upon known gene exon structure and spotted on the array surface to perform quantitative or
qualitative assessment of mRNA or miRNA expression.
Chapter 61 • Microarrays I 9 05
Ill. MICRDARRAY ASSAYS. Microarray assays are complex (e-Fig. 61.1)*, both because
of the amount of data generated and the exacting specimen requirements that are
necessary to produce high quality data.
A. Study design. To date, most microarray-based studies have been designed as
biomarker discovery experiments, with the aim of defining a panel of multiple
biomarkers that can then be transitioned into a more conventional clinical assay.
Microarray experiments generally fall into several classes.
1. Class discovery. In such studies, experimental specimens are classified based
solely upon their microarray data values, and the results of the classification
are reviewed to identify new, previously unappreciated clinical or pathologic
classifications. Perhaps the most elegant illustration of this approach has
been the reclassification of breast adenocarcinoma based upon microarray-
generated gene expression profiles (Clin Cancer Res. 2005;11:5678). Similar
studies have effectively identified other molecular subtypes of tumors as well
(] Clin Oncol. 2006;24:5079; N Engl] Med. 2003;348:1777).
2. Class distinction. This type of study is designed to identify novel predictive
biomarkers, patterns of gene expression, CNVs, or sequence alterations that
demonstrate a correlation to an already known parameter such as clinical
outcome or treatment response(] Clin Oncol. 2009;27:1160).
3. Single sample classification. Ultimately, to achieve clinical utility, it is neces-
sary to create a robust molecular signature that can be prospectively applied
to individual patient specimens to accurately predict clinical phenotype. Typ-
ically, a specific subset of probes on a microarray (sometimes a customized
array designed for a specific diagnostic purpose) is examined and a weighted
discriminate index is calculated. The resulting index provides a probability
measure that a given specimen falls into a specific, predefined diagnostic cat-
egory. Studies which independently validate a previously identified signature
are relatively rare to date, but are obviously a critical step in transitioning
any assay into routine clinical use (Clin Cancer Res. 2010;16:5222).
B. Statistical considerations. In principle, microarray data analysis is no differ-
ent than evaluating whether a single biomarker demonstrates a statistically
significant difference between defined sample classes using traditional statis-
tics. By definition, a traditional significance threshold of p = 0.05 allows for
a 5% false-positive (false discovery) rate. Therefore, when analyzing 50,000
to 2,000,000 independent biomarker values obtained by a microarray assay, as
many as 100,000 values will appear to be "significant" by chance alone. To con-
tend with this problem of multiple testing, several methods have been applied to
calculate a true significance threshold when analyzing thousands of variables in
relatively few numbers of samples (Genome Biol. 2003;4:210). Although these
approaches minimize false-positive results for a given sample set, they can in no
way substitute for data validation using multiple, independent sets of samples
across different technology platforms and laboratories.
C. Specimen requirements. Because of the inherent complexity of microarray-based
assays, specimen quality assurance is essential.
1. Specimen collection. Careful consideration must be given to specimen collec-
tion for microarray studies. While DNA and DNA methylation patterns are
relatively stable and probably less sensitive to environmental conditions, the
same is not true for mRNA and miRNA when targeted in microarray-based
gene expression profile assays. Global changes in gene expression can occur
in tissue biospecimens as a result of tissue warm ischemia time (] Clin Oncol.
2006;24:3763 ), creating artificial differences in gene expression patterns seen
between specimens based on collection procedures rather than important
*All e-figures are available online via the Solution Site Image Bank.
Chapter 61 • Microarrays I 9 07
clinical differences. For peripheral blood and bone marrow specimens, the
method in which a specimen is collected and processed can also influence gene
expression signatures (Physiol Genomics 2004;19:247). Finally, most tissue
specimens are inherently heterogeneous collections of many cell types. Vari-
able cellular composition between tissue specimens may lead to differences
in genomic and transcriptional profiles generated from microarray assays.
For example, two prostate tumor samples, one of which contains 5% neo-
plastic cellularity and a second which contains 70% neoplastic cellularity,
may demonstrate two different gene expression signatures based simply on
the content of neoplastic epithelial cells present in the tissue. Similarly, mea-
surement of a tumor-associated change in DNA copy number will vary con-
siderably depending upon the content of neoplastic epithelial cells. For this
reason, many investigators use techniques such as laser microdissection to
isolate more homogeneous cell populations for both gene expression and
DNA copy number microarray analysis (see Chap. 62).
2. Specimen processing. Generally, diagnostic surgical pathology tissue spec-
imens are subjected to formalin fixation and paraffin embedding, a pro-
cess that results in chemical cross-linking and degradation of nucleic
acids. DNA extracted from formalin-fixed, paraffin-embedded (FFPE) tis-
sue may be suitable for some microarray-based DNA analyses (Methods
Mol Biol. 2011;724:127). However, most investigators have found that
RNA derived from FFPE tissue is unsuitable for traditional gene expres-
sion microarray-based assays, although novel molecular amplification pro-
cedures and microarray platforms suggest that this may no longer be true
(]Mol Diagn. 2011;13:48; BMC Cancer. 2011;11:253). Nonetheless, freshly
procured, snap frozen biospecimens remain the "gold standard" for microar-
ray analysis, particularly for RNA-based gene expression assays. Since many
clinical centers do not have access to resources needed for the processing and
storage of frozen samples, a number of solutions have been proposed to cir-
cumvent the limited availability of fresh-frozen biospecimens for microarray
analysis(] Mol Diagn. 2006;8:31). Such advances enable routine prospective
analysis of clinical specimens for RNA- or DNA-based microarray assays.
D. Target preparation. To prepare samples for microarray assays, RNA or DNA
derived from a tissue or cell specimen is converted into a synthetic target in
the presence of labeled deoxynucleotides. In order to analyze clinical specimens
containing small amounts of cellular material, such as diagnostic core or needle
aspiration biopsies, most protocols for microarray target synthesis employ some
method of molecular amplification including PCR, isothermal DNA polymer-
ization, or in vitro transcription (Br] Cancer. 2004;90:1111).
IV. DATA ANALYSIS. Data analysis is by far the most complicated aspect of any microar-
ray study (Nat Rev Genet. 2001;2:418; BMC Bioinform. 2005;6:115). The prin-
cipal steps in microarray data analysis involve the following:
A. Image analysis. Current microarray technology allows for laser scanning of sev-
eral square centimeters of microarray surface at the resolution of micron-sized
image elements. The result is a primary image data file that can be hundreds of
megabytes in size. While a number of software solutions and data repositories
have been developed to hold and distribute experimental microarray data, reg-
ulatory issues related to storage and transfer of data in a clinical setting have
yet to be fully addressed.
The first step in micro array analysis involves the conversion of these raw, pix-
ilated images into numerical values that relate to hybridization signal intensity
at each feature (probe). For two color arrays, the fluorescence intensity must
be sequentially captured and analyzed for each emission spectrum. In other
microarray platforms such as the Affymetrix GeneChip, multiple probes are
used to assay for a single transcript or genomic locus and the signal from the
908 I SECTION XIII: ANCILLARY METHODS
multicellular organisms, this approach has led to sophisticated models for cell
signaling and transcriptional regulatory networks, whereas for humans this type
of analysis is still evolving.
E. Validation. Like any other diagnostic test, the results of a microarray assay must
be validated through independent testing. Validation of microarray assays is
particularly problematic for many reasons. First, microarray assays are relatively
expensive ($200 to $800 per sample), which creates financial constraints on the
number of samples that can be analyzed. Second, given the stringent specimen
requirements needed to perform most microarray assays, availability of suitable
specimens is often limiting. Finally, as discussed above, the large number of
variables associated with a microarray data set requires that a relatively large
number of observations (e.g., samples) must be analyzed to create any degree
of statistical confidence. To perform data validation for microarray results in
the face of these limitations, investigators have devised a number of approaches
(Expert Rev Mol Diagn. 2003;3:587).
1. Cross validation. One of the most popular approaches for data validation is
sequential sampling or "leave-one-out" cross validation analysis in a single
sample set. In an analysis of N study samples, N- 1 samples are used for the
initial statistical analysis to identify groups of signature genes. The ability
of these genes to correctly classify the Nth sample is then calculated and the
gene list modified, discarding biomarkers that perform poorly and solidifying
those with the best performance. This process is repeated, removing all N
samples, one at a time, until a list of genes with the best class prediction score
is created. The advantage of this method is that no additional data or exper-
imentation is needed for validation. Howevet; because the cross-validation
is still applied to a single set of samples (i.e., the "test" and "validation" sets
are one in the same), the ability to generalize conclusions to independent or
larger sample sets may still be limited.
2. Sample set splitting. If an initial microarray sample set is large enough, it
is also possible to divide the experiment into independent sets of test data
and validation data. In this scheme, patterns of "significant" gene expression
are identified using the first set of samples, and patterns are validated in a
second set of arrays. Although this approach utilizes two truly independent
data sets, it necessarily limits the number of independent samples available
for the discovery phase and validation phase. The desire to split a limited
number of samples into test and validation sets raises the question of sample
size requirement for performing microarray analyses with sufficient statis-
tical power (Physiol Genomics. 2003;16:24). While multiple methods have
been proposed to calculate required sample sizes, the number of samples
required will ultimately depend upon the expected biologic effect. For exam-
ple, relatively few study samples may be necessary to identify fundamental
genomic differences between acute myelogenous leukemia {AML) and acute
lymphocytic leukemia (ALL), as these tumor cell types are biologically very
distinct. On the other hand, a considerably larger study set may be required
to identify reliable differences in molecular signatures associated with clin-
ical outcome within ALL patients if the intrinsic biologic basis for patient
outcome is more subtle (BMC Med Genomics. 2011;4:31).
3. Meta-analyses. Microarray data results can be validated using multiple, inde-
pendent study data sets. As an increasing number of microarray studies are
published and corresponding data sets are made publicly available in micro ar-
ray data repositories, it has become increasingly possible to validate pat-
terns of gene expression identified in one experiment using other microarray
experiments in the published literature (PLoS Med. 2008;5:e184). In fact,
meta-analyses of microarray data are becoming more frequent, and while
some studies have shown that significant patterns of gene expression can be
910 I SECTION XIII: ANCILLARY METHODS
(Clin Cancer Res. 2005;11:5678). To date, these studies have been small and
retrospective. However, the ability to use tumor gene expression data to prospec-
tively manage patient treatment will be an important step toward the concept
of personalized medicine.
E. Nontumor pathology. Microarray technology has been applied to biomarker dis-
covery in other fields of pathology and clinical medicine such as neuropathology
(Alzheimer disease, Parkinson disease, epilepsy, schizophrenia), immunopathol-
ogy (systemic lupus, multiple sclerosis), organ transplantation, reproductive
endocrinology, trauma and sepsis, and cardiovascular disease. However, in mul-
tiorgan disease processes, the appropriate target cell population for study is
often not obvious, or often difficult to obtain from a large number of patients.
Therefore, many microarray studies focusing on noncancer disease processes
have been limited to very small sample sizes. Known and unknown variability
within these disease processes and between patient participants makes it diffi-
cult to establish definitive associations between patterns of gene expression and
disease phenotype.
F. Genotyping. DNA sequence alterations, either single nucleotide polymorphisms
(SNPs) present in germline DNA or somatic point mutations which occur in
tumor cells, are important diagnostic markers for disease predisposition, diag-
nosis, and treatment. Microarray technology is a high-throughput method for
genotyping individuals at as many as one million loci across the genome, and
is beginning to have enormous implications in clinical genetics. For example,
familial linkage studies designed to identify inherited disease genes associated
with tumor syndromes have greatly benefited from this technology. Microarray-
based genotyping has also been used in genetic association studies to define loci
associated with disease predisposition and clinical phenotype. Although most
applications of genotyping microarrays have been used to discover a single dis-
ease locus of interest, it is likely that complex multigenic diseases will require
genotyping at multiple loci in order to accurately classify a genetic phenotype.
G. Genome copy number. Another major application of microarray technology
involves assessment of genome copy number (Nature. 2008;452:553). Tumor
cells experience a wide variety of chromosomal gains and losses, and while these
events have been traditionally measured using cytogenetic techniques, microar-
rays are becoming an increasingly useful method to correlate gene copy number
changes with clinical and pathologic features of tumors. Microarray-based gene
copy number assessment has been used to define critical regions, and even sin-
gle genes, whose gain or loss has previously been measured only on the scale
of chromosome arm losses or gains, which has led to the rapid identification
of new oncogenes and tumor suppressor genes. Microarray analysis of gene
copy number has also identified genomic alterations that are characteristic of
particular tumor types and which can be associated with clinical outcome.
Microarray analysis of gene copy number has also demonstrated that the
germline genome of normal populations contains a large number of copy num-
ber polymorphisms as well (Nat Genet. 2007;39:516). This normal variability
in locus copy number has known significance for disease predisposition and
pharmacogenomics, making the use of copy number-based microarray analysis
a useful tool for patient management.
H. Methylation. Methylation of CpG dinucleotides occurs frequently in tumor
genomes and is associated with transcriptional silencing of key tumor suppres-
sor genes. Microarray measurement of methylation patterns has many potential
applications; for example, it has allowed for whole-genome methylation pro-
filing to distinguish subtypes of leukemia, and has made it possible to identify
patterns of methylation associated with treatment response.
I. DNA resequencing. A final, but perhaps most promising, application of microar-
rays involves their use in gene resequencing. Oligonucleotide microarrays can
912 I SECTION XIII: ANCILLARY METHODS
utilize single tube, "hands-off" chemistries that facilitate routine use in the
clinical laboratory. Self-contained microarray cassettes, such as those manufac-
tured by Affymetrix and other vendors, also provide an acceptable format for
regulated clinical tests. Finally, as diagnostic biomarkers become more estab-
lished, microarray platforms will need to evolve into clinical diagnostic tools
which meet all regulatory requirements of standard clinical tests, including addi-
tional standardization and inter-laboratory assay validation, which are both
now receiving appropriate attention (Nat Biotechnol. 2010;28:827).
C. Assay complexity. Microarrays are indisputably high complexity assays, and
although streamlined protocols, automation, and standardized array manu-
facturing can mitigate some inherent technical complexity, microarray assays
still remain in the domain of specialized clinical laboratories. Like most
hybridization-based assays, the time required to perform microarray analy-
sis even in its most automated format may still be three to four days. Assay
turnaround time could be problematic when assay results are needed to guide
immediate therapy. Finally, because microarray assays are based on signal detec-
tion by hybridization, there is an inherent limit to sensitivity as compared with
amplification-based PCR detection assays.
D. Sample requirements. Another technical limitation of microarray assays is their
requirement for high sample quality. Although exact specimen requirements
depend upon the analyte measured (i.e., mRNA, genomic DNA, methylated
DNA), the use of formalin-fixed tissue or other specimens that have been col-
lected under routine hospital conditions currently limits the use of microarray
technology in a number of clinical settings. Methods are available for amplify-
ing and labeling nanogram quantities of nucleic acids samples (both DNA and
RNA), so the ability to utilize small numbers of cells obtained from minimally
invasive procedures such as fine needle aspiration, swabs, or lavages allows
microarray-based assays to be used for diagnostics in an increasing number of
different clinical scenarios. Refinements in the use of routine pathology tissue
specimens that have been fixed and embedded in paraffin will allow full inte-
gration of this technology with routine histopathologic assessment.
E. Clinical relevance. The biggest challenge for implementing microarray technol-
ogy in the clinical laboratory, however, is not array technology itself but rather
the clinical validity of the array biomarker content. To date, the majority of
microarray platforms have been designed as whole genome discovery tools.
Studies have used a relatively small number of observations (patients) and a
large number of variables (genes), an approach which has consistently led to
high false-positive rates that are unacceptable for clinical assay validation. A
considerable amount of microarray data generated and reported in the liter-
ature (particularly gene expression data) has not been reproducible in inde-
pendent samples sets (PLoS Med. 2008;5:e184). Therefore, while microarray
technology itself still holds promise for use in a clinical laboratory, there is
a need for many larger, prospective correlative studies to support the use of
microarray-based biomarker panels in routine diagnostic pathology.
Biospecimen Banking
Mark Watson
I >
~
r==-:>
,[l
•> ..
~
I > u
Figure 62.1 From a single participant, a biospecimen resource may collect multiple specimen types and corresponding data to enable
a wide variety of translational research projects. A corresponding informatics system is often required to track and maintain these data.
q ---+
q - .__
q I
~ ~ ~ ~ ~
co
....
...,
Figure 62.2 Flow diagram of the many processes and resources required to operate a typical full service biospecimen resource
918 I SECTION XIII: ANCILLARY METHODS
*All e-figures are available online via the Solution Site Image Bank.
920 I SECTION XIII: ANCILLARY METHODS
blood specimen collected before and at multiple time points after a course of
therapy.
2. Biospecimen preservation. For molecular and genomic analyses, proper tissue
preservation is critical. A specimen's molecular profile (i.e., gene expression,
protein phosphorylation) can rapidly change from the time it is first col-
lected until the time when it is ultimately preserved by freezing or fixation
(J Clin Oncol. 2006;10;24:3763) due to so-called "warm ischemia time".
Although few evidence-based guidelines exist, warm ischemia time should
be limited and at least documented so as not to introduce additional prean-
alytical variability. Although snap-frozen tissue is the preferred substrate for
many studies, several commercial and "home-brew" tissue preservatives are
available that provide acceptable biospecimen preservation (BMC Genomics.
2004;5:88). Delays in blood processing can affect proteomic biomarker pro-
files (Expert Rev Proteomics. 2006;3:409); unlike tissue, whole blood cannot
be immediately snap frozen, but must be centrifuged to remove the spe-
cific plasma or serum component which then, in turn, must be aliquoted
and rapidly frozen. As with tissue, a number of commercial products are
available to preserve blood cell components at ambient temperature without
freezing, although these products are relatively expensive and require special-
ized downstream isolation procedures (Cytometry A. 2004;59:191; Physiol
Genomics. 2004;19:247).
3. Remote site collection. For multi-institutional research protocols, it may be
necessary to collect biospecimens from remote sites, often from health cen-
ters or medical offices which have little experience in dealing with intricate
biospecimen requirements such as snap-frozen tissue. Proper and efficient
collection of biospecimens from such sites may require preassembled spec-
imen procurement kits (e-Fig. 62.2) and personnel training to ensure the
uniform collection of biospecimens.
4. Specimen quantity. New molecular amplification strategies such as PCR,
whole genome amplification, and transcript amplification allow investigators
to use nanogram quantities of DNA and RNA for analysis. Instrumentation
for proteomic analysis has also become considerably more advanced and,
although it is not possible to amplify protein analytes from biospecimens,
tissue requirements have been minimized even for these technologies. There-
fore, even limited size biospecimens such as needle aspirates, tissue touch
preps, and core biopsies are valuable and frequently amenable to molecular
analyses. Obviously, however, such specimens must be judiciously distributed
to maximize their research potential. Conversely, the collection of large tissue
specimens is not necessarily desirable; for example, large resected carcinoma
specimens are often not properly preserved when fixed or frozen in their
entirety. For such cases, tissue specimens should be divided into samples no
> 1 cm3 for fixation and/or freezing.
5. Associated data collection. Clinical and pathologic data corresponding to
the patient and specimen (for clinical applications), and the participant and
specimen (for research applications), are as important as the specimen itself.
Without these associated data, the utility of the specimens for clinical or
correlative studies is lost. The scope of the data that are collected and stored
with the biospecimen will depend upon the mission of the biospecimen bank.
It is usually impractical to gather detailed clinical information represented
as written notes in a clinical chart or report; instead, it may be more efficient
to rely on electronic sources of data. Although data are usually represented
in text format, surgical pathology reports provide an accurate and detailed
source of pathology information for tissue specimens that are collected and
diagnosed as part of routine clinical care. In some centers, an electronic
medical record may provide access to basic patient demographics and clinical
Chapter 62 • Biospecimen Banking I 9 21
diagnostic data. For cancer patients and their specimens, the hospital tumor
registry is often a reasonably detailed and standardized source for cancer-
related clinical and pathology data. Except for specific studies, it is generally
advisable to collect a limited but standardized data set corresponding to each
collected case, rather than an exhaustive (and inevitably incomplete) data set
for every specimen.
E. Biospecimen data management. As depicted in Figure 62.1, biospecimen banks
may house a complex array of specimens and associated data. It is critical that
specimens be accurately tracked and annotated if they are to be useful (Cancer
Inform. 2008;6:127). While smaller banks may rely on written log books and
electronic spreadsheets to track specimen information, these methods become
rapidly constraining as the bank becomes larger and more diversified. Basic data
types that may be required in a biospecimen bank information system include
the following:
1. Receipt, study association, and consent tracking. An accurate record must
be maintained of when and from where biospecimens were received, and
whether they are intended for clinical or research use. For the lattet; the
intended protocol must also be recorded, as well as the consent under which
the specimens were collected, since the consent likely regulates how the spec-
imens may be used.
2. Inventory and storage. It is important to accurately maintain the availability
and storage location of every specimen so that it can be rapidly retrieved on
demand.
3. Quality assurance. Quality assurance measures such as tissue histology review,
warm ischemia time, details of specimen processing, and nucleic acid quality
should be maintained for each specimen.
4. Clinical annotation. As discussed above, a minimal set of clinical and pathol-
ogy annotation data should be associated with each specimen.
5. Distribution and utilization. The efficacy of a biospecimen bank is judged
almost entirely upon its distribution of biospecimens for clinical testing or
productive translational research. Therefore, detailed data concerning spec-
imen distribution is essential in justifying the activity and operation of the
bank.
Several commercial software packages are available to assist in the man-
agement of biospecimen banks. More frequently, individual centers develop
their own data systems, often creating rather arbitrary and custom data
schemes and data definitions. While such applications may serve the imme-
diate needs of the bank, they present a significant obstacle to collaboration
among different biospecimen resource centers. To address this challenge, the
NCI's Cancer Biomedical Informatics Grid (caBIG) program has fostered
a group of pathologists, biologists, and informatics specialists working to
develop data standards and tools for pathology and biospecimen resources
(https://cabig.nci.nih.gov/workspacesffBPT). Among these tools is the caTis-
sue software application, a freely available web-based tool for managing
biospecimen inventory (https://cabig.nci.nih.gov/tools/catissuesuite).
Ill. BIOSPECIMEN PROCESSING. The same principles of specimen processing used in
the clinical laboratory apply to biospecimens banked for clinical and/or research
purposes. Howevet; since specimens collected for research purposes may be used
for experimental studies that are not a part of traditional diagnostic histopathology,
special specimen processing needs must be considered.
A. Frozen tissue. Although the histologic quality of frozen tissue is inferior to that of
FFPE tissue, most molecular studies require high grade DNA, RNA, and protein
that can only be obtained from frozen specimens. Tissue may be frozen in liquid
nitrogen, an isopentane cryobath (such as is available in most pathology frozen
section rooms), or a make-shift dry ice ethanol bath. Tissue specimens should
922 I SECTION XIII: ANCILLARY METHODS
be no > 1 cm3 to ensure rapid and consistent freezing throughout the specimen.
If the specimen is to be used for histologic sectioning for quality review or
laser microdissection (LM), it may first be embedded in freezing media such as
optimal cutting temperature (OCT) compound. Since OCT material does not
interfere with nucleic acid isolation, but may have an effect on proteomic studies,
it may be advisable to freeze both embedded and nonembedded tissue for the
widest range of downstream uses. Once frozen, tissue may be stored in cassettes
in -80° ultralow freezers or liquid nitrogen (LN2) vapor inventory systems.
While the latter is more expensive and difficult to maintain, specimens stored
in LN2 are immune to the power failures and mechanical breakdowns that can
occur with electric freezers. Any frozen storage system should be equipped with
a temperature recording device to document appropriate storage conditions and
a remote alarm system that can contact the laboratory supervisor in the event
of machine failure or power loss.
B. Fixed tissue. FFPE tissue specimens are not as well suited for molecular and
genomic analyses as frozen tissue. However, the quality of fixed tissue specimens
can be improved using several quality control measures.
1. Control and documentation of fixation and processing times. By minimizing the
time for which tissues are fixed, over-fixation (which leads to excessive anti-
gen crosslinking and molecular degradation) can be avoided. Documentation
of processing times for each specimen can remove this preanalytical variable
from tissue specimens collected over different time periods or from different
locations.
2. Use of 11 mDiecularly friendly" fixatives. Several commercially available and
"home brew" precipitating fixatives preserve tissue and tissue histology while
minimizing the damaging effects to protein antigens and nucleic acids, caused
by cross-linking fixatives such as formalin (Mod Pathol. 2001;14:116; Diagn
Mol Pathol. 2011;20:52).
3. Vacuum storage. Although there are no convincing data to suggest an
added benefit, many biospecimen repositories recommend that paraffin tissue
blocks and, more importantly, cut unstained sections, be vacuumed, sealed,
and stored at 4o. The premise of this storage approach is that antigenicity
and nucleic acids are protected from further degradation by both removal of
oxygen and reduced temperature.
C. Blood components. For isolation and storage of serum and plasma, whole blood
must be immediately spun and the appropriate liquid component removed from
cellular material, aliquoted, and frozen. Processing must be performed rapidly
before cell lysis and protein degradation occurs. Consortia of proteomics inves-
tigators have published recommendations on blood processing for proteomic
studies (Expert Rev Proteomics. 2006;3:409) and while some of these guide-
lines are evidence based, robust guidelines have not been firmly established.
Many institutions freeze whole blood at the site of collection as a convenient
way to store blood for future genomic DNA isolation; however, freezing blood
in glass Vacutainer tubes presents a safety hazard, requires considerably more
freezer space to store, and inevitably causes cell lysis which results in DNA that
is frequently lower in yield, degraded, and contaminated with heme products
that can effect downstream assays. Although more labor intensive, immediate
spinning of whole blood followed by selective removal, washing, aliquoting, and
freezing of the huffy coat creates a high-quality specimen that is easy to store
and results in higher DNA yield and quality. In some cases, it may be desirable
to isolate the peripheral blood mononuclear cell (PBMC) fraction and preserve
these cells; this approach has the advantage of removing peripheral blood gran-
ulocytes and preserving the monocyte fraction which can be manipulated for
future uses such as flow cytometry and cell immortalization. However, viable
preservation of PBMC is expensive, requires additional expertise, and is seldom
Chapter 62 • Biospecimen Banking I 9 23
necessary for simple DNA isolation. When PBMC isolation is not a considera-
tion, whole blood may be stored at 4° for as long as 72 hours with little loss in
PBMC viability (Ann Epidemiol. 2000;10:538).
D. Laser microdissection. For some types of genomic analysis, it may be desirable to
have material derived from homogeneous cell populations. Laser microdissec-
tion (LM) is a method that makes it possible to use a laser to either "capture" or
cut the desired cells away from surrounding tissue under direct microscopic visu-
alization (e-Fig. 62.3). The isolated cells may then be used for DNA, RNA, or
protein isolation for molecular analysis (Acta Histochem. 2007;109:171; Meth-
ods Mol Bioi. 2005;293:187). Although LM instrumentation is expensive, and
the technique is time-consuming and requires expertise, it has become routine
in many biospecimen banks.
E. Tissue microarrays. TMAs provide another format to maximize utilization of
limiting tissue specimens (Methods Mod Med. 103:89, 2005). TMAs are con-
structed by sampling one to three 0.8 mm (up to as large as 3 mm) tissue cores
from a donor paraffin tissue block, and assembling them into an array of as
many as 300 cores in a recipient block (e-Fig. 62.4). The resulting recipient
block can then be sectioned as a traditional tissue block, making it possible to
perform simultaneous immunohistochemistry or fluorescence in situ hybridiza-
tion on each of the 300 cores on a single slide. TMAs allow researchers to
easily validate patterns of biomarker expression in a large sample series using
relatively inexpensive technology. Few special reagents are required and basic
TMA instrumentation is relatively inexpensive, but expert review and informat-
ics tracking of the 300 cores per slides is required. A recognized disadvantage
of the TMA format is that it is prone to sampling error as only a small sample
from each tissue block is incorporated into the array.
F. Nucleic acid. Genomic technologies have shifted requirements from fixed tissue
sections to derivative nucleic acids, and so many biospecimen banks now pro-
duce DNA and RNA for research investigators. Nucleic acid can be effectively
derived from frozen and fixed tissue using several standard protocols, although
the quality from the later is often inferim; as discussed above. In some cases, it
may be desirable to isolate nucleic acid directly from collected specimens, such
as DNA from peripheral blood. Nucleic acids (RNA and DNA) are more stable
once isolated and can be more conveniently stored as compared to the parent
tissue or fluid. However, the initial cost and effort to generate DNA and/or
RNA from every banked specimen as it is received is great, and so prospective
preparation of nucleic acids should only be considered when material will be of
immediate use.
IV. QUALITY ASSURANCE. A biospecimen resource should only provide material that is
subject to rigorous quality control. In fact, one of the main reasons for a pathologist-
supervised biospecimen resource is that it is pathologists who best understand the
approach to and importance of clinical specimen quality control.
A. Specimen identification. Just as in any clinical laboratory, proper specimen label-
ing and identification are critical, because a biospecimen bank may store biospec-
imens for prolonged periods of time; maintenance of proper specimen identifi-
cation is important. As biospecimen banks become subject to regulatory review
as with any clinical laboratory (as noted above), they will need to implement
standard clinical laboratory practices to prevent sample mislabeling including
use of preprinted barcode labels, double data entry, and routine data auditing.
B. Representative tissue sampling. In most cases, biospecimens that are submitted
for clinical diagnosis and those that are submitted to a bank are not identical.
They may be collected at slightly different times or locations, or may involve
sampling bias. For example, in a heterogeneous prostate tumor, a tissue biopsy
banked for research may contain no neoplastic cells although the routinely
processed tissue for diagnosis shows adenocarcinoma; distribution of the banked
924 I SECTION XIII: ANCILLARY METHODS
specimen under the assumption that it truly reflects the diagnostic material may
result in a compromised research study. Consequently, it is important that indi-
vidual biospecimens received by the bank undergo individual diagnostic review,
either prospectively or prior to distribution. Such a review of tissue or cellu-
lar samples may involve confirmation of a histopathologic diagnosis as well
as notation of general cellular features such as histologic preservation, tissue
cellularity, and necrosis. A special circumstance arises when a novel finding or
diagnosis is observed in a banked research specimen that was not reported from
the material for diagnosis; resolution of such a problematic circumstance will
depend upon the policies of both the IRB and pathology service, and should be
documented as part of the bank's standard operating procedures.
C. Tissue preservation and molecular integrity. When the bank will be responsible
for generating and distributing nucleic acids (DNA and RNA), it is important
that molecular integrity is verified. As discussed above, the quality of nucleic
acid is directly related to the manner in which the biospecimen was collected and
preserved. Fixation and delays in specimen processing (warm ischemia time) can
compromise nucleic acid quality as can repeated freeze/thaw cycles (e-Fig. 62.5).
Inherent necrosis and tissue cellularity in the specimen can also affect the quality
of nucleic acids. Documentation of molecular specimen quality can be accom-
plished by readily available (albeit often expensive) instrumentation designed for
quality review of small amounts of nucleic acid, including fiberoptic spectropho-
tometers and capillary microelectrophoresis systems. Metrics have been devel-
oped which relate quantitative results from these instruments to expected per-
formance in downstream applications and assays (BMC Mol Biol. 2007;22:8).
V. BIOSPECIMEN UTILIZATION. The success of any biospecimen bank is measured by the
utilization of collected biospecimens. Distribution of biospecimens for patient care
or funded research projects is also an important revenue source to support the larger
biospecimen banking effort. Although there is often a 3 to 5 year lag phase before
a new biospecimen bank is routinely utilized, while the bank is developing and
maturing its collection, a fundamental principle of biospecimen banking is that
the collection should be biased in favor of those biospecimens that will be used
for funded research projects. Measures which can help promote the productive
utilization of a biospecimen resource include the following.
A. Advertisement Df available resources. This is best accomplished through a web-
based catalog of available sources.
B. Streamlined administrative processes. Often, clinics and researchers are unfa-
miliar with procedures for requesting specimens, obtaining appropriate IRB
approval, and creating necessary MTAs. Having a standardized and facilitated
process for dealing with regulatory steps necessary for biospecimen banking
and subsequent distribution will greatly enhance utilization of a biospecimen
resource.
C. High quality annotated specimens. Clinical testing and translational research can
only be accelerated when the biospecimens distributed are properly quality con-
trolled and "assay ready." Minimal but sufficient clinical and/or pathology data
should be supplied with each specimen in order to easily enable the correlative
analysis.
D. Facilitated and judicious distribution. The process of case selection, specimen
retrieval and processing, specimen annotation, and distribution can be time-
consuming and expensive. As a biospecimen bank matures, increasing requests
may overtax the bank's available resources and can result in significant delays
in providing specimens. Competing interests for limited biospecimens may also
become problematic as bank utilization increases. Therefore, a biospecimen
bank should establish a utilization review committee so that the finite effort
and physical resources of the bank can be fairly allocated to the clinical and
research committees. These committees should be an unbiased representation of
Chapter 62 • Biospecimen Banking I 9 25
speed and optical physics, the latter of which is determined by the magnification
of the scanning objective. The actual image acquisition step occurs via a charge-
coupled device (CCD), which consists of several hundred thousand individual pic-
ture elements (pixels) that transform the optical image into a virtual image; whole
slide images are therefore acquired via a single optical lens that moves over the
slide. Some scanners shorten scanning time by using a meander rather than a linear
scanning pattern to acquire images, but both the meander and the linear meth-
ods have inherent physical limitations that become most apparent when acquiring
multiple images within each plane of section (i.e., when z-stacking). An emerging
technology, so-called lens array microscopy that uses arrays of detectors (miniatur-
ized lenses) to simultaneously capture information from larger areas of the tissue
section, may be able to markedly shorten scanning times but still meet the high
standards of diagnostic pathology (Hum Pathol. 2004;35:1303).
A. Setting up vinual microscopy in the surgical pathology laboratory requires an
electronic and organizational infrastructure as well as a slide scanning instru-
ment. The infrastructure requires an efficient interaction between information
technology (IT) and LIS personnel, and dedicated and trained technical person-
nel (an image technologist) are required to load and maintain the scanning
instrument, perform initial screening (and rescanning if necessary), monitor
scan quality, and distribute the virtual files in a manner that can be conve-
niently viewed by the pathologist (]. Pathol Inform. 2011;2:39). A file server
with the necessary storage capability (upgradeable tera- to petabyte range or
beyond), databases for the virtual files (linked and updated to the LIS), and
the necessary maintenance and updates must be managed by a knowledgeable
IT person (Human Pathol. 2003;34:968). For optimal use in routine patient
care, high-quality LCD monitors of sufficient size (at least 53 em diagonal)
for peripheral vision are required (Hum Pathol. 2006;37:1543), with extended
desktop computer functionality to be able to simultaneously view the LIS, the
gross description and gross images of the specimen, and the virtual slide viewer.
B. Diagnostic and clinical applications of virtual microscopy include diagnostic con-
sultations, archiving, primary diagnosis, and quality assurance (QA).
1. The use of virtual microscopy for diagnostic consultation (so-called
e-consultation) is one of the most useful applications of virtual slide
microscopy. In addition to convenience (no packing or mailing of slides is
required), there are several advantages to e-consultations. Electronic distri-
bution is virtually instantaneous and allows consulting pathologists to review
the scanned slides at any time and location. Online, real-time conferencing
and simultaneous viewing by two or more pathologists is easily achieved. In
addition, there are no risks of losing the primary data (i.e., tissue blocks or
glass slides).
2. Scanning of selected cases for archiving. Virtual slide creation of selected
slides sent in consultation (the medicolegal climate in the United States dic-
tates return of all diagnostic materials to the referring institution) makes
it possible for the consulting institution to have a permanent record of the
diagnostic slides. Such an archive enhances patient care by providing an
immediately available permanent record of the slides to guide frozen section
diagnosis, or final diagnosis at the time of definitive excision. In addition,
the virtual slides are available for subsequent clinicopathologic conferences
(Hum Pathol. 2010;41;751).
3. Primary diagnosis based on review of virtual slides has been shown to have the
same accuracy as primary diagnosis based on routine light microscopy (Arch
Path Lab Med. 2011;135;372), although the use of virtual slides for primary
diagnosis requires an extensive validation procedure. While the use of virtual
slides has dear applications for improved pathology services to underserved
areas (Natl Med] India. 2002;15:363; Ethiop Med ]. 2005;43:51), there is
928 I SECTION XIII: ANCILLARY METHODS
little evidence that digitizing all slides in routine practice results in cost or
time savings (]Path Inform. 2011;2:39; Anal Cell Pathol. 2011;34:1).
4. QA programs may be enhanced by review of virtual slides from cases with diag-
nostic discrepancies. Files can be flagged for subsequent review, and trends
in diagnostic errors easily identified (Dis Mark. 2007;23:459).
C. Education. Virtual microscopy is already an indispensable tool for medical educa-
tion in pathology, including medical student teaching, teaching of pathology resi-
dents and fellows, and continuing medical education for practicing pathologists.
Many national pathology conferences and slide seminars now post virtual slides
on websites before the meeting, and some organizations are building a collection
ofteaching cases (e.g., see the United States and Canadian Academy of Pathology
[USCAP] Virtual Slide Box at www.uscap.org). Other examples of educational
activities based on virtual slides include teaching of histology to medical students
(Anat Rec. 2006;289B:128), a tutorial on Gleason grading of prostatic adeno-
carcinoma (Hum Pathol. 2005;36:3 81), didactic presentations using a combina-
tion of text and virtual microscopy (Ann Diagn Pathol. 2003;7:67), and online
virtual atlases (e.g., of breast pathology at www.webmicroscope.net).
Education and experience with interpretation of virtual slides is critical
for pathology residents and fellows; at the very least, it prepares trainees for
the American Board of Pathology virtual microscopy practice examination
(www.abpath.org/VMinstr.htm).
D. Research applications for virtual microscopy in surgical pathology include quan-
titative image analysis, and imaging of tissue micro arrays used to study patterns
of gene and protein expression. Virtual images can also provide documenta-
tion of specimens or tissue microarrays retained in tissue banks (Eur] Can-
cer. 2006;42:3110), or for patients enrolled in clinical trials. As for diagnostic
pathology, virtual slides make it possible to easily share specimens among inves-
tigators at different sites, anytime. Virtual slides also allow for the electronic
publication of whole slides rather than selected fields of slides, which facilitate
the transfer of new information to practicing pathologists.
Image analysis can be performed on any of the virtual slide file formats pro-
duced by the current generation of slide scanners, and widely accepted freeware
and a variety of subprograms are available to support clinical applications. The
most versatile solution for image processing and analysis is the Nlli freeware
named Image] (Biophot Int. 2004;11:36, available at http://rsb.info.nih.gov/ij/).
The versatility of this program derives from its open architecture that
allows users to implement small and typically customized subprograms (called
"plug-ins"). Over the years, many plug-ins (mostly research-derived) have been
developed; for example, 'Image J for microscopy' (www.macbiophotonics.ca/
imagej) includes numerous tools such as intensity and time analysis, particle
analysis, colocalization analysis, intensity processing, color processing, stack-
slice manipulation, z-functions, t-functions, deconvolution, and annotation.
Ill. TELEPATHOLOGY is the practice whereby pathologists render diagnoses from a dis-
tance by viewing electronically transmitted images rather than by examination of
the glass slides themselves by light microscopy. Electronic Images can be transmitted
by ordinary telephone lines, high-speed digital lines, or satellites, but increasingly
the images are transmitted via the Internet. With some overlap, three systems are
currently available: dynamic, static, and virtual.
A. Dynamic telepathology systems. In these systems, pathologists view images in real
time by electronic control of a distant robotic microscope that has motorized
optics and a motorized stage. Dynamic-robotic telepathology has been primarily
used to provide intraoperative frozen section diagnoses to hospitals without on-
site pathologists (Hum Pathol. 2007;38:1330). Typically, in less than a minute,
a digital overview of the slide is created; virtual controls enable the pathologist
Chapter 63 • Imaging Technologies in Surgical Pathology I 929