Atlas of Gastrointestinal Pathology
Atlas of Gastrointestinal Pathology
DORA M. LAM-HIMLIN, MD
Associate Professor
Department of Laboratory Medicine and Pathology
Mayo Clinic
Scottsdale, Arizona
ELIZABETH A. MONTGOMERY, MD
Professor of Pathology, Oncology, and Orthopedic Surgery
Department of Pathology
Division of Gastrointestinal and Liver Pathology
Johns Hopkins Medical Institutions
Baltimore, Maryland
CHRISTINA A. ARNOLD, MD
Associate Professor
Department of Pathology
Division of Gastrointestinal and Liver Pathology
The Ohio State University Wexner Medical Center
Columbus, Ohio
Acquisitions Editor: Ryan Shaw
Editorial Coordinator: Lindsay Ries
Marketing Manager: Julie Sikora
Production Project Manager: Linda Van Pelt
Design Coordinator: Holly McLaughlin
Manufacturing Coordinator: Beth Welsh
Prepress Vendor: TNQ Technologies
All rights reserved. This book is protected by copyright. No part of this book may be reproduced or transmitted in any
form or by any means, including as photocopies or scanned-in or other electronic copies, 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. To request permission, please contact
Wolters Kluwer at Two Commerce Square, 2001 Market Street, Philadelphia, PA 19103, via email at
[email protected], or via our website at shop.lww.com (products and services).
987654321
Printed in China
This work is provided “as is,” and the publisher disclaims any and all warranties, express or implied, including any
warranties as to accuracy, comprehensiveness, or currency of the content of this work.
This work is no substitute for individual patient assessment based upon healthcare professionals’ examination of each
patient and consideration of, among other things, age, weight, gender, current or prior medical conditions, medication
history, laboratory data and other factors unique to the patient. The publisher does not provide medical advice or
guidance and this work is merely a reference tool. Healthcare professionals, and not the publisher, are solely
responsible for the use of this work including all medical judgments and for any resulting diagnosis and treatments.
Given continuous, rapid advances in medical science and health information, independent professional verification of
medical diagnoses, indications, appropriate pharmaceutical selections and dosages, and treatment options should be
made and healthcare professionals should consult a variety of sources. When prescribing medication, healthcare
professionals are advised to consult the product information sheet (the manufacturer’s package insert) accompanying
each drug to verify, among other things, conditions of use, warnings and side effects and identify any changes in
dosage schedule or contradictions, particularly if the medication to be administered is new, infrequently used or has a
narrow therapeutic range. To the maximum extent permitted under applicable law, no responsibility is assumed by the
publisher for any injury and/or damage to persons or property, as a matter of products liability, negligence law or
otherwise, or from any reference to or use by any person of this work.
shop.lww.com
To my family:
Matt: For being my rock.
Madeline: Stay brave. Stay curious.
Matthew: Always have the heart of a gentleman.
Tommy: Yes, I’d rather be outside, too.
Dora M. Lam-Himlin, MD
To my loving family:
Mary and Andy: Mike and I didn’t realize how much we needed parents, until
we became parents ourselves.
Thank you for always being there for us, and for reminding us about the
importance of family, love, and laughter.
Mom: Thank you for being my first best friend, believing in me always, and
teaching me the value of hard work.
Our Wednesday afternoon brownies are some of my favorite memories.
Jackson and Madelyn: Objects in the mirror are sometimes closer than they
appear. If it is important,
NEVER give up—hunker down and dig in!
Mike: The keys to a good road trip are fun, excitement, danger, and Johnny Cash.
OGN: This book and my career would not be possible without you. Although I
don’t deserve you,
I thank the heavens for you every day. Here’s to Baltimore, the Inca trail, and all
of tomorrow’s
mischievous adventures.
Christina A. Arnold, MD
PREFACE
• Each chapter opens with a “Chapter Outline” that outlines the enclosed structure and
allows the reader to quickly hone in on select patterns and pertinent differential
diagnostic considerations. Similar checklists are found throughout the chapter to neatly
organize complicated topics.
• “The Unremarkable X”: Normal histology is sometimes overlooked in textbooks
because it is assumed to be widely understood, much to the frustration of junior
trainees. A firm understanding of normal is essential to recognizing subtle injury
patterns. As such, each chapter begins with a brief discussion of normal histology to
contrast to the succeeding mucosal injury patterns and to highlight helpful diagnostic
clues.
• The “Pearls & Pitfalls” boxes include lessons from real life sign-out experience with
an emphasis on important diagnostic clues, mimics, and hazards.
• The “Frequently Asked Questions” sections stem from our busy consult service and
teaching sessions. In these sections, we discuss real-life diagnostic dilemmas and
offer diagnostic tips and tools to sort through commonly encountered sign-out
challenges.
• All major topics close with a “Key Features” section that summarizes the essential
elements of the subtopic for handy reference.
• A “Sample Note” section accompanies the more challenging topics. In these sections,
an example pathology report is included with the top-line diagnosis, pertinent
discussion, and salient references. These notes offer a template of how to synthesize
complicated topics and are based on real-life cases and interactions with clinicians.
The select references are included for those interested in further reading but also can
be included in pathology reports to help guide clinical management.
• Each chapter features a corresponding “Quiz” section in the appendix to emphasize
important teaching points. These sections offer the reader experience and confidence
with high-yield teaching topics. Questions are in the format of the board type
examinations and can also serve as useful board preparatory materials.
ACKNOWLEDGMENTS
We thank our institutions, colleagues, and trainees for invaluable resources and support.
We are indebted to our inquisitive trainees and clinicians whose fresh perspectives and
lively discussions drove the direction of this book. We particularly thank our families
for understanding the numerous late night, early morning, and weekend marathon writing
sessions.
We thank our Acquisition Editor, Ryan Shaw, for taking a chance on this project, and
our Editorial Coordinator, Lindsay Ries, for working diligently with us to ensure timely
completion. We thank Frank M. Corl, MS, for the custom medical illustrations; Rick
Marshall for computer assistance in identifying pertinent teaching material; and Shawn
Scully for photography editing on select topics.
Lastly, we thank the production team led by Ramkumar Soundararajan for their
careful attention to detail.
CONTENTS
1 ESOPHAGUS
The Unremarkable Esophagus
Some Esophageal Polyps
Barrett Esophagus
Squamous Neoplasia
Near Misses
2 STOMACH
The Unremarkable Stomach
Polyps
Adenocarcinoma
Well-Differentiated Neuroendocrine Tumors (Formerly “Carcinoid”)
MALT Lymphoma
Mesenchymal Lesions
Near Miss
3 SMALL BOWEL
The Unremarkable Small Bowel
Polyps
Adenocarcinoma
Neuroendocrine Tumors and Neuroendocrine Carcinomas
Near Miss
4 COLON
The Unremarkable Colon
Polyps
Dysplasia in Inflammatory Bowel Disease
Adenocarcinoma
Near Miss
5 ANUS
The Unremarkable Anus
Nondysplastic Polyps
HPV-Associated Neoplasms
Adenocarcinoma
Extramammary Paget Disease
Malignant Melanoma
Near Miss
6 MESENCHYMAL LESIONS
The Layout of the Real Estate
Immunolabeling Comments
Mesenteric Lesions
Translocation Sarcomas Associated With the Gastrointestinal Tract
Other Spindle Cell Lesions
Summary of Gastrointestinal Tract Lesions Containing Many Inflammatory
Cells
Select Colorectal Lesions Likely to Be Encountered on Biopsies
Summary of Neural Polyps
Near Miss.
A Few Vascular Tumors.
Near Miss.
APPENDIX A
APPENDIX B
INDEX
ESOPHAGUS 1
CHAPTER OUTLINE
The Unremarkable Esophagus
Some Esophageal Polyps
□ Granular Cell Tumor
□ Leiomyoma
□ Gastrointestinal Stromal Tumors of the Esophagus
Barrett Esophagus
□ Grading Barrett Dysplasia
• Barrett Esophagus, Negative for Dysplasia
• Barrett Esophagus Indefinite for Dysplasia
• Low-Grade Dysplasia
• High-Grade Dysplasia
• Intramucosal Adenocarcinoma and Adenocarcinoma
• Endoscopic Mucosal Resection Specimens
• Dysplasia Recapitulation
Squamous Neoplasia
□ Risk Factors
□ Squamous Carcinoma Precursor Lesions
• Squamous Dysplasia
• Epidermoid Metaplasia
• Squamous Cell Carcinoma
Near Misses
□ What Went Wrong?
• Neuroendocrine Tumors of the Esophagus
• Adenosquamous Carcinoma
• Spread of Extraesophageal Carcinoma to the Esophagus
• Melanoma
• Esophageal Hematopoietic Disorders
Figure 1-1. Esophageal mucosal biopsy. This sample is slightly tangentially embedded. Note
that the biopsy contains all three layers of the mucosa, namely, the squamous epithelium, the
loose lamina propria with a few delicate blood vessels, and the muscularis mucosae (this is
Latin for the muscle of the mucosa). The epithelium has only a few layers of the darker basal
cells, and the more superficial cells are pink (eosinophilic), with their long axes arranged
parallel to the basement membrane, which is normal polarity for squamous epithelium. For
columnar epithelium, the long axes of the nuclei are normally arranged perpendicular to the
basement membrane. This sample is essentially normal and fairly well oriented.
Figure 1-2. Esophageal mucosal biopsy. This mucosal biopsy has been embedded in a
disorderly fashion such that it is a bit trickier to interpret than the sample shown in Fig. 1.1
There is a lymphoid aggregate at the left. The muscularis mucosae is tangentially embedded
and appears thick, but this is not muscularis propria. The loose connective tissue at the upper
right is lamina propria rather than submucosa.
LEIOMYOMA
Leiomyoma is by far the most common spindle cell tumor of the esophagus, but it is still
uncommon. Esophageal leiomyomas arise in young patients (well, at least compared
with one of the authors—median age, 35 years),9 with a male predominance. They
consist of cells with eosinophilic cytoplasm (Fig. 1.11) and express desmin and alpha-
smooth muscle actin, but not CD117 and CD34. The important pitfall to be aware of in
diagnosing gastrointestinal tract leiomyomas is that, if one performs immunolabeling for
CD117 and DOG1, these stains label Cajal cells that are either entrapped in or
proliferating along with the lesion (Fig. 1.12). For this reason, confident morphologists
avoid these stains. Esophageal leiomyomas are easy to diagnose on staining with
hematoxylin and eosin (H&E)—they are hypocellular, pink, and benign.
Figure 1-7. Granular cell tumor. The tumor consists of plump eosinophilic cells with granular
cytoplasm and small nuclei. This type of tumor is notorious for stimulating hyperplasia of the
overlying squamous epithelium (so-called pseudoepitheliomatous hyperplasia), which can be
mistaken for carcinoma.
Figure 1-8. Pseudoepitheliomatous hyperplasia associated with an adenocarcinoma. The
adenocarcinoma at the bottom of the field has undermined the squamous epithelium at the top
of the field. The squamous process is benign and reactive.
Figure 1-9. Pseudoepitheliomatous hyperplasia associated with an adenocarcinoma. This is a
high-magnification image of the same lesion as that seen in Fig. 1.8. The surface squamous
component is benign and simply reacting to the adenocarcinoma beneath it.
Figure 1-10. Granular cell tumor. This stunning S100 protein immunohistochemical stain shows
striking nuclear and cytoplasmic expression.
Figure 1-11. Esophageal leiomyoma. The lesional cells are brightly eosinophilic, and the tumor
has low cellularity. The cytoplasm is fibrillary, and paranuclear vacuoles are present.
Figure 1-12. Esophageal leiomyoma. This is a CD117/KIT stain. Do not be fooled by the
scattered labeled cells. Whether these are entrapped Cajal cells or an integral part of the
leiomyoma is not clear, but they should not result in an interpretation of gastrointestinal stromal
tumor.
Most patients in Miettinen’s early series died of their tumors, but the study was from
the era before targeted therapy. Regardless, in contrast to GISTs of the stomach,
esophageal examples should be considered aggressive.
BARRETT ESOPHAGUS
Evaluating for Barrett esophagus and the neoplasia associated with it is a large part of
the practice of Western pathologists. Columnar metaplasia in the esophagus is the source
of anxiety for patients and pathologists alike. This section is approached with a short
list of questions before dysplasia is discussed.
Answer: It depends on whom you ask! In the United Kingdom and Japan the
definition differs from that in the United States, and at this writing, there is
some flux at play in the United States.
British (and Japanese) definition of Barrett mucosa 2014:
• Columnar epithelium with or without goblet cells extending ≥1 cm above the
gastric folds10
American Gastroenterological Association definition of Barrett mucosa 2011:
• Columnar epithelium in the esophagus that contains goblet cells—no length
requirement11
American College of Gastroenterologists’ definition of Barrett mucosa 2016:
• Columnar epithelium with goblet cells extending ≥1 cm above the top of the
gastric folds12
The last definition (the 2016 one from the American College of Gastroenterologists)
makes the life of the pathologist challenging. In some instances, we have a good idea
about the length of a segment of columnar epithelium in question, whereas in others, the
only information we have is “esophagus.” Obviously, if we have a sample labeled
“esophagus, 40 cm” and we see intestinal metaplasia and we have a second sample that
is labeled “esophagus, 34 cm” and we see intestinal metaplasia, it is clear that the
affected segment of lesion measures at least 1 cm. Interestingly, the gastroenterology
colleagues who compiled the recommendations even went so far as to caution our
endoscopy colleagues to refrain from biopsies of the gastroesophageal junction unless
there was a visible alteration. At least in our hospitals, there seems to be little
compliance with the latter suggestion. The American College of Gastroenterology
suggested the term “specialized intestinal metaplasia of the esophagogastric junction”
for lesions that contain goblet cells but do not satisfy the requirement for the mucosal
irregularity to extend at least 1 cm above the top of the gastric folds.12
To get around the length issue, we have developed two notes that we find useful for
situations for which (1) we see intestinal metaplasia and we do not know the segment
length or (2) the sample is labeled “gastroesophageal junction” and we see intestinal
metaplasia.
Note: The diagnosis of Barrett esophagus is made owing to the presence of goblet cells
(intestinal metaplasia), with the assumption that the biopsies were obtained from
columnar mucosa in the distal esophagus and the mucosal irregularity extends at least 1
cm above the top of the gastric folds as per 2016 American College of Gastroenterology
(ACG) guidelines.
Reference:
Shaheen NJ, Falk GW, Iyer PG, Gerson LB; American College of Gastroenterology.
ACG clinical guideline: diagnosis and management of Barrett’s esophagus. Am J
Gastroenterol. 2016;111(1):30-50.
Note: This biopsy shows gastric-type mucosa with scattered goblet cells. The diagnosis
in this case depends on the location of this biopsy. If this biopsy was taken from the
tubular esophagus and the mucosal irregularity extends at least 1 cm above the top of the
gastric folds, the diagnosis is Barrett mucosa of the distinctive type. If this biopsy was
taken from the gastric cardia, the diagnosis is intestinal metaplasia of the gastric cardia.
Reference:
Shaheen NJ, Falk GW, Iyer PG, Gerson LB; American College of Gastroenterology.
ACG clinical guideline: diagnosis and management of Barrett’s esophagus. Am J
Gastroenterol. 2016;111(1):30-50.
FAQ: The ACG 2016 criteria state: BE should be diagnosed when there is
extension of salmon-colored mucosa into the tubular esophagus
extending ≥1 cm above the top of the gastric folds with biopsy
confirmation of IM (strong recommendation, low level of evidence). So, if
there is a 0.5-cm island that is >1 cm above the top of the gastric folds
that contains intestinal metaplasia, is it Barrett esophagus?
FAQ: Why do some observers want to eliminate the requirement for goblet
cells for a diagnosis of Barrett mucosa in the United States?
Answer: There are some studies that suggest that most esophageal
adenocarcinomas that are detected arise in the absence of intestinal
metaplasia. In one of them, the authors used EMR samples and found adjoining
intestinal metaplasia in less than half of the samples with early cancers.14
However, these colleagues did not attempt to learn if the patients had separate
samples that contained intestinal metaplasia. In two West Coast US studies,
intestinal metaplasia essentially always accompanied high-grade columnar
epithelial dysplasia and carcinomas.15,16 We found similar results in an East
Coast study17 and would endorse retaining the requirement for goblet cells, but
others have suggested eliminating the requirement.18 Regardless, there are a
few cases of esophageal adenocarcinomas that are unassociated with
intestinal metaplasia, but they are not numerous in our Western population.
Elimination of the requirement for goblet cells to diagnose Barrett esophagus in
the United States would even further reduce the cost-effectiveness of
surveillance of patients with Barrett esophagus because it would open the
floodgates to patients with reflux. A key issue is that, when cardiac mucosa is
found (without intestinal metaplasia), it could be a normal finding, so the
presence of intestinal metaplasia offers some assurance that at least abnormal
tissue has been biopsied.
Answer: This topic has been addressed in the non-neoplastic volume preceding
this volume (Atlas of Gastrointestinal Pathology. A pattern based approach to
non-neoplastic biopsies), although we have changed our practice patterns. We
performed up front periodic acid–Schiff/alcian blue staining on upper
gastrointestinal tract samples (including all biopsies of the esophagus) in the
past; we no longer follow this practice, but there are occasional cases for
which it is instructive. Both pancreatic acinar heterotopia (Figs. 1.5, 1.6, 1.16
and 1.17) and some gastric foveolar cells (Figs. 1.18 and 1.19) can suggest
intestinal metaplasia. Pancreatic heterotopia contains cells with granules, so
these cells can be easily distinguished from goblet cells, but some gastric
foveolar cells can take on a bluish hue or show weak alcianophilia on alcian
blue staining. These latter cells lack the true intestinal differentiation that we will
see later.
Figure 1-16. Pancreatic acinar cell heterotopia. This image allows us to further consider the
layer in which pancreatic acinar cell heterotopia is found. In this case, there is squamous rather
than cardiac-type mucosa associated with the pancreatic acinar cell heterotopia. Note that it is
found in the lamina propria. There is squamous epithelium over it, cardiac glands to the right,
and muscularis mucosae below it.
Figure 1-17. Pancreatic acinar cell heterotopia. This high-magnification image shows zymogen
granules in the cells to full advantage.
Figure 1-18. “Fake” goblet cells. A few of the cells in this cardiac gland have a bluish tint but
lack the demarcation of goblet cells.
Figure 1-19. “Fake” goblet cells. This is a periodic acid–Schiff/alcian blue stain. It shows a bit of
nonspecific alcianophilia, but no goblet cells are present. This does not qualify as Barrett
mucosa.
Figure 1-20. Barrett esophagus with complete and incomplete intestinal metaplasia. In
incomplete intestinal metaplasia, the mucosa has incompletely transformed from gastric
cardiac type to an intestinal phenotype such that there are goblet cells (see the arrow beneath
the “incomplete” annotation) interspersed with gastric foveolar cells with apical mucin. In
contrast, the complete type of intestinal metaplasia fully recapitulates intestinal epithelium and
cells with brush borders are seen between goblet cells (a goblet cell is marked with an arrow
underneath the zone marked as “complete”).
Figure 1-21. Barrett esophagus with complete and incomplete intestinal metaplasia. This is a
high-magnification image of the area shown in Fig. 1.20. It is not necessary to specify the type
of metaplasia in reports unless there is a specific protocol, but, on epidemiologic grounds, the
incomplete type is more likely to progress to carcinoma. In the individual case, it is not an
important factor.
Figure 1-22. Multilayered epithelium. This is a high-magnification image showing epithelium and
a tiny sliver of lamina propria at the bottom of the image. At first glance, the basal cells appear
hyperchromatic, but a quick glance at the lymphocytes at the lower right is reassuring. The
epithelial cell nuclei are only slightly larger than the lymphocyte nuclei, so this is not squamous
dysplasia. Note that the cells at the surface contain bubbly mucin-filled cytoplasm that is bluish,
a feature similar to the mucin in goblet cells, but the mucin does not form a single discreet
droplet in the manner of goblet cells. The appearance of esophageal multilayered epithelium is
similar to that of immature squamous metaplasia of the uterine cervix. This type of epithelium is
found in patients with reflux and can also accompany conventional Barrett mucosa. There is
also some experimental evidence that this type of epithelium gives rise to conventional Barrett
mucosa.21 We do not currently report this finding.
Figure 1-23. Multilayered epithelium. This is a periodic acid–Schiff/alcian blue stain. Some of
the mucin-laden cells display alcianophilia (have a blue appearance), but their morphology is
different from that of actual goblet cells like those seen in Fig. 1.21.
Our endoscopy colleagues use a wonderful system to describe the extent of Barrett
esophagus called the Prague system (of course they got to have a fancy meeting in
Prague, probably with nice concerts and fancy food and a posh hotel, whereas we have
to meet in Baltimore or Columbus, but that is fine because we get to sit down when we
do our daily work) in which the distance of the circumferential length of Barrett mucosa
(C) and the maximum length (M) are recorded.22 This method has allowed
standardization of endoscopy reports and is important for pathologists to also
understand. When these data are provided to us, they can help us understand whether the
samples are from long-segment of short-segment Barrett mucosa—long segment means
more than 3 cm. This scheme is seen in Fig. 1.26.
Figure 1-26. Schematic for the Prague classification for Barrett mucosa. In this scheme, which
is used worldwide by endoscopy colleagues, the distance of the circumferential length of
Barrett mucosa (C) and the maximum length are recorded (M). If the information is provided to
the pathologist, it can be used to decide whether the findings in any given case meet criteria for
Barrett mucosa because a minimal length criterion has been introduced.
Answer: The short answer is “nope,” but, of course, we all occasionally need a
bit of help. However, we would note that, when Panarelli and Yantiss reviewed
the topic, they concluded that neither histochemical nor immunohistochemical
stains add value over H&E stains because they tend to produce false-positive
results.23
In the past, the idea that CK7/CK20 stains could be used to differentiate
esophageal intestinal metaplasia from gastric cardiac intestinal metaplasia was
entertained but did not really catch on.24,25 Long-segment Barrett esophagus
cases (>3 cm) were characterized by superficial and deep CK7
immunoreactivity in the intestinalized mucosa, with only superficial CK20
staining in the intestinalized zones. In contrast, distal gastric intestinal
metaplasia was characterized by patchy, superficial, and deep CK20 staining in
areas of incomplete intestinal metaplasia; strong, superficial, and deep CK20
staining in areas of complete intestinal metaplasia; and patchy or absent CK7
staining in either type of gastric intestinal metaplasia. Many other immunostains
have been studied, including mucin core (MUC) polypeptides, to better
characterize gastric cardiac versus esophageal intestinal metaplasia. The MUC
polypeptides seem to be of little practical value in any given patient and are
presently of academic interest only. They include MUC5 (gastric foveolar
mucin), MUC6 (cardiac glands, antral glands, Brunner glands), and MUC2
(goblet cells). Some have used CDX2 staining to label areas of intestinal
metaplasia.26 Others have used it to note that some cases lacking goblet cells
express these markers (illustrated later) anyway to make the point that the
United States should drop the requirement for goblet cells to diagnose Barrett
esophagus.27 Others have suggested that hepatocyte antigen (Hepar-1,
carbamoyl phosphate synthetase 1) is helpful in detecting intestinal metaplasia
or processes that are intestinalized in the absence of goblet cells.28 It seems
more practical to simply search for goblet cells because that can be done
consistently in any laboratory without the added costs of immunolabeling. In
fact, none of the immunostains intended to detect goblet cells offers added
value over H&E stains.23 Fig. 1.27 was prepared from a consultation case. It is
a CDX2 stain of Barrett mucosa in which every single nucleus is labeled,
including both those of goblet cells and those of gastric foveolar cells.
Figure 1-27. Esophageal columnar mucosa with scattered goblet cells, CDX2 stain. This is
interesting in that cells that are clearly goblet cells, such as the one near the tip of the villuslike
structure at the upper left, and all other columnar cells are reactive. This finding suggests that
the cardiac-type epithelium already has some traits that overlap with those of the intestinal-type
epithelium. However, at least in a US population, goblet cells are probably a better marker for
patients who require surveillance than columnar epithelium alone.17
The case shown in Figs. 1.37–1.40 has reactive features and illustrates another
pitfall. The deep glands have slightly enlarged hyperchromatic nuclei in a context of
inflammation and gradual surface maturation. A TP53 (P53) stain is also shown for this
case and shows a wild-type pattern. Many laboratories endorse the use of TP53
immunolabeling in all Barrett cases, and this is a common practice in Europe. We have
not adopted this practice, but if P53 labeling is always used in your practice, it is
important to think before reacting to light staining. This protein is a tumor suppressor
protein, and thus it is active during cell proliferation and has a quick half-life. Because
it is busy preventing cancer during cell proliferation, a bit of labeling is to be expected
in the proliferative compartment of the mucosa. As such, there is always a bit of nuclear
labeling in the basal layer of the squamous epithelium and in the pits of the stomach.
When the TP53 gene is mutated, this results in a TP53 protein with an extra-long half-
life, so it accumulates in the nuclei of the cells and can be detected by immunolabeling
and is a wonderful marker for dysplasia, but intense staining should be used to confirm
dysplasia. Fig. 1.41 shows an area of intense labeling in a zone that could be interpreted
as basal pattern dysplasia (basal crypt dysplasia31), which is discussed later. The so-
called null pattern of P53 labeling is discussed in the “High-Grade Dysplasia” section.
Figure 1-38. Barrett mucosa with reactive changes and inflammation, negative for dysplasia.
This is a very-high-magnification image of the gland indicated in Fig. 1.37. This was not a good
idea! Mitoses are present (acceptable in deep glands), and the nuclei appear somewhat
jumbled and hyperchromatic out of context.
Figure 1-39. Barrett mucosa with reactive changes and inflammation, negative for dysplasia.
This is another high-magnification image of some of the deep glands in the case seen in Fig.
1.37. The nucleoli in the gland with cells containing abundant eosinophilic cytoplasm and a
somewhat syncytial arrangement have all of the features of a reparative process. This sort of
appearance can be seen after any type of injury (including mucosal ablation). This field shows
reactive nuclei rather than dysplastic ones.
Figure 1-40. Barrett mucosa with reactive changes and inflammation, negative for dysplasia.
This is a TP53 (P53) immunohistochemical stain. It shows a wild-type (not mutated) pattern
that would be expected in mucosa with reparative/reactive changes. The P53 protein is a tumor
suppressor protein encoded by a tumor suppressor gene. Its job is to suppress neoplastic
transformation while cells are going through the proliferation process every time our mucosa is
made and remade. As such, there should be a little of this protein in cells that are proliferating
or regenerating an area (our mucosa turns over constantly). As such, there is staining in the
basal layer of the squamous epithelium and in the deeper glands. The normal gene produces a
protein that has a short half-life, so we just see just a little bit of it accumulate in proliferating
nuclei. Notice that there is none at the columnar surface, and only the basal squamous nuclei
are labeled. A few of the lymphocyte nuclei are also labeled lightly. When the TP53 gene is
mutated, it produces a protein that is not properly degraded and has a longer half-life, so the
cells with mutant TP53 show prominent nuclear labeling. Do not equate light labeling such as
that in this image as evidence of dysplasia.
Figs. 1.42–1.45 are images taken from biopsies from a single patient. Are the
features reactive? In some areas the four lines are present, but in other areas these lines
are jumbled and some of the surface nuclei are arranged in a disorderly fashion. Are the
nuclei hyperchromatic? Maybe. In one area a deeper gland appears hyperchromatic but
perhaps it is crushed. Based on the uncertainty, this process was interpreted as
indefinite for dysplasia. Figs. 1.46–1.51 show similar alterations in the setting of
inflammation that were interpreted as indefinite for dysplasia. There is no shame in
diagnosing lesions as indefinite for dysplasia, but the number can be reduced by
showing colleagues the case, and sometimes the presence of a wild-type P53
immunolabeling pattern can be reassuring. Sometimes fresh eyes can clarify the
findings! This diagnosis should be used in only a small percentage of cases (up to 3% to
5%). Some colleagues (personal communications) essentially never use this category.
Perhaps these colleagues are very good and always know. We wish we were that good!
Figure 1-42. Barrett esophagus, indefinite for dysplasia. In this case, the findings are difficult to
interpret. There is a large complex gland at the right and adjoining squamous epithelium at the
left. Many of the nuclei on the surface at the right are hyperchromatic, but it is not clear if they
are reactive. The lines are obscured at the upper right, but the nuclei are not particularly
enlarged. Perhaps this is all reactive, but it is difficult to be entirely certain.
Figure 1-43. Barrett esophagus, indefinite for dysplasia. This is from the same case as that
seen in Fig. 1.42. One could argue that the lines are intact at the upper right and assume that
the findings at the upper left reflect tangential embedding. However, the image seen in Fig. 1.44
is from the same case as well.
Figure 1-44. Barrett esophagus, indefinite for dysplasia. There is an atypical gland at the left of
center, but the section appears thick in that focus. The overlying nuclei are not particularly
enlarged, but the lines are obscured. Is this reactive? Not sure. Is this dysplastic (adenoma
like)? Not sure.
Figure 1-45. Barrett esophagus, indefinite for dysplasia. This is a higher magnification of the
field seen in Fig. 1.44. The nucleoli in the glands at the left suggest that the findings are
reactive, but the jumbled nuclei on the surface at the left are concerning, but they gradually
merge with an area to the right that has the four lines.
Figure 1-46. Barrett esophagus, indefinite for dysplasia. In this sample, the surface shows
prominent nuclear stratification and the lines are obscured. However, the nuclei are smaller
than the ones in the deep glands. The features are adenoma-like (low-grade dysplasia), but the
key issue is obscuring inflammation. It is no problem to consider high-grade dysplasia in the
setting of lots of inflammation, but obscuring inflammation is problematic for diagnosing low-
grade dysplasia.
Figure 1-47. Barrett esophagus, indefinite for dysplasia. This is a high-magnification image of
the lesion seen in Fig. 1.46. The nucleoli at the surface do suggest that the findings are reactive
in the setting of obscuring inflammation, but the features at low magnification are striking such
that a “treat and repeat” scenario is not unreasonable.
Figure 1-48. Barrett esophagus, indefinite for dysplasia. This image is from a different case
from the one seen in Figs. 1.44–1.47. In this example, inflammation is prominent as is “chatter
artifact,” but there seem to be some surface alterations that are concerning for low-grade
dysplasia.
Figure 1-49. Barrett esophagus, indefinite for dysplasia. This image is from the same case as
the one in Fig. 1.48. In this area, the surface lines are obscured but there are many neutrophils
embedded in the epithelium. The nuclei are dark but not particularly enlarged. Compare their
sizes with those of the inflammatory cell nuclei.
Figure 1-50. Barrett esophagus, indefinite for dysplasia. This is from the same case as that
seen in Figs. 1.48 and 1.49. The specimen is tangentially embedded and there are apparent
surface hyperchromatic nuclei, but it is not clear if in fact the surface is well represented.
Figure 1-51. Barrett esophagus, indefinite for dysplasia. Although the lines are obscured in this
zone, the adjoining areas seem to have preservation of cell polarity. The nuclei are enlarged but
not crisply distinct from nuclei that appear unremarkable.
Low-Grade Dysplasia
Low-grade dysplasia should be clearly neoplastic (adenoma-like) and should involve
the surface epithelium. It is important not to overdiagnose it because current guidelines
endorse mucosal ablation for low-grade dysplasia and mucosal ablation confers a risk
for stricture formation even in skilled hands. In prospectively evaluated patients the
incidence of low-grade dysplasia should be on the order of 2% to 3% but not more than
5%.33
The nuclei in the cells of low-grade dysplasia are larger than those of normal
Barrett mucosa, and generally, there is little inflammation in samples confidently
diagnosed with low-grade dysplasia. A helpful clue that low-grade dysplasia is present
is that one observes an abrupt transition between the dysplastic zone and adjoining
zones that are clearly not dysplastic. The surface lines that help to confirm
nondysplastic Barrett mucosa are effaced. Classic examples of low-grade dysplasia
with intestinal differentiation can resemble colorectal tubular adenomas. When this
happens, it is important to consider the possibility of a sample switch with an actual
colorectal adenoma before reporting the case. Examples of low-grade dysplasia appear
in Figs. 1.52–1.61. In general, low-grade dysplasia should demonstrate loss of the four
lines (an overall indication of altered cell architecture and cell polarity) but not loss of
nuclear polarity. The long axes of the nuclei should remain more or less perpendicular
to the basement membrane. This general concept is further discussed and illustrated in
“Colon” chapter with the construct of well aligned rows of nuclei. There is some
subjectivity in differentiating low-grade dysplasia from high-grade dysplasia, but this is
less important than it was in the past because all dysplasia are currently managed by
endoscopic ablation, although gastroenterology societies do suggest peer review of
dysplasia cases before mucosal ablation is performed.10,12
Staining for TP53 is sometimes useful in confirming an interpretation of low-grade
dysplasia—a few darkly stained nuclei on the surface can be identified and the basal
glands are labeled. In contrast, alpha-methylacyl-CoA racemase and other markers have
not proven useful, at least in our hands.
Most dysplasia cases show intestinal differentiation in that the epithelium is
stratified and punctuated by goblet cells in a fashion similar to the appearance of
colorectal adenomas, but not all do.34 On the order of 10% of low-grade dysplasia
cases can display gastric-type differentiation, which can manifest either as an
appearance similar to that of gastric pyloric gland adenoma (see “Stomach” chapter) or
a lesion that resembles a gastric foveolar-type adenoma. In such lesions in Barrett
mucosa the background may or may not contain goblet cells but the surface shows
dysplastic-appearing nuclei coated by an apical mucin cap akin to that seen in gastric
foveolar epithelium, except that the lines are absent. This type of dysplasia is
characterized by smaller nuclei than those in intestinal-type dysplasia, but they are
typically slightly hyperchromatic. Examples of this pattern of dysplasia are seen in Figs.
1.62–1.65. Unfortunately, immunolabeling for TP53 is not particularly helpful.
Figure 1-52. Low-grade dysplasia. This case is easy! This lesion is adenoma-like. The surface
is involved, and the nuclei at the surface appear similar to those in the glands. The lines are
completely obscured throughout the process and most of the nuclei are lined up with their long
axes oriented perpendicular to the basement membrane. With a case such as this, some
observers might prefer a diagnosis of high-grade dysplasia, but, importantly, presumably all
observers would regard this process as dysplastic and endoscopic treatment would be offered.
Figure 1-53. Low-grade dysplasia. This is a high-magnification image of the lesion depicted in
Fig. 1.52. Overall, the nuclei are oriented perpendicular to the basement membranes.
Figure 1-54. Low-grade dysplasia. This is a P53 stain from the lesion depicted in Figs. 1.52 and
1.53. It shows a wild-type pattern and was not particularly helpful in confirming the
interpretation.
Figure 1-55. Low-grade dysplasia. Note the abrupt demarcation between the low-grade
dysplasia and the nondysplastic mucosa. Note also that the lines are obscured and most of the
nuclei are oriented in an alignment that is perpendicular to the basement membrane. This latter
alignment is lost in high-grade dysplasia.
Figure 1-56. Low-grade dysplasia. A sharp demarcation is present between cardiac-type and
Barrett mucosa at the left of the image versus the dysplasia at the right.
Figure 1-57. Low-grade dysplasia. This field shows that, despite the stratification and loss of
the overall cellular polarity (loss of the lines), the nuclear polarity is maintained with the
elongated nuclei aligned perpendicularly to the basement membrane.
Figure 1-58. Low-grade dysplasia. This high magnification intends to show the alignment of the
nuclei with respect to the basement membrane.
Figure 1-59. Low-grade dysplasia. This image shows areas with sharp demarcations between
foci of low-grade dysplasia versus nondysplastic epithelium.
Figure 1-60. Low-grade dysplasia. The arrow indicates a zone of demarcation between
dysplastic and nondysplastic epithelium. Note that the dysplastic nuclei lack nucleoli in most
cells.
Figure 1-61. Low-grade dysplasia. This very-high-magnification image shows the nuclear
features of low-grade dysplasia.
Figure 1-62. Low-grade dysplasia. Rare examples of low-grade dysplasia show gastric foveolar
differentiation. The lines are obscured and the nuclei are hyperchromatic, but there are no
goblet cells in the area of dysplasia in contrast to the situation in Figs. 1.52–1.61. This pattern of
dysplasia can be encountered in a background of Barrett mucosa with goblet cells, but the
dysplasia itself shows gastric foveolar differentiation.
Figure 1-63. Low-grade dysplasia. This is a high-magnification image of the foveolar-type
dysplasia seen in Fig. 1.62. Note that the nuclei themselves appear similar to those in the
intestinal-type dysplasia seen in Figs. 1.60 and 1.61.
Figure 1-64. Foveolar pattern dysplasia. This case is a bit controversial. It could be interpreted
as low-grade dysplasia based on the disorderly alignment of the cells, but some cells have lost
their relationship to the membrane (a feature of high-grade dysplasia). This was interpreted as
low-grade dysplasia because the nuclei are not particularly enlarged. There are no goblet cells
in this area.
Figure 1-65. Foveolar pattern dysplasia. This is a high-magnification image of the lesion seen in
Fig. 1.64.
High-Grade Dysplasia
Generally, high-grade dysplasia is not difficult to recognize and is difficult to overlook.
At low magnification the area appears hyperchromatic and stands out from any
nondysplastic mucosa in the sample. It is generally not particularly inflamed, but even
examples showing inflammation appear extremely hyperchromatic at low magnification.
The alterations usually can be detected in the surface epithelium. Prominent nucleoli are
not a usual feature of high-grade dysplasia, but there are exceptions. In most cases, there
is still plenty of lamina propria between the glands. Figs. 1.66 and 1.67 show a
characteristic example of high-grade dysplasia with all the key features. Although there
are a few neutrophils (indicated), the nuclear hyperchromasia is in excess of that which
can be explained by a reparative process. Furthermore, there are often prominent
nucleoli in a reactive process and the nuclei shown are quite dense appearing. Many
nuclei both in the pits and at the surface have completely lost their relationship to the
basement membrane and have rounded up and are arranged in a jumbled configuration.
A similar lesion is shown in Figs. 1.68 and 1.69. Nuclear hyperchromasia is the key
finding, although the glands are crowded in this example as well. Some examples of
high-grade dysplasia feature markedly enlarged nuclei, sometimes in the absence of
glandular crowding, such as the case shown in Figs. 1.70 and 1.71.
Figure 1-66. High-grade dysplasia. In a case like this, finding acute inflammation (arrow) need
not detract from the diagnosis. In this case, the degree of nuclear hyperchromasia is in excess
of that which can be attributed to inflammation. Note that the nuclei at the surface are
hyperchromatic and many have lost their relationship. There are also many nuclei in the glands
that are hyperchromatic and have rounded up and lost their alignment with the basement
membrane.
Figure 1-67. High-grade dysplasia. This is a high-magnification image of the lesion seen in Fig.
1.66. The rounded hyperchromatic nuclei are the key feature. Note that nucleoli are not a
prominent feature.
Figure 1-68. High-grade dysplasia. The key finding is striking nuclear hyperchromasia.
Figure 1-69. High-grade dysplasia. This is a P53 immunostain. This strong diffuse nuclear
labeling pattern is characteristic of high-grade dysplasia. Some examples of low-grade
dysplasia show strong labeling, but usually just a few surface cells are labeled. In this example,
the surface nuclei are strongly reactive.
Most cases of high-grade dysplasia show some degree of nuclear elongation and
stratification akin to the features in colorectal adenomas. A subset of cases shows an
unusual pattern of small tubules, each lined by a monolayer of hyperchromatic nuclei
(Figs. 1.72 and 1.73). Because the monolayer appearance is unusual and lacks nuclear
stratification, it has been referred to as a “nonadenomatous” form of high-grade
dysplasia by some,35 but others have considered this pattern as evidence for gastric
differentiation in high-grade dysplasia.36,37 None of this has any effect on management,
but such cases are still part of high-grade dysplasia.
Figure 1-70. High-grade dysplasia. Even though there is no glandular crowding, the giant
hyperchromatic nuclei in some of the glands and on the surface at the left part of the image are
sufficient for a high-grade dysplasia diagnosis.
Figure 1-71. High-grade dysplasia. This is a very-high-magnification image of the lesion
depicted in Fig. 1.70. The nuclei are extremely hyperchromatic. Compare their sizes (including
the smaller ones) to the sizes of the lamina propria inflammatory cells.
Figure 1-72. High-grade dysplasia. Although the surface shows loss of nuclear polarity and
somewhat stratified nuclei, the arrow indicates some deep glands that also show high-grade
dysplasia in a pattern consisting of small tubules each lined by a monolayer of tiny
hyperchromatic nuclei that are not elongated like those of classic dysplasia. Because of the
lack of nuclear stratification, some colleagues have referred to this pattern as
“nonadenomatous” and others have regarded it as a form of gastric-type differentiation. The
main thing is to be aware that some examples of high-grade dysplasia lack nuclear
stratification.
Figure 1-73. High-grade dysplasia. This is a high-magnification image of the deep dysplastic
glands seen in Fig. 1.72.
The second: If there is pagetoid extension of single adenocarcinoma cells into the
squamous epithelium (Figs. 1.101 and 1.102), there is invariably an associated deeply
invasive underlying carcinoma, but this pattern is uncommon in biopsies.42,46
Most adenocarcinomas that have invaded beyond the lamina propria are
straightforward to diagnose and not subtle. They can have mucinous and signet cell
patterns akin to those in gastric carcinoma, and some are poorly differentiated and can
require immunolabeling to be classified (Figs. 1.103–1.105). At present, HER2 testing
is added for esophageal adenocarcinomas and the scheme for scoring is the same as that
for gastric carcinomas and addressed in detail in “Stomach” chapter.
Figure 1-96. Intramucosal carcinoma. This lesion has features of high-grade dysplasia with
hyperchromatic nuclei, but some of the glands bud off from larger ones, some have luminal
necrosis, and some grow parallel to the surface. Cases such as this are often interpreted as
high-grade dysplasia, which is acceptable for the purposes of modern treatments, as lesions
such as this are unlikely to metastasize.
Figure 1-97. Intramucosal carcinoma. This is a high-magnification image of the lesion seen in
Fig. 1.96. It is the complex architecture of the process that merits an interpretation of
intramucosal carcinoma rather than high-grade dysplasia. The glands do not form individual
tubules but instead interanastomose.
Figure 1-98. Intramucosal carcinoma. This is another high-magnification image of the lesion
shown in Figs. 1.96 and 1.97 intended to show the luminal necrosis at the lower right and the
abnormal complex architecture of the glands.
Figure 1-99. Intramucosal carcinoma. This is a CDX2 stain from the case seen in Figs.
1.96–1.98. There was no reason to perform this labeling—the laboratory that handled the
sample was in the habit of adding CDX2 labeling to all esophageal biopsies. The strong nuclear
labeling certainly supports that the lesion displays intestinal differentiation.
Figure 1-100. Adenocarcinoma. This adenocarcinoma is accompanied by desmoplasia
(scarring in response to the lesion). This finding on a biopsy sample suggests that there may
be deeper invasion than into the lamina propria.
Figure 1-101. Adenocarcinoma. In this case, there is pagetoid extension of single
adenocarcinoma cells into accompanying squamous mucosa in the sample. This finding
suggests that a deeply invasive carcinoma is present and is an ominous sign.
Figure 1-102. Adenocarcinoma. This is PAS/AB stain from the case seen in Fig. 1.101. The
PAS highlights glycogen in the superficial squamous epithelium (the basal cells lack glycogen),
but the adenocarcinoma cells contain bluish alcian blue–reactive mucin.
Figure 1-103. Poorly differentiated carcinoma associated with columnar epithelium in the
esophagus. This lesion has a suggestion of gland formation (adenocarcinoma), but it is difficult
to assure that it is not a squamous cell carcinoma.
Figure 1-104. Poorly differentiated carcinoma. This is a high-magnification image of the lesion
seen in Fig. 1.103. The cells are arranged in sheets and some have prominent nucleoli.
Figure 1-105. Poorly differentiated carcinoma. This strongly reactive BEREP4 stain supports an
interpretation of poorly differentiated adenocarcinoma. This staining was performed to
determine if HER2 testing should be done. HER2 testing is discussed in the “Stomach”
chapter, but the same criteria are used for gastric and esophageal adenocarcinomas.
Estimated
Likelihood of
Dysplasia Grade Further Documentation Follow-up, ACG/AGA Progression
High-grade or Expert confirmation. EMR Every 3 months for the 7%/year (HGD)
intramucosal recommended if biopsies are first year, every 6
carcinoma taken from an area of mucosal months for the second
irregularity coupled with RFA year, then annually
EMR, endoscopic mucosal resection; HGD, high-grade dysplasia; RFA, radiofrequency ablation.
Adapted from Shaheen NJ, Falk GW, Iyer PG, Gerson LB; American College of Gastroenterology. ACG
clinical guideline: diagnosis and management of Barrett’s esophagus. Am J Gastroenterol.
2016;111(1):30-50.
Endoscopic Mucosal Resection Specimens
EMR specimens are the more common samples received compared with submucosal
dissections and are more prone to interpretation issues because they are often simply
tossed into a jar of formalin, which causes them to curl. Ideally, they should be pinned
to a corkboard or wax board, but they are often too tiny to pin without ruining them. The
main thing is to have a way to orient them before they are sectioned. Ideally, they are
breadloafed and embedded so that each slice shows the surface mucosa and some deep
submucosa.44 It can be helpful to ink the deep (submucosal) margin, but if this is not
done, diathermy (cautery) is usually apparent to offer a clue about the location of the
margins.
There are two forms of artifact that are often encountered in EMRs, and once they
are understood, most EMRs can be readily assessed. The first is that the surface is often
iatrogenically damaged. This is because, to perform an EMR, often the surface mucosa
is sucked into a plastic cap after injecting the submucosa to create a polyplike lesion
that is then removed by polypectomy. As such, the surface is rubbed and sometimes the
most superficial epithelium is denuded. The second artifact is that, once the sample is
removed and tossed into fixative, the sample curls as noted earlier. This gives an
appearance that the lateral (mucosal) margin is a deep margin.
It is especially important when evaluating EMRs to be aware that, in patients with
Barrett mucosa, the muscularis mucosae becomes disorganized and duplicated or even
triplicated in response to cycles of damage and repair.2,51-53 This can easily result in
misinterpretation of lamina propria as submucosa and overstaging of the lesion as T1b
when the stage is in fact T1a. Remember that T1a indicates invasion into lamina propria
and T1b indicates invasion into submucosa.1 The space between the duplicated and
original muscularis mucosae is still lamina propria!!! A number of EMR samples,
some annotated to highlight the issue with the muscularis mucosae, are shown in Figs.
1.106–1.120.
For clinical treatment purposes, it does not matter which layer of the mucosa is
invaded, but clinical colleagues like to know a depth of invasion because they like a
number and a quantitation. They also like to know the status of the sample margins.
There are some formalized schemes for reporting this, but in our experience, it is safest
to simply describe the depth of invasion in words. For example, “the tumor invades into
the space between the initial and duplicated muscularis mucosae.” Table 1.2 shows the
various schemes that have been developed, and several authors have used “m1-m3” or
something similar to account for the various depths that a tumor can invade the mucosa.
We usually avoid the classifications with “m1-m3” or “m1-m4” because an oncologist
invariably misinterprets the “m” for “mucosal invasion” as the “M” for “metastasis” in
the staging manual. Argh.
Figure 1-106. Endoscopic mucosal resection (EMR) specimen. This image illustrates some
key issues in interpreting these samples. There is a lesion present in the upper right portion of
the specimen. Note that the right lesion is covered by squamous epithelium (buried or
pseudoregression pattern). The sample was not pinned to a corkboard such that it has curled,
but it is well embedded. A thick black line separates the muscularis mucosae from the
submucosa. The original muscularis mucosae is annotated, as is the duplicated muscularis
mucosae. The loose tissue between these two muscle layers in not submucosa, it is part of the
lamina propria! Note also that the surface epithelium appears damaged—this is probably from
the procedure to perform the EMR, in which a cap is applied to the lesion’s surface and suction
is applied. In this example, the muscularis mucosae layers are so thick that the endoscopist
barely obtained any submucosa.
Figure 1-107. Endoscopic mucosal resection (EMR) specimen. This is a higher-magnification
image of the right side of the lesion seen in Fig. 1.106. There is an intramucosal carcinoma that
invades the lamina propria. The squamous epithelium and the accompanying lamina propria
has curled such that the lateral margin masquerades as a deep margin. The actual deep
margin does not begin until the middle of the image, where a submucosal vessel is seen; the
muscularis mucosae is draped over it!
Figure 1-108. Endoscopic mucosal resection (EMR) specimen. This is a higher-magnification
image of the lesion seen in Figs. 1.106 and 1.107. The intramucosal carcinoma consists of
glands with luminal necrosis and angulated growth. Note that the surface at the left part of the
image has been altered during the EMR operation itself.
Figure 1-109. Endoscopic mucosal resection (EMR) specimen. In this example, the junction
between the mucosa and submucosa is delineated with a thick black line. It drapes over some
submucosal glands; their presence is proof that the cordoned off area is indeed submucosa!
The thick green line thus is the true deep (submucosal) margin, and the rest of the apparent
“deep” area is in fact lateral margin! The squamous epithelium itself has curled around to the
apparent deep margin. This is an artifact of tissue retraction associated with diathermy
(cautery) used in the EMR. In the top center, a few glands are arranged parallel to the surface in
the lamina propria (not the submucosa).
Figure 1-110. Endoscopic mucosal resection (EMR) specimen. This is a higher-magnification
image of the left side of the tissue seen in Fig. 1.109. The thick black line separates the
mucosa and submucosa. The thick muscularis mucosae has draped over the submucosal
gland to the left. The deep (submucosal) margin is indicated by the thick green line, but the
apparent deep tissue inked with black India ink is in fact a lateral margin consisting of curled
squamous epithelium and muscularis mucosae. There is an intramucosal carcinoma invading
into duplicated disorganized muscularis mucosae at the upper right of the field.
Figure 1-111. Endoscopic mucosal resection (EMR) specimen. This is a high-magnification
image of the lateral margin seen at the lower left of Fig. 1.110. Note the muscularis mucosae
curling over the submucosal gland at the upper right and painted with India ink at the bottom of
the image.
Figure 1-112. Endoscopic mucosal resection (EMR) specimen. This is a high-magnification
image of the lesion seen in Figs. 1.109–1.111. The intramucosal carcinoma has invaded into
the disorganized muscularis mucosae at the upper right of the image and is in the space
between the original muscularis mucosae and the superficial duplicated muscularis mucosae.
Note that there is no desmoplastic response to the invasion in this intramucosal carcinoma. In
this case, it is easy to see the extent of the lesion because this is an EMR sample but this field
should serve as a caution—were this a small biopsy, it would be easy to incorrectly assume
that there was submucosal invasion and that the smooth muscle at the bottom was muscularis
propria. However, a moment’s thought should rectify this idea because the muscularis propria
is practically never seen on mucosal biopsies of columnar esophagus.
Figure 1-113. Endoscopic mucosal resection (EMR) specimen. This example was pinned well
and has not curled much. As such, the bottom of the sample is submucosa and the thick band
of smooth muscle near the bottom is the muscularis mucosae. There is additional disorganized
smooth muscle above the thick muscularis mucosae.
Figure 1-114. Endoscopic mucosal resection (EMR) specimen. This is a higher-magnification
image of the EMR seen in Fig. 1.113. This particular EMR lacks dysplasia but shows only
reactive Barrett mucosa. However, it gives a good look at the thickened original (lower part of
field) muscularis mucosae and disorganized duplicated muscularis mucosae in the center of
the image above the original muscularis mucosae.
Figure 1-115. Endoscopic mucosal resection (EMR) specimen. This EMR is curled and
contains a carcinoma that has invaded the space between the original and duplicated
muscularis mucosae (T1a lesion). A thick black line separates the mucosa and submucosa. A
thick green line shows the deep (submucosal) margin, and thick blue lines show the lateral
margins. There is intramucosal carcinoma at the lateral (mucosal) margin on the left. This is
not at the deep margin! This is a common occurrence because endoscopists often resect
lesion by means of several side-by-side EMRs. In this case, much of the lesion is buried under
squamous epithelium but the endoscopist found it anyway.
Figure 1-116. Endoscopic mucosal resection (EMR) specimen. This EMR contains an
intramucosal carcinoma that has invaded nearly to the submucosa. A black line separates the
mucosa and submucosa. A thick green line shows the deep (submucosal) margin, and a blue
line shows the lateral margin. This T1a neoplasm is present at the lateral margin.
Figure 1-117. Endoscopic mucosal resection (EMR) specimen. This is a high-magnification
image of the involved lateral margin from the lesion seen in Fig. 1.115. The thick black line
shows the demarcation between the mucosa and submucosa. A submucosal gland is present
at the right side of the image beneath the black line. The green line shows the deep
(submucosal) margin, and the blue line shows the lateral (mucosal) margin. The muscularis
mucosae can be seen draping over a large submucosal vessel in the lower central part of the
field.
Figure 1-118. Endoscopic mucosal resection (EMR) specimen. This EMR shows a carcinoma
that invades the submucosa in the center of the image. In this case, it is worthwhile to measure
the depth of invasion starting from the bottom of the original muscularis mucosae.
Figure 1-119. Endoscopic mucosal resection (EMR) specimen. This is a higher-magnification
image of the lesion seen in Fig. 1.118. A bit of muscularis mucosae is seen in the center of the
image, and the carcinoma at the left is invading into the submucosa. Large submucosal
vessels are seen at the lower right.
Figure 1-120. Endoscopic mucosal resection (EMR) specimen. A carcinoma has entered the
submucosa. Note the large thick vessels in this space!
Dysplasia Recapitulation
Composite Fig. 1.121 summarizes the key issues of Barrett dysplasia grading:
Answer: This is a pattern seen after either injury from reflux or injury from
mucosal ablation in which squamous mucosa grows on top of columnar mucosa
(Barrett mucosa). There were early case reports of carcinomas appearing
beneath the squamous mucosa following ablation for dysplasia, but it is
currently believed that this concern was overblown and not an issue. In fact,
there is virtually always a surface lesion accompanying buried lesions
(pseudoregression lesions).38 In addition, using modern high-resolution
endoscopes, many endoscopists are able to see areas that harbor buried
lesions. We have already seen examples of these in Figs. 1.94, 1.106, 1.107
1.109, 1.110, 1.115, 1.116, 1.118, and 1.122. In some of these examples, the
endoscopist detected the lesions even though they were buried beneath
squamous epithelium and even labeled them “lesion” on the specimen jars. Of
course, she is a good endoscopist, but the point is that such lesions can be
found by careful gross (endoscopic) examination.
Figure 1-122. “Buried” Barrett-associated neoplasia/pseudoregression. This was once believed
to be an issue, but additional data do not support such a concern. In this example, an
intramucosal carcinoma, is seen buried under the squamous epithelium.
FAQ: What changes can we expect in samples obtained in patients who have
had mucosal ablation?
SQUAMOUS NEOPLASIA
RISK FACTORS
Esophageal squamous neoplasia is uncommon in the United States compared with other
regions of the world. Most patients with squamous carcinomas are men, and most are
adults at least in their fifties. In contrast to the demographics for adenocarcinomas,
which typically affect white men, squamous carcinoma predominates among African-
American men.57 Recent advances in improvement in detecting and treating
adenocarcinomas are not mirrored for squamous carcinoma.58
The incidence of squamous cell carcinoma is diminishing in comparison with
esophageal adenocarcinomas in the United States. In contrast, squamous carcinomas
have a high incidence in developing countries, e.g., in southern Africa and China. In
Southeast Asians, polymorphisms in ALDH, the gene that encodes aldehyde
dehydrogenase, are associated with esophageal squamous cell cancer.59,60 The effects of
these polymorphisms are synergistic with alcohol and smoking. ALDH polymorphisms
also result in accumulation in acetaldehyde, which causes flushing upon ingestion of
alcohol in about a third of East Asians (Chinese, Japanese, and Koreans).
Any factor that causes chronic irritation and inflammation of the esophageal mucosa
predisposes one to esophageal squamous cell carcinoma. Even skin disease affecting
the esophagus can initiate the development of dysplasia/carcinoma of the esophagus. An
example of this is lichen planus. However, substantial alcohol intake, especially in
combination with smoking, exponentially increases the risk of squamous cell carcinoma
(but not adenocarcinoma) and may account for the vast majority of squamous cell
carcinoma of the esophagus in the developed world. The combination of smoking and
alcohol abuse also results in an increased risk of head and neck cancer. Squamous cell
carcinoma of the esophagus is in fact discovered incidentally in 1% to 2% of patients
with head and neck cancers.61
Chronic esophageal irritation can also result from achalasia and esophageal
diverticula such that food is retained and decomposes, releasing various chemical
irritants. Frequent consumption of extremely hot beverages seems to increase the
incidence of squamous cell carcinoma. Lastly, persons who have ingested lye or other
caustic fluids require lifelong surveillance for the development of this cancer.62
Nonepidermolytic palmoplantar keratoderma (tylosis) is a rare autosomal dominant
disorder defined by RHBDF2 mutations on chromosome 17q2563 associated with
squamous cell carcinoma of the esophagus.64 Patients have hyperkeratosis of the palms
and soles and thickening of the oral mucosa. Although it confers up to a 95% risk of
squamous cell carcinoma of the esophagus by the age of 70 years, it is a rare syndrome.
Squamous cell carcinoma (but not adenocarcinoma) is associated with low
socioeconomic status, presumably a reflection of poor nutrition and other lifestyle
factors. However, deficiency syndromes associated with this cancer, such as the
Plummer-Vinson syndrome (dysphagia, iron-deficiency anemia, and esophageal webs),
are becoming uncommon in the developed world as overall nutrition improves. A role
for human papillomavirus (HPV) in the development of esophageal squamous cell
carcinoma is debatable, even though it is well-established in the anal canal, as
discussed in “Anus” chapter. Although HPV DNA detection rates are minimal (0% to
2%) in some studies from low-incidence areas,65,66 higher rates are reported in high-
incidence areas, such as China and Iran.67,68 However, one Mexican study, an area of
low tumor incidence, reported the presence of high-risk HPV DNA in 25% of
esophageal squamous cell carcinomas.69 The rate in the United States is about 10%.70
Figure 1-123. Low-grade squamous dysplasia. These lesions are subtle and often difficult to
differentiate from reparative changes. The nuclei are hyperchromatic, and there may be
mitoses and apoptotic nuclei as seen in the left part of the image. The epithelial changes are
restricted to the lower half of the epithelium. In the setting of prominent inflammation, it can be
impossible to determine if epithelial changes are reactive or dysplastic. Note that nucleoli are
not prominent.
Figure 1-124. Low-grade squamous dysplasia. This case is difficult, but there are no
inflammatory features to explain the findings and there are a few apoptotic bodies in the
proliferation, which occupies just under half of the epithelial thickness.
Figure 1-125. Low-grade squamous dysplasia. This is a high-magnification image of the case
seen in Fig. 1.123.
Figure 1-126. Low-grade squamous dysplasia. The nuclei in the basal portion of the epithelium
are markedly hyperchromatic.
Figure 1-127. Low-grade squamous dysplasia. The basal zone is thickened, and the nuclei are
enlarged and hyperchromatic.
Figure 1-128. Low-grade squamous dysplasia. High magnification shows scattered mitoses
and apoptotic bodies.
Figure 1-129. Low-grade squamous dysplasia. Note the marked nuclear hyperchromasia in the
center of the image. There is no inflammatory process to explain the findings.
Figure 1-130. Low-grade squamous dysplasia. This is a high-magnification image of the
process seen in Fig. 1.129.
Figure 1-131. High-grade squamous dysplasia. Despite the inflammation, the epithelial changes
are in excess of those attributable to inflammation. In Japan, lesions such as this would be
regarded as intramucosal carcinomas.
Figure 1-132. High-grade squamous dysplasia. The keratinization is concerning for invasion.
Figure 1-133. Lichenoid esophagitis with marked reactive epithelial changes. The findings are
similar to those of low-grade dysplasia, but the striking inflammatory process can explain them.
Patients with lichen planus of the esophagus and related lesions are at risk for squamous cell
carcinomas and are monitored accordingly, so if these changes are in fact dysplastic and
interpreted as reactive, this will not end patient surveillance.
Figure 1-134. Lichenoid esophagitis with marked reactive epithelial changes. The prominent
nucleoli suggest a reactive process.
Figure 1-135. Pill esophagitis with striking reactive changes. In the context of the exudate and
embedded pill material, these changes are best regarded as reparative.
Figure 1-136. Pill esophagitis with striking reactive changes. This is a high-magnification image
of the lesion seen in Fig. 1.135. Note the prominent intracellular edema at the lower right. The
edema causes the intercellular bridges to appear prominent.
Epidermoid Metaplasia
A peculiar pattern of hyperkeratosis and hypergranulosis, which can be termed
“esophageal leukoplakia” (because the endoscopist notes white plaques), but more
accurately “epidermoid metaplasia,” can be encountered as an isolated finding, but it
can also be seen associated with samples showing squamous dysplasia and
carcinoma.72,73 We suspect that this is a precursor lesion. There is also some more
scientific rather than histologic evidence that epidermoid metaplasia is a precursor
lesion; we have noted that molecular alterations detected by next-generation sequencing
mirror those in the associated dysplasias and carcinomas.74
Epidermoid metaplasia is easy to overlook unless one is in the know because it
appears normal at first glance except that it is metaplastic. Unfortunately, the magnitude
of the risk for associated squamous cell dysplasia and carcinoma is not known. The
subtle finding to be sought is simply the presence of a granular layer in esophageal
squamous epithelium, which is not normal for the esophagus but perfectly normal in
skin! Examples of epidermoid metaplasia, one of which is associated with neoplasia,
are shown in Figs. 1.137–1.142.
Figure 1-137. Epidermoid metaplasia. This sample appears rather unremarkable at first glance
but note the granular layer at the left. Note also that the lesion is sharply demarcated from the
uninvolved squamous epithelium.
Figure 1-138. Epidermoid metaplasia. This is a high-magnification image of the lesion seen in
Fig. 1.137. It shows the sharp demarcation between the normal area and the zone with
epidermoid metaplasia.
Figure 1-139. Epidermoid metaplasia. Note the granular layer and the surface hyperkeratosis.
Figure 1-140. Epidermoid metaplasia. This example is from a resection specimen—the
indication for resection was squamous cell carcinoma and there was extensive epidermoid
metaplasia in the adjoining tissue.
Figure 1-141. Squamous cell carcinoma associated with epidermoid metaplasia. This
squamous cell carcinoma was associated with epidermoid metaplasia.
Figure 1-142. Squamous cell carcinoma associated with epidermoid metaplasia. This is a high-
magnification image of the carcinoma seen in Fig. 1.141.
Figure 1-143. Epidermoid metaplasia. This is a tricky case. There is epidermoid metaplasia and
apparently dysplasia in the lower half of the mucosa. But, wait, there is more! The patient was
taking a taxane medication for a separate carcinoma (breast), and in fact there is taxane effect
(note the ring mitoses) in association with epidermoid metaplasia.
Figure 1-144. Taxane effect. In this tangentially embedded focus, it is a real mimicker of
dysplasia but note the mitotic arrest.
Figure 1-145. Taxane effect. This field is from the same biopsy as the images seen in Figs.
1.143 and 1.144. The mitotic arrest (ring mitoses) is apparent.
Figure 1-146. Esophageal squamous cell carcinoma. Squamous cell carcinoma of the
esophagus has the same appearance as it does elsewhere in the body with abnormally
keratinized overtly malignant cells.
However, in the spirit of trickery and to introduce the near misses section of this
chapter, note that Figs. 1.143–1.145 show a lesion in which a medication effect (taxane
effect75) mimics dysplasia in a patient with esophageal epidermoid metaplasia! The
clue is the ring mitoses.
NEAR MISSES
Case 1
Figs. 1.152 and 1.153 were taken from the esophagus and diagnosed as adenocarcinoma
arising in association with Barrett mucosa with high-grade dysplasia. As a result, the
patient was referred for endoscopic treatment and the biopsies were rereviewed in
preparation for this.
Figure 1-159. High-grade neuroendocrine carcinoma, large cell type. The cells are nested and
some are large with large nucleoli.
Like well-differentiated NETs elsewhere, they can be graded by assessing the
mitotic activity (to the extent possible in small biopsies, which means that they cannot
really be graded) or by performing Ki-67 immunolabeling.76 As of the 2010 WHO
classification of gastrointestinal tumors, well-differentiated NETs were graded as G1 or
G2 and the rare lesions with well-differentiated morphology but a high Ki-67 labeling
index were included with high-grade neuroendocrine carcinoma, but this has sometimes
proved unsatisfactory (because there is different treatment of small cell carcinoma and
large cell neuroendocrine carcinoma). As such, the 2017 WHO Classification of
Tumours of Endocrine Organs, at least with good data for pancreatic lesions, has added
a new category, termed NET G3.78 This latter category encompasses neoplasms that
have an appearance like that of a well-differentiated neuroendocrine (carcinoid) tumor
but that have a high proliferation index when Ki-67 labeling is performed. Because this
latter category is rare and NET of the esophagus is rare as well, we have not
encountered such a lesion. To summarize for neuroendocrine tumor76,78:
On the other hand, classic neuroendocrine carcinomas (always grade 3) are high-
grade lesions and can be classified as “small cell” or “large cell” types.76,78 Both types
are aggressive lesions. Most arise in men (although they are rare), sometimes in
association with Barrett mucosa. These are rare tumors that have not been well studied,
but in one Chinese series of patients with resectable disease, the mean age was about 60
years and most patients (about 70%) were men. About 60% were of the small cell
rather than of the large cell type. Although nearly 70% were alive after a year, only a
third were alive at 3 years, even though the patients had resectable disease.79 Patients
with these carcinomas are often treated with platinum-based chemotherapy. Large cell
neuroendocrine carcinomas manifest an organoid pattern with solid nests or acinar
structures, whereas small cell carcinomas form solid sheets and nests and are composed
of cells with small dark nuclei and minimal cytoplasm or larger cells with more
cytoplasm. Adjoining zones of adenocarcinoma or squamous cell carcinoma may be
present (the latter may require p63/p40 or CK5/6 staining to detect). They express
keratins, synaptophysin, and chromogranin. Some observers also use CD56
immunolabeling, which is hampered by its lack of specificity. Both small cell and large
cell neuroendocrine carcinomas are G3 neoplasms:
Case 2
Figs. 1.165 and 1.166 are from an esophageal biopsy. A mass was seen at
endoscopy in the middle third of the esophagus. The sample was interpreted as
squamous cell carcinoma. Indeed, the lesion does appear to “drip,” but there is
something amiss for squamous cell carcinoma. Fig. 1.166 gives the clue in that there is
mucin in some of the cells. This is an adenosquamous carcinoma. However, it is not
horrible to miss this feature—both types (adenocarcinoma and squamous cell
carcinoma) are treated the same way, although HER2 testing might be added for
adenosquamous carcinoma. This is different from the situation in the prior case—
detecting a high-grade neuroendocrine carcinoma results in a different chemotherapy
regimen than that for adenocarcinoma.
Figure 1-166. Adenosquamous carcinoma of the esophagus. Note the mixture of squamous
cells and cells that contain mucin.
Figure 1-167. Thyroid papillary carcinoma extending directly into the esophagus. The
carcinoma has an odd appearance for an esophageal adenocarcinoma.
Figure 1-168. Thyroid papillary carcinoma extending directly into the esophagus. A few grooves
and intranuclear inclusions are present, but they are easier to see if you know that the patient
has a history of a large thyroid carcinoma!
Adenosquamous Carcinoma
Adenosquamous carcinoma arises only rarely in the esophagus. It consists of a mixture
of infiltrating squamous cell carcinoma and adenocarcinoma elements. Like squamous
cell carcinoma, this variant preferentially affects the middle third of the esophagus.
Adenosquamous carcinoma also seems to present at an earlier stage than pure
adenocarcinoma or squamous cell carcinoma.80 The squamous component tends to be
more conspicuous than the glandular areas with gradual transitions between the two.
Areas of accompanying Barrett mucosa may rarely be identified. The outcome is more
favorable than that associated with pure squamous or adenocarcinoma, presumably a
result of the smaller size and lower stage at presentation.80
Case 3
Figs. 1.167 and 1.168 are images taken from an esophageal biopsy in a 58-year-old
woman who complained of dysphagia. The sample was diagnosed as well-
differentiated adenocarcinoma, and the patient was referred to one of our institutions for
treatment. On review, it was noted that the nuclei appeared rather small compared with
those that usually form esophageal adenocarcinomas and also that nucleoli were not a
feature. This prompted extensive queries into the clinical history, and we learned that
the patient had a history of papillary thyroid carcinoma. Based on this history, we added
a thyroglobulin stain, which is seen in Fig. 1.169. In this case, the presence of unusual
features in the lesion prompted some digging in the patient’s records. Once we
considered thyroid carcinoma, the nuclear inclusions and grooves became apparent to
us, even though the sample was suboptimal.
Case 4
Fig. 1.172 shows a spindle cell lesion that was biopsied from the esophagus. On
immunolabeling, it was KIT reactive but CD34 nonreactive. A diagnosis of esophageal
GIST was made. The mitotic count was high in keeping with a high-risk lesion, and a
resection was performed.
Melanoma
Primary esophageal melanoma is rare, with only about 300 cases reported. It is
encountered in adults with a mean age of about 60 years.86-91 There is a male
predominance but no racial predominance. Most esophageal melanomas arise in the
distal esophagus. At endoscopy, most are polypoid and pigmented (about 85%). Imaging
studies show bulky polypoid masses that bulge intraluminally without associated
obstruction. Many examples are pigmented, an obvious diagnostic clue. The rest are
whitish and poorly marginated. Primary melanomas may display an in situ component.
When present, this finding is extremely useful in establishing the esophagus as the
primary site. The malignant cells are spindled to epithelioid with variable pigment and
prominent nucleoli, and sometimes prominent intranuclear pseudoinclusions can be
found.
The key differential diagnosis is with poorly differentiated carcinoma, which is far
more common, and also with high-grade lymphomas. Immunohistochemistry can be
critical in establishing the diagnosis of esophageal melanoma, as it is with melanomas
elsewhere. Useful antibodies include S100 protein, Sox 10, MART, HMB45, and Melan
A. Remember that melanomas are also reactive with CD117/KIT antibodies about 40%
of the time, so a panel approach is best; overall, melanomas are usually more
pleomorphic than GISTs. The host of masqueraders is usually unmasked using a panel
including lymphoid markers (among which is CD30 for anaplastic lymphoma) and
pankeratins. Sarcomas are rarely in the differential diagnosis, but, of course, spindled
melanomas may lack other melanoma markers aside from S100 protein and Sox 10.
Another spindle cell tumor that is strongly S100 protein reactive is cellular (benign)
schwannoma. The distinction is on cytologic grounds, with attention to nuclear
pleomorphism and large nucleoli, features of melanoma but not of cellular schwannoma.
These unusual primary esophageal melanomas have had a dismal prognosis. Based on
the literature, only rare patients, whose tumors present early, can be cured. However,
newer treatments exploiting PDL1/PD1 blockade may improve the situation, but there
are no outcome data available at this point for esophageal melanoma specifically. In one
case report a good tumor response was achieved with nivolumab.92
Figure 1-172. Esophageal melanoma with spindle cell features. This lesion displays the
monotonous cytologic features that are often seen in gastrointestinal stromal tumors.
Figure 1-173. Melanoma extending into the lamina propria of the esophagus. In this case, a
pigmented in situ component gives away the diagnosis.
Case 5
Fig. 1.174 shows a biopsy from a mass lesion of the distal esophagus. It was
submitted on a rush protocol on a Friday night by a surgeon eager to plan an operation
for Monday morning and was reported as poorly differentiated carcinoma. The surgeon
scheduled the patient for 7 AM the following Monday. The resection resulted in a sinking
feeling.
Case 6
Fig. 1.175 is from a biopsy taken from a 75-year-old man with dysphagia, and a
subtle early lesion was seen by the endoscopist. A diagnosis of intramucosal
adenocarcinoma was made. The next day, the endoscopist called and said that she was
confused because the patient’s symptoms were out of proportion to the histologic
diagnosis and to the results of endoscopic ultrasound performed at the time of the
endoscopic evaluation.
Figure 1-174. Diffuse large B cell lymphoma involving the esophagus. It is easy to consider a
poorly differentiated carcinoma in this location.
Figure 1-175. Esophageal intramucosal carcinoma with extensive associated inflammation. Be
sure to check the areas of exudate!
References
1. Amin M, Edge S, Greene F, et al. AJCC Cancer Staging Manual. 8th ed. Springer
International Publishing AG Switzerland; 2017.
2. Abraham SC, Krasinskas AM, Correa AM, et al. Duplication of the muscularis
mucosae in Barrett esophagus: an underrecognized feature and its implication for
staging of adenocarcinoma. Am J Surg Pathol. 2007;31(11):1719-1725.
3. Glickman JN, Chen YY, Wang HH, Antonioli DA, Odze RD. Phenotypic
characteristics of a distinctive multilayered epithelium suggests that it is a
precursor in the development of Barrett’s esophagus. Am J Surg Pathol.
2001;25(5):569-578.
4. Glickman JN, Spechler SJ, Souza RF, Lunsford T, Lee E, Odze RD. Multilayered
epithelium in mucosal biopsy specimens from the gastroesophageal junction region
is a histologic marker of gastroesophageal reflux disease. Am J Surg Pathol.
2009;33(6):818-825.
5. Lack EE, Worsham GF, Callihan MD, et al. Granular cell tumor: a
clinicopathologic study of 110 patients. J Surg Oncol. 1980;13(4):301-316.
6. Johnston J, Helwig EB. Granular cell tumors of the gastrointestinal tract and
perianal region: a study of 74 cases. Dig Dis Sci. 1981;26(9):807-816.
7. Ohmori T, Arita N, Uraga N, Tabei R, Tani M, Okamura H. Malignant granular cell
tumor of the esophagus. A case report with light and electron microscopic,
histochemical, and immunohistochemical study. Acta Pathol Jpn. 1987;37(5):775-
783.
8. Yoshizawa A, Ota H, Sakaguchi N, et al. Malignant granular cell tumor of the
esophagus. Virchows Arch. 2004;444(3):304-306.
9. Miettinen M, Sarlomo-Rikala M, Sobin LH, Lasota J. Esophageal stromal tumors:
a clinicopathologic, immunohistochemical, and molecular genetic study of 17 cases
and comparison with esophageal leiomyomas and leiomyosarcomas. Am J Surg
Pathol. 2000;24(2):211-222.
10. Fitzgerald RC, di Pietro M, Ragunath K, et al. British Society of Gastroenterology
guidelines on the diagnosis and management of Barrett’s oesophagus. Gut.
2014;63(1):7-42.
11. American Gastroenterological Association, Spechler SJ, Sharma P, Souza RF,
Inadomi JM, Shaheen NJ. American Gastroenterological Association medical
position statement on the management of Barrett’s esophagus. Gastroenterology.
2011;140(3):1084-1091.
12. Shaheen NJ, Falk GW, Iyer PG, Gerson LB; American College of
Gastroenterology. ACG clinical guideline: diagnosis and management of Barrett’s
esophagus. Am J Gastroenterol. 2016;111(1):30-50; quiz 51.
13. Epstein JA, Cosby H, Falk GW, et al. Columnar islands in Barrett’s esophagus: do
they impact Prague C&M criteria and dysplasia grade? J Gastroenterol Hepatol.
2017;32(9):1598-1603.
14. Aida J, Vieth M, Shepherd NA, et al. Is carcinoma in columnar-lined esophagus
always located adjacent to intestinal metaplasia?: a histopathologic assessment.
Am J Surg Pathol. 2015;39(2):188-196.
15. Chandrasoma P, Wijetunge S, DeMeester S, et al. Columnar-lined esophagus
without intestinal metaplasia has no proven risk of adenocarcinoma. Am J Surg
Pathol. 2012;36(1):1-7.
16. Smith J, Garcia A, Zhang R, DeMeester S, Vallone J, Chandrasoma P. Intestinal
metaplasia is present in most if not all patients who have undergone endoscopic
mucosal resection for esophageal adenocarcinoma. Am J Surg Pathol.
2016;40(4):537-543.
17. Salimian KJ, Waters KM, Eze O, et al. Definition of Barrett esophagus in the
United States: support for retention of a requirement for goblet cells. Am J Surg
Pathol. 2018;42(2):264-268.
18. Bennett C, Moayyedi P, Corley DA, et al. BOB CAT: a large-scale review and
Delphi consensus for management of Barrett’s esophagus with no dysplasia,
indefinite for, or low-grade dysplasia. Am J Gastroenterol. 2015;110(5):662-682;
quiz 683.
19. Shields HM, Rosenberg SJ, Zwas FR, Ransil BJ, Lembo AJ, Odze R. Prospective
evaluation of multilayered epithelium in Barrett’s esophagus. Am J Gastroenterol.
2001;96(12):3268-3273.
20. Takubo K, Honma N, Arai T. Multilayered epithelium in Barrett’s esophagus. Am J
Surg Pathol. 2001;25(11):1460-1461.
21. Jiang M, Li H, Zhang Y, et al. Transitional basal cells at the squamous-columnar
junction generate Barrett’s oesophagus. Nature. 2017;550(7677):529-533.
22. Sharma P, Dent J, Armstrong D, et al. The development and validation of an
endoscopic grading system for Barrett’s esophagus: the Prague C & M criteria.
Gastroenterology. 2006;131(5):1392-1399.
23. Panarelli NC, Yantiss RK. Do ancillary studies aid detection and classification of
Barrett esophagus? Am J Surg Pathol. 2016;40(8):e83-e93.
24. Ormsby AH, Goldblum JR, Rice TW, et al. Cytokeratin subsets can reliably
distinguish Barrett’s esophagus from intestinal metaplasia of the stomach. Hum
Pathol. 1999;30(3):288-294.
25. Ormsby AH, Vaezi MF, Richter JE, et al. Cytokeratin immunoreactivity patterns in
the diagnosis of short-segment Barrett’s esophagus. Gastroenterology.
2000;119(3):683-690.
26. Phillips RW, Frierson HF Jr, Moskaluk CA. Cdx2 as a marker of epithelial
intestinal differentiation in the esophagus. Am J Surg Pathol. 2003;27(11):1442-
1447.
27. Hahn HP, Blount PL, Ayub K, et al. Intestinal differentiation in metaplastic,
nongoblet columnar epithelium in the esophagus. Am J Surg Pathol.
2009;33(7):1006-1015.
28. Chu PG, Jiang Z, Weiss LM. Hepatocyte antigen as a marker of intestinal
metaplasia. Am J Surg Pathol. 2003;27(7):952-959.
29. Montgomery E, Bronner MP, Goldblum JR, et al. Reproducibility of the diagnosis
of dysplasia in Barrett esophagus: a reaffirmation. Hum Pathol. 2001;32(4):368-
378.
30. Reid BJ, Haggitt RC, Rubin CE, et al. Observer variation in the diagnosis of
dysplasia in Barrett’s esophagus. Hum Pathol. 1988;19(2):166-178.
31. Lomo LC, Blount PL, Sanchez CA, et al. Crypt dysplasia with surface maturation:
a clinical, pathologic, and molecular study of a Barrett’s esophagus cohort. Am J
Surg Pathol. 2006;30(4):423-435.
32. Riddell RH, Goldman H, Ransohoff DF, et al. Dysplasia in inflammatory bowel
disease: standardized classification with provisional clinical applications. Hum
Pathol. 1983;14(11):931-968.
33. Curvers WL, ten Kate FJ, Krishnadath KK, et al. Low-grade dysplasia in Barrett’s
esophagus: overdiagnosed and underestimated. Am J Gastroenterol.
2010;105(7):1523-1530.
34. Vieth M, Montgomery EA, Riddell RH. Observations of different patterns of
dysplasia in Barrett’s esophagus - a first step to harmonize grading. Cesk Patol.
2016;52(3):154-163.
35. Rucker-Schmidt RL, Sanchez CA, Blount PL, et al. Nonadenomatous dysplasia in
Barrett esophagus: a clinical, pathologic, and DNA content flow cytometric study.
Am J Surg Pathol. 2009;33(6):886-893.
36. Mahajan D, Bennett AE, Liu X, Bena J, Bronner MP. Grading of gastric foveolar-
type dysplasia in Barrett’s esophagus. Mod Pathol. 2010;23(1):1-11.
37. Patil DT, Bennett AE, Mahajan D, Bronner MP. Distinguishing Barrett gastric
foveolar dysplasia from reactive cardiac mucosa in gastroesophageal reflux
disease. Hum Pathol. 2013;44(6):1146-1153.
38. Bronner MP, Overholt BF, Taylor SL, et al. Squamous overgrowth is not a safety
concern for photodynamic therapy for Barrett’s esophagus with high-grade
dysplasia. Gastroenterology. 2009;136(1):56-64; quiz 351-352.
39. Vieth M, Riddell RH, Montgomery EA. High-grade dysplasia versus carcinoma:
east is east and west is west, but does it need to be that way? Am J Surg Pathol.
2014;38(11):1453-1456.
40. Coco DP, Goldblum JR, Hornick JL, et al. Interobserver variability in the
diagnosis of crypt dysplasia in Barrett esophagus. Am J Surg Pathol.
2011;35(1):45-54.
41. Voltaggio L, Montgomery EA. Diagnosis and management of Barrett-related
neoplasia in the modern era. Surgical Pathol Clin. 2017;10(4):781-800.
42. Zhu W, Appelman HD, Greenson JK, et al. A histologically defined subset of high-
grade dysplasia in Barrett mucosa is predictive of associated carcinoma. Am J
Clin Pathol. 2009;132(1):94-100.
43. Pech O, May A, Manner H, et al. Long-term efficacy and safety of endoscopic
resection for patients with mucosal adenocarcinoma of the esophagus.
Gastroenterology. 2014;146(3):652.e651-660.e651.
44. Greene CL, Worrell SG, Attwood SE, et al. Emerging concepts for the endoscopic
management of superficial esophageal adenocarcinoma. J Gastrointest Surg.
2016;20(4):851-860.
45. Manner H, Pech O, Heldmann Y, et al. The frequency of lymph node metastasis in
early-stage adenocarcinoma of the esophagus with incipient submucosal invasion
(pT1b sm1) depending on histological risk patterns. Surg Endosc.
2015;29(7):1888-1896.
46. Abraham SC, Wang H, Wang KK, Wu TT. Paget cells in the esophagus: assessment
of their histopathologic features and near-universal association with underlying
esophageal adenocarcinoma. Am J Surg Pathol. 2008;32(7):1068-1074.
47. van der Wel MJ, Jansen M, Vieth M, Meijer SL. What makes an expert Barrett’s
histopathologist? Adv Exp Med Biol. 2016;908:137-159.
48. Terheggen G, Horn EM, Vieth M, et al. A randomised trial of endoscopic
submucosal dissection versus endoscopic mucosal resection for early Barrett’s
neoplasia. Gut. 2017;66(5):783-793.
49. Shaheen NJ, Sharma P, Overholt BF, et al. Radiofrequency ablation in Barrett’s
esophagus with dysplasia. N Engl J Med. 2009;360(22):2277-2288.
50. Canto MI, Shin EJ, Khashab MA, et al. Safety and efficacy of carbon dioxide
cryotherapy for treatment of neoplastic Barrett’s esophagus. Endoscopy.
2015;47(7):591.
51. Estrella JS, Hofstetter WL, Correa AM, et al. Duplicated muscularis mucosae
invasion has similar risk of lymph node metastasis and recurrence-free survival as
intramucosal esophageal adenocarcinoma. Am J Surg Pathol. 2011;35(7):1045-
1053.
52. Kaneshiro DK, Post JC, Rybicki L, Rice TW, Goldblum JR. Clinical significance
of the duplicated muscularis mucosae in Barrett esophagus-related superficial
adenocarcinoma. Am J Surg Pathol. 2011;35(5):697-700.
53. Lewis JT, Wang KK, Abraham SC. Muscularis mucosae duplication and the
musculo-fibrous anomaly in endoscopic mucosal resections for Barrett esophagus:
implications for staging of adenocarcinoma. Am J Surg Pathol. 2008;32(4):566-
571.
54. Westerterp M, Koppert LB, Buskens CJ, et al. Outcome of surgical treatment for
early adenocarcinoma of the esophagus or gastro-esophageal junction. Virchows
Arch. 2005;446(5):497-504.
55. Vieth M, Stolte M. Pathology of early upper GI cancers. Best Pract Res Clin
Gastroenterol. 2005;19(6):857-869.
56. Kissiedu J, Thota PN, Gohel T, Lopez R, Gordon IO. Post-ablation lymphocytic
esophagitis in Barrett esophagus with high grade dysplasia or intramucosal
carcinoma. Mod Pathol. 2016;29(6):599-606.
57. Brown LM, Hoover R, Silverman D, et al. Excess incidence of squamous cell
esophageal cancer among US black men: role of social class and other risk factors.
Am J Epidemiol. 2001;153(2):114-122.
58. Njei B, McCarty TR, Birk JW. Trends in esophageal cancer survival in United
States adults from 1973 to 2009: a SEER database analysis. J Gastroenterol
Hepatol. 2016;31(6):1141-1146.
59. Brooks PJ, Enoch MA, Goldman D, Li TK, Yokoyama A. The alcohol flushing
response: an unrecognized risk factor for esophageal cancer from alcohol
consumption. PLoS Med. 2009;6(3):e50.
60. Cui R, Kamatani Y, Takahashi A, et al. Functional variants in ADH1B and ALDH2
coupled with alcohol and smoking synergistically enhance esophageal cancer risk.
Gastroenterology. 2009;137(5):1768-1775.
61. Erkal HS, Mendenhall WM, Amdur RJ, Villaret DB, Stringer SP. Synchronous and
metachronous squamous cell carcinomas of the head and neck mucosal sites. J Clin
Oncol. 2001;19(5):1358-1362.
62. Csikos M, Horvath O, Petri A, Petri I, Imre J. Late malignant transformation of
chronic corrosive oesophageal strictures. Langenbecks Arch Chir.
1985;365(4):231-238.
63. Blaydon DC, Etheridge SL, Risk JM, et al. RHBDF2 mutations are associated with
tylosis, a familial esophageal cancer syndrome. Am J Hum Genet. 2012;90(2):340-
346.
64. Ellis A, Risk JM, Maruthappu T, Kelsell DP. Tylosis with oesophageal cancer:
diagnosis, management and molecular mechanisms. Orphanet J Rare Dis.
2015;10:126.
65. Turner JR, Shen LH, Crum CP, Dean PJ, Odze RD. Low prevalence of human
papillomavirus infection in esophageal squamous cell carcinomas from North
America: analysis by a highly sensitive and specific polymerase chain reaction-
based approach. Hum Pathol. 1997;28(2):174-178.
66. Poljak M, Cerar A, Seme K. Human papillomavirus infection in esophageal
carcinomas: a study of 121 lesions using multiple broad-spectrum polymerase
chain reactions and literature review. Hum Pathol. 1998;29(3):266-271.
67. Chang F, Syrjanen S, Shen Q, et al. Human papillomavirus involvement in
esophageal carcinogenesis in the high-incidence area of China. A study of 700
cases by screening and type-specific in situ hybridization. Scand J Gastroenterol.
2000;35(2):123-130.
68. Farhadi M, Tahmasebi Z, Merat S, Kamangar F, Nasrollahzadeh D, Malekzadeh R.
Human papillomavirus in squamous cell carcinoma of esophagus in a high-risk
population. World J Gastroenterol. 2005;11(8):1200-1203.
69. Herrera-Goepfert R, Lizano M, Akiba S, Carrillo-Garcia A, Becker-D’Acosta M.
Human papilloma virus and esophageal carcinoma in a Latin-American region.
World J Gastroenterol. 2009;15(25):3142-3147.
70. Syrjanen K. Geographic origin is a significant determinant of human
papillomavirus prevalence in oesophageal squamous cell carcinoma: systematic
review and meta-analysis. Scand J Infect Dis. 2013;45(1):1-18.
71. Salaria SN, Abu Alfa AK, Cruise MW, Wood LD, Montgomery EA. Lichenoid
esophagitis: clinicopathologic overlap with established esophageal lichen planus.
Am J Surg Pathol. 2013;37(12):1889-1894.
72. Singhi AD, Arnold CA, Crowder CD, Lam-Himlin DM, Voltaggio L, Montgomery
EA. Esophageal leukoplakia or epidermoid metaplasia: a clinicopathological study
of 18 patients. Mod Pathol. 2014;27(1):38-43.
73. Taggart MW, Rashid A, Ross WA, Abraham SC. Oesophageal hyperkeratosis:
clinicopathological associations. Histopathology. 2013;63(4):463-473.
74. Singhi AD, Arnold CA, Lam-Himlin DM, et al. Targeted next-generation
sequencing supports epidermoid metaplasia of the esophagus as a precursor to
esophageal squamous neoplasia. Mod Pathol. 2017;30(11):1613-1621.
75. Daniels JA, Gibson MK, Xu L, et al. Gastrointestinal tract epithelial changes
associated with taxanes: marker of drug toxicity versus effect. Am J Surg Pathol.
2008;32(3):473-477.
76. Bosman F, Carneiro F, Hruban R, Theise N, eds. WHO Classification of Tumours
of the Digestive System. Lyon: IARC; 2010. Bosman F, Jaffee E, Lakhani S, Ohgaki
H, eds. World Health Organization Classification of Tumours.
77. Modlin IM, Sandor A. An analysis of 8305 cases of carcinoid tumors. Cancer.
1997;79(4):813-829.
78. Lloyd R, Osamura R, Kloppel G, Rosai J. WHO Classification of Tumours of
Endocrine Organs. Lyon: IARC Press; 2017.
79. Deng HY, Ni PZ, Wang YC, Wang WP, Chen LQ. Neuroendocrine carcinoma of the
esophagus: clinical characteristics and prognostic evaluation of 49 cases with
surgical resection. J Thorac Dis. 2016;8(6):1250-1256.
80. Yachida S, Nakanishi Y, Shimoda T, et al. Adenosquamous carcinoma of the
esophagus. Clinicopathologic study of 18 cases. Oncology. 2004;66(3):218-225.
81. Aulakh K, Chisholm C, Speights V. TTF-1 and napsin-A frequently positive in
esophageal and pulmonary adenocarcinomas: dispelling myths and offering new
insights. Mod Pathol. 2011;24(suppl 1):142A; (Abstract 596).
82. Antonescu CR, Busam KJ, Francone TD, et al. L576P KIT mutation in anal
melanomas correlates with KIT protein expression and is sensitive to specific
kinase inhibition. Int J Canc. 2007;121(2):257-264.
83. Carvajal RD, Antonescu CR, Wolchok JD, et al. KIT as a therapeutic target in
metastatic melanoma. JAMA. 2011;305(22):2327-2334.
84. Carvajal RD, Hamid O, Antonescu CR. Selecting patients for KIT inhibition in
melanoma. Meth Mol Biol. 2014;1102:137-162.
85. Carvajal RD, Lawrence DP, Weber JS, et al. Phase II study of nilotinib in
melanoma harboring KIT alterations following progression to prior KIT inhibition.
Clin Canc Res. 2015;21(10):2289-2296.
86. Gollub MJ, Prowda JC. Primary melanoma of the esophagus: radiologic and
clinical findings in six patients. Radiology. 1999;213(1):97-100.
87. Syrigos KN, Konstadoulakis MM, Ricaniades N, Leandros M, Karakousis CP.
Primary malignant melanoma of the esophagus: report of two cases and review of
the literature. In Vivo. 1999;13(5):421-422.
88. Takubo K, Kanda Y, Ishii M, et al. Primary malignant melanoma of the esophagus.
Hum Pathol. 1983;14(8):727-730.
89. Volpin E, Sauvanet A, Couvelard A, Belghiti J. Primary malignant melanoma of the
esophagus: a case report and review of the literature. Dis Esophagus.
2002;15(3):244-249.
90. Wayman J, Irving M, Russell N, Nicoll J, Raimes SA. Intraluminal radiotherapy
and Nd:YAG laser photoablation for primary malignant melanoma of the
esophagus. Gastrointest Endosc. 2004;59(7):927-929.
91. Yoo CC, Levine MS, McLarney JK, Lowry MA. Primary malignant melanoma of
the esophagus: radiographic findings in seven patients. Radiology.
1998;209(2):455-459.
92. Inadomi K, Kumagai H, Arita S, et al. Bi-cytopenia possibly induced by anti-PD-1
antibody for primary malignant melanoma of the esophagus: a case report.
Medicine. 2016;95(29):e4283.
93. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health
Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375-
2390.
STOMACH 2
CHAPTER OUTLINE
The Unremarkable Stomach
Polyps
□ Pancreatic Heterotopia
□ Hamartomatous Polyps and Syndromic Considerations
• Peutz-Jeghers Polyp
• Juvenile Polyp
• Cronkhite-Canada Syndrome Polyp
• PTEN Hamartoma Tumor Syndrome and Cowden Syndrome
Polyp
□ Epithelial Polyps
• Hyperplastic Polyp
• Fundic Gland Polyp
□ Adenomatous Polyps
• Gastric Adenoma, Intestinal Type
• Gastric Adenoma, Foveolar Type
• Pyloric Gland Adenoma
• Oxyntic Gland Polyp/Adenoma
Adenocarcinoma
□ Approach to the Biopsy
• Tumor Location and Staging: Esophageal Versus Gastric
□ Tumor Classification: Lauren, Ming, and WHO Morphologic
Variants
□ Risk Factors and Genetic Considerations
• Background Mucosa
• Environmental Risk Factors
• Familial Predisposition
□ Biomarker Testing 101
• Programed Death Receptor-1/Programed Death Ligand-1 (PD-
1/PD-L1)
• Human Epidermal Growth Factor Receptor 2 (HER2)
Well-Differentiated Neuroendocrine Tumors (Formerly “Carcinoid”)
□ Gastric Well-Differentiated Neuroendocrine Tumor, Type I
□ Gastric Well-Differentiated Neuroendocrine Tumor, Type II
□ Gastric Well-Differentiated Neuroendocrine Tumor, Type III
MALT Lymphoma
Mesenchymal Lesions
□ Inflammatory Fibroid Polyp
□ Gastrointestinal Stromal Tumor
□ Leiomyoma
□ Granular Cell Tumor
Near Miss
□ Metastatic Lobular Breast Carcinoma
□ Gastric Xanthoma
□ Gastritis Cystica Profunda
□ Inactive Chronic Gastritis Hides Tumors
The lamina propria between the pits and glands is normally devoid of inflammatory
cells and contains inconspicuous lymphovascular channels accessible for metastatic
spread of tumor cells. The muscularis mucosae is composed of a thin delicate layer of
smooth muscle cells, separating the mucosa from the underlying submucosa, which
contains abundant larger lymphatic and vascular structures, also readily able to
facilitate metastasis. The muscularis propria of the stomach is composed of three sets of
smooth muscle fibers: longitudinal, circular, and oblique. The entire organ is encased by
the mesothelial-derived serosa, which forms the boundary to the peritoneal space.
Staging of invasive tumors requires accurate identification of each of these layers.
POLYPS
Several features contribute to challenges in gastric polyp classification, even for the
skilled pathologist. For example, gastric polyps are far less common than colonic
polyps, resulting in a more recent and sparse body of literature; they show significant
histologic overlap with one another; and classification is influenced by features of the
background flat mucosa. Unlike polyps elsewhere in the tubular gastrointestinal (GI)
tract, which are frequently isolated findings, polyps of the stomach often arise in
association with an inflammatory backdrop or a polyposis syndrome. Thus, although
they may be troublesome to classify, careful attention to the background mucosa with a
deliberate effort toward an integrated interpretation will provide important information
about prognosis or risk for familial syndromes. Gastric polyps are found in 6% of upper
endoscopies and are seen as projections above the adjacent flat mucosa (Fig. 2.8).1
These proliferative lesions may arise from the epithelium (most common) or from other
compartments in the mucosa and submucosa. Table 2.1 lists the most common gastric
polyps by chief proliferative compartment and serves as an outline for this segment.
This section provides a diagnostic approach for epithelial and hamartomatous polyps;
mesenchymal lesions are discussed in a dedicated chapter (see “Mesenchymal Lesions”
chapter). An understanding of the normal regions and histologic compartments,
reviewed at the beginning of this chapter, will facilitate application of this approach.
Figure 2-8. Endoscopically, gastric polyps appear exophytic and are mucosal or submucosal
based. All gastric polyps require histologic diagnosis.
TABLE 2.1: Gastric Polyps: Categorized by Proliferative Compartment
Heterotopic Polyps
• Pancreatic acinar heterotopia
Hamartomatous Polyps
• Peutz-Jeghers polypa
• Juvenile polypa
• Cronkhite-Canada syndrome–associated polypa
• PTEN hamartoma tumor syndrome and Cowden syndrome–associated polypa
Mesenchymal
• Inflammatory fibroid polyp
• Gastrointestinal stromal tumor
• Leiomyoma
• Vascular lesions
• Granular cell tumor
PANCREATIC HETEROTOPIA
Heterotopic pancreatic tissue is an incidental finding most commonly seen in the wall of
the gastric antrum. Endoscopically, it can appear as a submucosal nodule with central
umbilication and surface erosion (Fig. 2.9). These are commonly biopsied with clinical
concern for a gastrointestinal stromal tumor (GIST), which is endoscopically similar in
appearance. However, because the bulk of these lesions are submucosal, superficial
biopsies may lack diagnostic material and rebiopsy may be necessary. When well
sampled or mucosally resected, these lesions consist of a variable admixture of benign
pancreatic acinar cells, islets, and ducts, the latter of which may even connect to the
mucosal surface for drainage in larger lesions (Figs. 2.10–2.14). These structures
attempt to recapitulate normal pancreatic tissue and maintain a lobular architecture but
may appear slightly disorganized. Although the pancreatic acinar cells may be sparse,
they are identified by their typical triangular shape, eccentric round nuclei, and abundant
eosinophilic to amphophilic granular cytoplasm. Importantly, there is no cytologic
atypia, infiltrative border, or desmoplastic stromal reaction. Adjacent or overlying
mucosa may show reactive changes or intestinal metaplasia (IM) owing to the digestive
secretions of the pancreatic acinar cells.
Figure 2-9. Pancreatic heterotopia. A submucosal nodule with a central umbilication is seen in
the gastric antrum.
Figure 2-10. Pancreatic heterotopia. Pancreatic heterotopia is found in the wall of the stomach.
This EMR specimen shows deep submucosal location as well as mucosal involvement. Note
the lobulated appearance of the glands.
Figure 2-11. Pancreatic heterotopia. This example contains a few lobules of pancreatic acini in
the submucosa. The surface epithelium shows mild erosive and reactive changes.
Figure 2-12. Pancreatic heterotopia. Most examples are straightforward with well-developed
pancreatic acinar lobules, as seen here. In challenging cases, the presence of a duct (arrow)
can be a helpful clue.
Figure 2-13. Pancreatic heterotopia. Some cases can resemble neuroendocrine tumors, but
careful examination for a population of pancreatic acinar cells with eosinophilic granular
cytoplasm (arrows) can steer one away from this pitfall. Trypsin or chymotrypsin immunostains
would also be reactive in pancreatic acinar cells.
Figure 2-14. Pancreatic heterotopia. A small pancreatic duct (arrow) is a helpful clue in
differentiating pancreatic heterotopia from pancreatic metaplasia and neuroendocrine tumor.
Figure 2-15. Pancreatic acinar metaplasia (PAM). These pancreatic acinar cells (arrow) are
admixed with normal antral glands and lack the lobulated organization of pancreatic heterotopia.
The acinar cells of either PAM or pancreatic heterotopia may secret enzymes causing adjacent
intestinal metaplasia (arrowheads).
Peutz-Jeghers Polyp
Peutz-Jeghers syndrome is characterized by hamartomatous polyps of the GI tract and
melanocytic mucocutaneous hyperpigmentation.4 Up to 25% of documented cases are
sporadic, but this condition is best known as an autosomal dominant inherited syndrome
with 80% of affected families harboring a germline mutation in the STK11/LKB1
gene.5,6 Patients with Peutz-Jeghers syndrome have a 93% cumulative lifetime risk for
cancer, including carcinomas of the GI tract, breast, ovary, and testis.7-9 In this context,
early recognition of the syndrome allows for appropriate screening and surveillance for
patients and family members. World Health Organization (WHO) criteria for the clinical
diagnosis of Peutz-Jeghers syndrome are:
Figure 2-20. Higher magnification of the previous figure. Disorganized small bundles and wisps
of smooth muscle are present in the lamina propria and stream at intersecting angles.
Figure 2-21. Gastric Peutz-Jeghers polyp. This polyp is not as well developed as previous
examples, but it was retrieved from a patient known to have Peutz-Jeghers syndrome. The
glands appear organized and lack dilation or architectural changes, but note the presence of
smooth muscle within the lamina propria (arrowheads).
Figure 2-22. Higher magnification of the previous figure. Small disorganized bundles and wisps
of smooth muscle expand the lamina propria and branch in different directions.
Figure 2-23. Gastric Peutz-Jeghers polyp. Wisps of intersecting smooth muscle surround
glands creating small lobules, a helpful clue in some examples.
PEARLS & PITFALLS
Although a low threshold for diagnosis of hamartomatous polyps is advocated,
caution is advised against using gastric polyps to fulfill any of the WHO criteria
requiring “histologically confirmed” Peutz-Jeghers polyps. In the stomach, the
histologic features are not reliable enough to differentiate syndromic polyps
from reactive lesions such as inflammatory/hyperplastic polyps, and the
implications are considerable. Instead, compose a descriptive sign out with an
explanatory note (see the following note). If the patient has prior GI polyps, it
may be worthwhile to review the histology.
Comment
There are no reliable histologic features to distinguish gastric hamartomatous polyps
from reactive lesions (i.e., gastric inflammatory/hyperplastic polyps) or to reliably
differentiate subtypes of hamartomatous polyps. Nevertheless, the current specimen
contains some features that suggest hamartomatous development, such as cystically
dilated irregular glands and admixed smooth muscle. Syndromes involving
hamartomatous GI polyps (e.g., Peutz-Jeghers syndrome, juvenile polyposis syndrome,
Cowden syndrome) should be ruled out clinically. In addition, bidirectional endoscopy
is advised, with biopsy of any polypoid lesions in either the upper or lower GI tract, as
polyps found outside the stomach are more likely to retain pathognomonic features.
FAQ: I have a patient with single classic Peutz-Jeghers polyp, but this patient
has no other polyps or family history of Peutz-Jeghers syndrome. What does
this mean?
Juvenile Polyp
Juvenile polyposis syndrome, the most common of the hamartomatous polyposis
syndromes, affects one in 100,000. The syndrome is largely sporadic but can be
inherited as autosomal dominant familial syndrome (30%).15 Both inherited and
sporadic forms share similar genetics, with germline mutations in SMAD4 (also known
as DPC4) (15%) and the related gene BMPR1A (25%), whereas ENG is associated
with early childhood presentation.16,17 These genetic changes cause a disruption in the
transforming growth factor beta signal transduction pathway and result in an increased
risk of malignancy. The overall risk of GI malignancies in these patients is 55%, with
colorectal cancer presenting at an average age of 37 years.18,19
Figure 2-24. Gastric hamartomatous polyp. This gastric polyp was retrieved from a patient
without a known syndrome but shows brightly eosinophilic disorganized smooth muscle
bundles within the lamina propria suggestive of a hamartomatous polyp.
Figure 2-25. Gastric hamartomatous polyp, higher magnification of the previous figure. The
foveolar epithelium is separated by disorganized smooth muscle bundles that stream at
intersecting angles. Isolated sporadic Peutz-Jeghers polyps are rare but can occur. These
patients share the same cumulative lifetime risk of cancer as patients with Peutz-Jeghers
syndrome.
Figure 2-26. Juvenile polyp. This gastric polyp was retrieved from a patient known to have
juvenile polyposis syndrome. There are broad fingerlike projections with an excess of lamina
propria and inflammation. The glands are abnormally shaped and dilated.
Figure 2-27. Juvenile polyp. These polyps can resemble inflammatory polyps owing to their
edematous lamina propria, chronic inflammation, dilated glands, and surface erosion.
WHO criteria for the clinical diagnosis of juvenile polyposis syndrome are:
Figure 2-35. Cronkhite-Canada syndrome. Diffuse polyposis is seen in the stomach (pictured)
and throughout the upper and lower gastrointestinal tract.
Figure 2-36. Cronkhite-Canada syndrome, gastric antrum. Multiple biopsies of the stomach
show the diffuse nature of this disease. The mucosa is atrophic with abundant edematous
lamina propria, similar to that seen in juvenile polyps. The similarities between the antrum,
body, and fundus (see the next two figures) emphasize the diffuse process.
Figure 2-37. Cronkhite-Canada syndrome, gastric body. These biopsies from the gastric body
show cystically dilated glands and marked lamina propria edema. At first glance, they resemble
juvenile polyps. However, these changes are found in random samples of nonpolypoid mucosa.
One of the key diagnostic clues to Cronkhite-Canada syndrome is the involvement of
nonpolypoid mucosa.
Figure 2-38. Cronkhite-Canada syndrome, gastric fundus. As in the previous two figures,
multiple biopsies of the gastric fundus show diffuse changes involving all tissue fragments.
There is atrophy of the oxyntic glands, abundant lamina propria edema, and cystically dilated
glands with inspissated proteinaceous material.
Figure 2-39. Cronkhite-Canada syndrome. Polypoid and nonpolypoid mucosa is histologically
similar to juvenile polyps. The edematous lamina propria contains variable inflammation, and
focal areas may show erosion and reactive epithelial changes.
Figure 2-40. Cronkhite-Canada syndrome. There is an excess of edematous lamina propria
and dilated irregular glands. In isolation, the findings suggest a juvenile polyp or inflammatory
polyp. When these changes are found diffusely in both polypoid and nonpolypoid mucosa,
consider Cronkhite-Canada syndrome and look for the clinical triad of alopecia,
onychodystrophy (changes in nail color or quality), and hyperpigmentation.
Figure 2-41. Cronkhite-Canada syndrome. The mucosa contains cystically dilated glands and
edematous lamina propria with sparse inflammatory cells, similar in appearance to juvenile
polyps.
Figure 2-42. Cowden syndrome gastric polyp. Gastrointestinal lesions in PTEN hamartoma
tumor syndrome or Cowden syndrome include hamartomatous polyps, lipomas,
ganglioneuromas, and inflammatory polyps. This gastric hamartomatous polyp from a patient
with Cowden syndrome contains mucosal adipocytes (arrowhead ) and dilated, distorted
glands.
Figure 2-43. Cowden syndrome gastric polyp. This polyp shows lobules of glands, similar to
that of Peutz-Jeghers polyps, but the presence of adipocytes (arrowhead ) and neural tissue
(arrow) argue against this diagnosis. This mixed hamartomatous polyp is from a patient with
Cowden syndrome.
Figure 2-44. Cowden syndrome gastric polyp. The lamina propria between these distorted
gastric glands is myxoid and contains hamartomatous elements, such as adipose tissue.
EPITHELIAL POLYPS
Epithelial polyps are the most commonly encountered gastric polyps. These include
gastric hyperplastic polyps, fundic gland polyps (FGPs), and adenomatous polyps, all of
which are associated with distinctly different clinical contexts, as discussed later. Less
common epithelial lesions manifesting as polyps include NETs, discussed separately in
this chapter.
Hyperplastic Polyp
These benign epithelial proliferations are the second most common type of gastric
polyp, and, in contrast to the incidental colonic hyperplastic polyp, gastric hyperplastic
polyps are highly associated with background mucosal injury (85%), including
Helicobacter infection (25%), reactive/chemical gastropathy (21%), AMAG (12%),
and environmental metaplastic atrophic gastritis (8%).28 Other associated conditions
include mucosal ulcerations and erosions, ostomy sites, and gastroesophageal reflux
disease, which, notably, are also forms of mucosal injury. Although they may be found at
any age, gastric hyperplastic polyps occur more frequently with increasing age (mean
age 65 to 75 years) and are female predominant. The lesions are solitary in 75% of
cases and, owing to their foveolar origin, are found in all regions of the stomach with
fairly even distribution. When large, these lesions may cause gastric outlet obstruction
(Fig. 2.46); recurrence following polypectomy or endoscopic resection is common (up
to 50%).29,30
Figure 2-45. A flow diagram for interpreting unusual gastric polyps. The histologic overlap of
hyperplastic and syndromic gastric polyps can be a challenge during routine signout, but most
can be handled by applying the simple algorithmic approach illustrated in this figure.
Figure 2-46. Gastric hyperplastic polyp. This antral-based gastric polyp protrudes into the
lumen with a lobulated appearance. The entire lesion should be resected for histologic
evaluation, and a separate jar containing tandem biopsies of the surrounding flat mucosa
should also be submitted by the endoscopist to evaluate for background etiologic factors (e.g.,
H. pylori, AMAG).
Histologically, the polypoid mucosa often has a broad pedicle and shows elongated
and distorted pits lined by a single layer of foveolar epithelial cells. There is wide
histologic variability: the gastric pits and glands may show cystic dilation separated by
edematous lamina propria with mixed inflammatory cells (Fig. 2.47), or there may be
glandular crowding with gastric foveolar hyperplasia (Fig. 2.48). Surface erosion is
common and may be accompanied by reactive epithelial changes (Figs. 2.49 and 2.50).
Always ensure that a quick search for IM and dysplasia is performed (Figs. 2.51 and
2.52). Given the high association with surrounding mucosal inflammation and damage,
and the potential for neoplastic progression, pathologists should carefully review the
background flat mucosa when such material is available. Because not all endoscopists
are in the habit of doing so, this necessitates advising them to submit separate samples
of flat mucosa every time a gastric polyp is encountered. Recommended biopsy
protocols exist, the most widely accepted of which is the Sydney protocol, which
includes five mucosal samples: two each from greater and lesser curvatures (to include
both antrum and body) and one from incisura (transition zone).
Figure 2-47. Gastric hyperplastic polyp. Gastric hyperplastic polyps have a wide range of
appearances. This example shows a broad, rounded fingerlike projection at low magnification,
with tortuous distorted and dilated glands, abundant lamina propria, and surface erosion. The
features are similar to the juvenile polyp in Fig. 2.25, but this patient does not have a history of
juvenile polyposis syndrome.
Figure 2-48. Gastric hyperplastic polyp. The histologic spectrum ranges from stroma-rich, like
the previous example, to gland-rich, as seen here. This polyp contains tightly packed glands
and marked foveolar hyperplasia with elongated tortuous foveolar pits, similar to
chemical/reactive gastropathy. In this case, the gastric pits comprise about 80% of the mucosal
thickness (normal is up to 25% in the body/fundus and up to 50% in the antrum).
Figure 2-49. Reactive atypia in a gastric hyperplastic polyp. Epithelial cells may show reactive
changes, such as mild nuclear hyperchromasia and enlargement due to inflammation or
erosion.
Figure 2-50. Reactive atypia in a gastric hyperplastic polyp, higher magnification of the previous
figure. Reassuring features for reactive atypia are the presence of preserved nuclear to
cytoplasmic ratio, indistinct nucleoli, lack of nuclear crowding, and absence of an abrupt
transition from normal.
Figure 2-51. Low-grade dysplasia arising in a hyperplastic polyp. Most dysplasia can be
identified at low magnification by the presence of stark hyperchromasia and nuclear crowding.
An abrupt transition from normal to dysplastic (arrow) is a helpful clue. The nuclei in this
example remain elongated, and the glandular architecture is simple, supporting classification
as low-grade dysplasia.
Figure 2-52. High-grade dysplasia. Both glands and cells are crowded in this example. The
glands are back to back and architecturally complex. The cells are no longer elongated and
show loss of nuclear polarity. Prominent nucleoli and irregular nuclear contours are present.
Frequent mitoses and apoptoses are evident.
PEARLS & PITFALLS: The Background Flat Mucosa May Reveal the Etiology
of the Hyperplastic Polyp
Always search for an etiologic factor when encountering these reactive lesions.
Treatment of an underlying condition, such as eradication of Helicobacter
infection, can prevent further polyp formation and recurrence.
Comment
Gastric hyperplastic polyps are reactive lesions highly associated with mucosal injury
and inflammation. Should repeat endoscopy be performed, separately submitted
biopsies of the background flat mucosa to evaluate the gastric environment would be of
interest.
Despite their classification as a regenerative growth, large (>2 cm) polyps may
harbor IM or dysplasia and subsequent risk of malignant transformation (2% to
20%).28,31-33 Similar to other areas of the GI tract, gastric dysplasia is categorized as
negative for dysplasia, low-grade dysplasia (LGD), high-grade dysplasia (HGD), or
indefinite for dysplasia. The microscopic features also parallel those seen in other areas
of the GI tract with LGD showing epithelial changes of hyperchromasia, nuclear
elongation, and pseudostratification extending to the mucosal surface (Fig. 2.51). Other
features include nuclear atypia, prominent nucleoli, and increased mitoses and
apoptoses. High-grade epithelial dysplasia is characterized by greater nuclear
pleomorphism, anisonucleosis, loss of cell polarity, and architectural complexity with
back-to-back or cribriform gland formation (Fig. 2.52). These descriptive terms sound
quite similar to the words used to describe the conventional tubular adenoma, yet
gastric dysplasia remains slightly more challenging to interpret. Contributors to this
struggle include the array of gastric dysplastic lesions (e.g., gastric adenomas include
foveolar, intestinal, pyloric gland, and oxyntic gland types; see later discussion) and the
frequency of background gastric inflammatory changes, from which one must
differentiate reactive and dysplastic lesions. The abrupt histologic transition of
dysplasia remains the best practical tool in dysplasia assessment and is evident even at
low magnification.
Figure 2-53. Gastric antral prolapse. Gastric antral prolapse can produce polypoid areas, not to
be mistaken for hyperplastic polyps. The presence of tortuous gastric pits and exuberant
smooth muscle (arrows) streaming perpendicular to the luminal surface are features of antral
prolapse.
Figure 2-54. Gastric hyperplastic polyp. This example is small, rounded and contains an
excess of pale edematous lamina propria, variably distorted glands, and inflammation.
Compare this example with the juvenile polyp from Fig. 2.26. There are no reliable histologic
features to differentiate the two entities aside from the presence of background mucosal
damage (e.g., H. pylori, AMAG, gastritis), which favors a gastric hyperplastic/inflammatory
polyp.
F, female; FAP, familial adenomatous polyposis; FGP, fundic gland polyp; LGD, low-grade dysplasia; M,
male.
Figure 2-57. Gastric hyperplastic polyp. Lamina propria edema, mild foveolar hyperplasia, and
scattered chronic inflammatory cells can be seen in both hyperplastic and inflammatory polyps.
This small polyp arose in a backdrop of portal hypertensive gastropathy (not pictured) and
contains ectatic vessels in the lamina propria (arrowheads). Findings in the background flat
mucosa aid in classifying gastric polyps.
Histologically, these polyps consist of dilated cystic oxyntic glands with distorted
glandular architecture admixed with normal-appearing glands (Figs. 2.59 and 2.60).
Parietal cells balloon into the lumen with snoutlike protuberances, sometimes resulting
in exfoliated anucleate blebs with eosinophilic granules that clog the gland outlets (Figs.
2.61 and 2.62). Some cystic spaces may be lined by columnar epithelium as a result of
adjacent gastric pit dilation (Fig. 2.63), but most are lined by chief cells and parietal
cells. The findings, for practical purposes, are identical to that of PPI effect, and
distinction requires correlation with an endoscopic lesion (Fig. 2.64). Likewise, there
are no differentiating histologic features between syndromic and sporadic FGPs, but the
clinicopathologic features are distinct. For example, nearly all patients with FAP have
FGPs (12.5% to 88% of patients with FAP, depending on age at time of endoscopy) and
these polyps are more numerous than sporadic FGPs, often with hundreds or thousands
in a carpetlike distribution.37 FAP-associated FGPs also occur earlier than in the
sporadic setting (third decade vs. fifth to sixth decade) and can be found among the
pediatric population, which is exceptionally rare for sporadic FGPs. A key distinction
of FAP-associated FGPs is their association with low-grade epithelial dysplasia, which
is reported in up to half of cases (Figs. 2.65–2.69). Despite the high prevalence of
dysplasia in these lesions, the risk of malignant transformation is exceptionally low,
with only case reports in the literature.38,39 By comparison, dysplasia develops in less
than 1% of sporadic FGPs and has never been associated with progression to
carcinoma.34,40 Given the rarity of both HGD and carcinoma in either sporadic or FAP
settings, surveillance is unnecessary for LGD in an FGP and surgical resection is never
advised. Patients with FAP do require continued surveillance for other risk factors,
however, such as duodenal polyposis and periampullary adenomas and
adenocarcinomas.41 Upper endoscopy and biopsy of selected polyps is recommended
every 1 to 3 years, with closer screening warranted when a gastric adenoma is detected.
Figure 2-58. Fundic gland polyps (FGP). Multiple small sessile lesions (arrowheads) are seen
in the gastric body. Sporadic FGPs may be multiple but usually few in number. When
innumerable FGPs carpet the gastric corpus, consider a syndromic cause, such as familial
adenomatous polyposis syndrome, MutYH-associated polyposis, or gastric adenocarcinoma
and proximal polyposis syndrome.
Figure 2-59. Fundic gland polyps, scanning magnification. These polypoid tissue fragments
show dilated cystic glands with distorted architecture. Multiple polyps are frequently found,
although endoscopists typically only sample one or two.
Figure 2-60. Fundic gland polyp. These small round polyps are expanded by cystically dilated
oxyntic glands, and there is no significant increase in stroma. The surface epithelium is foveolar
and unremarkable.
Figure 2-61. Fundic gland polyp. The cystic spaces are dilated oxyntic glands and are lined by
pink parietal cells and blue chief cells. The parietal cells balloon into the lumen with snoutlike
protuberances, and the lumen contains exfoliated cells.
Figure 2-62. Fundic gland polyp. At high magnification, the parietal cells have brightly
eosinophilic granular cytoplasm that protrudes toward the lumen, similar to apocrine metaplasia
in the breast. The apical snouts produce anucleate granular blebs within the cystic space.
Figure 2-63. Fundic gland polyp. Some cystic spaces are partially lined by foveolar mucous
cells (arrowhead ), but this variation does not indicate a mixed polyp. The background dilated
oxyntic glands and apical snouting of parietal cells are characteristic for fundic gland polyp.
Figure 2-64. Proton pump inhibitor (PPI) effect. PPIs are associated with diffuse oxyntic gland
dilation and apical snouting of parietal cells, similar to that seen in fundic gland polyps.
Distinction between the two requires endoscopic correlation with a polypoid lesion or with
medication history.
Figure 2-65. Low-grade dysplasia in a fundic gland polyp. Low-grade dysplasia is best identified
as an area of eye-catching hyperchromasia at low magnification. An abrupt transition
(arrowhead ) between normal foveolar epithelium and an area of hyperchromasia is a helpful
clue.
Figure 2-66. Low-grade dysplasia in a fundic gland polyp. Dysplasia in FGPs occurs in half of
patients with FAP syndrome. Dysplasia can also be found in sporadic FGPs but is seen in
<1%. An abrupt transition (arrow) from nondysplastic to dysplastic epithelium can usually be
identified. The dysplastic foveolar epithelial cells show pseudostratification and nuclear
crowding, imparting an area of distinct hyperchromasia.
Figure 2-67. Low-grade dysplasia in a fundic gland polyp. Dysplasia affects the surface foveolar
cells overlying the dilated oxyntic glands and extends down the foveolar-lined gastric pits. An
abrupt surface transition (arrowhead ) from nondysplastic cells is present.
Figure 2-68. Low-grade dysplasia in a fundic gland polyp. Dysplasia affects the foveolar
epithelium overlying the fundic glands and shows cytologic features of pseudostratification,
nuclear crowding, increased mitotic activity, and apoptotic activity (arrowhead ).
Figure 2-69. Low- and high-grade dysplasia in a fundic gland polyp. A distinct change in
architecture and cytology is seen in the right side of the figure and can be arguably interpreted
as high-grade dysplasia, in contrast to the characteristic low-grade dysplasia seen on the left.
Dysplasia in fundic gland polyps, whether low or high grade, has negligible risk for malignant
transformation.
An exception in FAP that is discussed later in this chapter is the syndrome termed
gastric adenocarcinoma and proximal polyposis syndrome (GAPPS). GAPPS is a rare
subvariant of FAP characterized by many FGPs and gastric adenocarcinoma.
FAQ: What is the surveillance protocol for LGD in an FGP? Are there
differences in follow-up for LGD in sporadic versus FAP-associated FGPs?
ADENOMATOUS POLYPS
By convention, polypoid dysplasia in the stomach is designated “gastric adenoma,” and
the degree of dysplasia is graded similar to other parts of the GI tract: low grade, high
grade, or indefinite for dysplasia (Figs. 2.70–2.77). Similar to colon tubular adenomas,
a diagnosis of gastric adenoma implies at least LGD. Gastric adenomas come in several
varieties, including the intestinal type, foveolar type, pyloric gland adenoma (PGA), and
oxyntic gland polyp/adenoma (see Table 2.3). Our understanding of this area continues
to evolve, and much of the previous literature is difficult to interpret owing to several
factors: (1) Divergent histologic criteria between Japanese and Western practices;
Japanese pathologists, who contributed a significant bulk of the 20th century literature
on this subject, did not require invasion to diagnose “carcinoma,” whereas invasion is
an explicit criterion of Western practice.45 (2) Revision of nomenclature as a result of
this discrepancy; definitions of gastric epithelial neoplasia were harmonized in the
Vienna classification published in 2000, resulting in a system congruent with Western
understanding.46 (3) Subsequent ongoing revisions of polyp classification and
nomenclature; oxyntic gland polyp/adenoma was previously classified as “gastric
adenocarcinoma with chief cell differentiation.”47 (4) Lack of recognition or
underreporting of certain gastric adenomas; for example, in one study, PGA was the
third most common gastric polyp, yet only rare case reports were published on this
entity previously.48 (5) Histologic blends of “hybrid” polyps; these defy specific
classification and continue to muddle the literature. Ongoing study in this area will most
certainly yield new updates before a revised edition of this text. In the meantime, the
following section depicts our current understanding for each of the gastric adenomas.
Some bear risk for malignant progression, but each is remarkably different in prognosis
and clinicopathologic associations, discussed later. As with gastric hyperplastic polyps,
sampling the background flat mucosa is important to identify other risk factors (e.g., H.
pylori, IM) and associated conditions such as AMAG.
Figure 2-70. Reactive changes, negative for dysplasia. Areas of intestinal metaplasia
(arrowhead ) may be hyperchromatic at low power. However, this polyp shows a gradual
gradient toward the lighter mature surface. Surface maturation is a reassuring feature for
nondysplastic reactive changes.
Figure 2-71. Gastric adenoma, intestinal type (low-grade dysplasia implied). The designation as
“gastric adenoma” implies at least low-grade dysplasia. The hyperchromasia is striking at low
magnification and extends to the mucosal surface. Note the abrupt transition from
nondysplastic adjacent mucosa.
Figure 2-72. Gastric adenoma, intestinal type. LGD is dramatically hyperchromatic at low
magnification. The crowded cells extend to the mucosal surface, and there is an abrupt
transition from nondysplastic epithelium. Reactive changes can mimic LGD, and this
juxtaposition is a nice contrast, showing slight hyperchromasia but no nuclear crowding. Also
note the gradual color gradient in the reactive area versus the abrupt change in LGD.
Figure 2-73. Gastric adenoma, intestinal type. Involvement of the surface epithelium is a helpful
clue to dysplasia, but beware of tangential embedding. In this example, the area of LGD is
hyperchromatic and eye-catching but does not involve the overlying surface epithelium. Search
out areas of background intestinal metaplasia (arrowhead ) for comparison. The marked
contrast in hyperchromasia between the two areas supports LGD.
Figure 2-74. Low-grade dysplasia. Gastric adenoma, intestinal type (goblet cells not seen in this
field). Dysplasia in this example is conventional, similar to that seen in a colonic tubular
adenoma. The architecture is simple with well-formed glands, and the nuclei are elongated,
pseudostratified, and crowded. Mitotic activity (arrow) and apoptoses (arrowhead ) are
increased.
Figure 2-75. Gastric adenoma, intestinal type (goblet cells not seen in this field). The cells at the
top of this photo are starting to show more rounded nuclei (loss of nuclear polarity) instead of
elongated nuclei. The combination of architectural complexity, loss of nuclear polarity, and
increased cytologic atypia are criteria for high-grade dysplasia.
Figure 2-76. High-grade dysplasia and low-grade dysplasia in gastric adenoma, intestinal type.
A diagnosis of gastric adenoma implies low-grade dysplasia. When high-grade dysplasia is
seen, it is listed first. The cells in high-grade dysplasia show disorganization with loss of
nuclear polarity and marked nuclear atypia characterized by variability in both size and shape
as compared with neighboring cells. In contrast, the cells of low-grade dysplasia retain nuclear
polarity and appear organized and relatively uniform, despite the hyperchromasia and nuclear
crowding.
Figure 2-77. High-grade dysplasia in gastric adenoma, intestinal type (goblet cells not seen in
this field). These cells have an increased nuclear to cytoplasmic ratio, appear disorganized
owing to loss of nuclear polarity, and show marked nuclear variability in size and shape.
Pyloric
Intestinal Foveolar Gland Oxyntic Gland
Features Type Type Adenoma Polyp/Adenoma
Morphology low power Fig. 2.77 Fig. 2.93 Fig. 2.99 Fig. 2.119
Morphology high power Fig. 2.83 Fig. 2.96 Fig. 2.110 Fig. 2.122
- - + +
+ - - -
(Background
IM will stain)
AMAG, autoimmune metaplastic atrophic gastritis; EMAG, environmental metaplastic atrophic gastritis; F,
female; FAP, familial adenomatous polyposis; GERD, gastroesophageal reflux disease; HGD, high-grade
dysplasia; IHC, immunohistochemistry; IM, intestinal metaplasia; LGD, low-grade dysplasia; M, male.
Figure 2-78. Gastric adenoma, intestinal type. The most common gastric adenoma subtype is
the intestinal type. It is fairly easy to identify owing to the marked hyperchromasia at low
magnification and presence of goblet cells.
Figure 2-79. Gastric adenoma, intestinal type. There is an abrupt transition (arrow) from
nondysplastic to dysplastic epithelium.
Figure 2-80. Gastric adenoma, intestinal type. The marked hyperchromasia and presence of
goblet cells make intestinal-type adenomas easy to classify as compared with other subtypes.
Figure 2-81. Gastric adenoma, intestinal type. Goblet cells may be sparse in some intestinal
type adenomas, but the adjacent mucosa may provide clues, such as foci of intestinal
metaplasia (arrow).
Figure 2-82. Gastric adenoma, intestinal type. The cytologic features of intestinal-type gastric
adenomas are similar to the conventional colonic tubular adenoma, with elongated and
crowded pseudostratified nuclei. Goblet cells (arrow) are usually present.
Figure 2-83. Gastric adenoma, intestinal type. These lesions are readily diagnosed at low
magnification, with strong resemblance to the colonic tubular adenoma. The presence of goblet
cells (arrow) are also a helpful clue to classification.
Figure 2-84. Gastric adenoma, intestinal type. By definition, gastric adenomas are at least low-
grade dysplasia. Although crowded and pseudostratified, the nuclei remain polarized and
organized. There may be increased mitoses and apoptoses (arrow). The presence of goblet
cells (arrowheads) classifies this gastric adenoma as intestinal type.
Figure 2-85. Gastric adenoma, intestinal type. Cytoplasmic features are key to differentiating
the gastric adenoma subtypes. For intestinal-type gastric adenomas, the presence of goblet
cells (arrowheads) is the easiest clue to spot. However, also note the smooth pale eosinophilic
cytoplasm (arrows) in the intervening cells. The luminal surface has a stiff eosinophilic border,
and the cytoplasm lacks clearing, foaminess, or a mucin cap.
Figure 2-86. Gastric adenoma, intestinal type. These cells share a stiff eosinophilic border at
the luminal surface, and a microvillous brush border can sometimes be seen (arrowheads).
The cytoplasm is smooth, pale, and eosinophilic. The absence of clearing, foaminess, and
mucin distinguish this from other gastric adenoma subtypes, especially when goblet cells are
absent.
Figure 2-87. Gastric adenoma, intestinal type. When goblet cells are abundant, classification of
the intestinal-type gastric adenoma is straightforward. Take a moment to note the cytoplasmic
qualities of the intervening cells, which appear pale and smooth. The stiff luminal border
(arrowheads) is distinctly eosinophilic.
Figure 2-88. Gastric adenoma, intestinal type. A microvillous brush border (arrow) is present in
this intestinal-type gastric adenoma. In the absence of goblet cells, the smooth pink cytoplasm
and lack of a mucin cap or cytoplasmic clearing are helpful features.
Figure 2-89. Gastric adenoma, foveolar type (low-grade dysplasia implied). By definition, gastric
adenomas are at least low-grade dysplastic. At low magnification, this gastric polyp is distinctly
hyperchromatic and easy to spot as a dysplastic lesion.
Figure 2-90. Gastric adenoma, foveolar type. This gastric polyp is hyperchromatic a low
magnification due to the presence of low grade dysplasia.
Figure 2-91. Gastric adenoma, foveolar type. The cells of the foveolar type gastric adenoma
recapitulate normal foveolar cells, which have a clear mucin cap (arrowheads). This
cytoplasmic feature distinguishes foveolar adenomas from other gastric adenomas.
Figure 2-92. Gastric adenoma, foveolar type. The nuclei of this foveolar-type adenoma are
slightly crowded and disorganized, but they remain basally located, without the degree of
pseudostratification seen in intestinal-type gastric adenomas. The key to classification,
however, is the presence of a pale pink to clear apical mucin cap along the luminal surface
(arrowheads). Mitotic activity is present (arrow).
Figure 2-93. Gastric adenoma, foveolar type. The nuclei of this foveolar-type adenoma are
slightly elongated and crowded but basally located and lack the degree of elongation and
pseudostratification seen in intestinal-type gastric adenomas. The presence of pale pink to
clear cytoplasmic mucin at the apical border (arrowheads) is key to classification. Mitotic
activity is present (arrow).
Figure 2-94. Gastric adenoma, foveolar type. At low magnification, this polyp is distinctly
hyperchromatic, indicating that it contains at least low-grade dysplasia and should be
categorized as a gastric adenoma. The clear mucinous cap along the surface epithelial cells,
evident even at low magnification, indicates foveolar subtype.
Figure 2-95. Gastric adenoma, foveolar type, higher magnification of the previous figure. The
cells are columnar with crowded and basally located nuclei. The presence of an apical mucin
cap indicates foveolar differentiation.
Figure 2-96. Gastric adenoma, foveolar type. These columnar cells have crowded nuclei and
some pseudostratification but not to the degree seen in intestinal-type gastric adenomas. The
absence of goblet cells and the presence of a pale pink to clear apical mucin cap identify this
gastric adenoma as foveolar subtype.
Figure 2-97. Gastric adenoma, foveolar type. Compare these epithelial cells to those found in
intestinal-type gastric adenomas. These nuclei are crowded and pseudostratified, but the key
differentiating feature is the cytoplasmic quality. Foveolar-type gastric adenomas have a pale
pink to clear apical mucin cap (arrows and arrowheads) not found in intestinal-type gastric
adenomas.
Figure 2-98. Gastric adenoma, foveolar type. This low-grade dysplastic gland is lined by cells
with crowded pseudostratified nuclei and apical mucin caps (arrows). Adjacent nondysplastic
foveolar epithelium (arrowhead ) is in the field for comparison.
Figure 2-99. Gastric adenoma, foveolar type. The apical mucin cap on these dysplastic cells
identifies this polyp as a foveolar-type gastric adenoma. Unlike intestinal-type and pyloric-type
gastric adenomas, these lesions rarely harbor high-grade dysplasia or carcinoma, and the
background mucosa is usually normal and nonatrophic.
The nomenclature ranges from benign (oxyntic gland polyp, chief cell
hyperplasia, chief cell hamartoma) to benign neoplasia (oxyntic gland
polyp/adenoma) to malignant (gastric adenocarcinoma of fundic gland/chief cell
predominant type and gastric adenocarcinoma with chief cell differentiation),
but clinicians may be assured that despite the evolving nomenclature, the data
are quite clear that these lesions are biologically benign. These lesions have
been reported to persist when incompletely resected, but no reported cases
have shown true recurrence or progression of disease. Thus, complete
endoscopic excision of these lesions is sensible, whereas surgical intervention
is overkill.
Figure 2-120. Oxyntic gland polyp/adenoma. These are typically small subcentimeter lesions
found in the gastric fundus with a proliferation of cords and clusters of oxyntic glands. The
surface foveolar epithelium is unchanged.
Figure 2-121. Oxyntic gland polyp/adenoma. This example shows an abrupt change from
normal oxyntic gland architecture at the left to distorted cords of oxyntic cells at the right. The
surface foveolar epithelium is normal.
Figure 2-122. Oxyntic gland polyp/adenoma. There is a deep proliferation of oxyntic glands
arranged in cords and clusters, whereas the overlying foveolar epithelium is unchanged. The
far right of this biopsy shows normal oxyntic mucosa for comparison.
Figure 2-123. Oxyntic gland polyp/adenoma. The cells in this example are bland and exquisitely
uniform. They are arranged in long tubules or cords separated by smooth muscle strands in the
lamina propria (arrowhead ).
Figure 2-124. Oxyntic gland polyp/adenoma. Another example demonstrates some
heterogeneity in the cells of this lesion. Although the cells are oxyntic in origin, this may not be
evident at high-power views. The arrangement of the cells in cords separated by smooth
muscle strands (arrowheads) and the deep location within oxyntic mucosa are clues to the
diagnosis.
Figure 2-125. Oxyntic gland polyp/adenoma. Note the deep proliferation of glands (bracket) that
has embedded smooth muscle between the cords of cells. This example arises in a backdrop
of autoimmune metaplastic atrophic gastritis.
Figure 2-126. Oxyntic gland polyp/adenoma, MUC6 immunohistochemistry. These lesions
demonstrate MUC6 (pyloric differentiation) immunoreactivity and are negative for MUC5AC
(foveolar).
ADENOCARCINOMA
Globally, gastric cancer is among the top five leading causes of cancer and a leading
cause of cancer death. The highest incidence worldwide is in Asia, central Europe, and
South America.62,63 In the United States, the overall incidence rates are modest but
remain greater than those of esophageal cancer, and when compared with Caucasians,
the incidence is increased in all non-Caucasian ethnic and racial groups including
Hispanics, Asian, and African-Americans.64 Although gastric cancer remains a deadly
disease, the incidence and mortality rates have fallen dramatically over the past 80
years.62 Advances in endoscopic techniques have improved detection of early gastric
cancer (defined as pT1), and adjuvant chemotherapy contributes to improved overall
mortality and 5-year survival rates.65,66 Stage remains the most important prognostic
indicator, but a number of considerations affect biopsy interpretation, such as tumor
location and stage, Lauren classification, morphologic variants, background mucosa
characteristics, genetic considerations, and biomarker profile, as discussed later.
Tumors entirely proximal or entirely distal to the GEJ are simply classified
as esophageal or gastric, respectively. For tumors that involve the GEJ, the
distance of the tumor’s midpoint is taken into account. A somewhat arbitrary
distance of 2 cm is the cutoff: the tumor is considered esophageal if its
midpoint is ≤2 cm into the stomach, and gastric if >2 cm (see Fig. 2.128).
Signet ring cell carcinoma is a specific subset of poorly cohesive carcinomas that is
composed predominantly of discohesive single cells with distended cytoplasmic mucin
that displaces and eccentrically compresses the nucleus. This designation should be
restricted to tumors composed predominantly of the signet ring cell type, as other
variants of poorly cohesive carcinomas may also infiltrate in a diffuse or single cell
pattern, but are mucin poor (Figs. 2.135–2.163). In either case, these are the carcinomas
that provide the most challenge on endoscopic biopsies because they lack the helpful
red flag of an endoscopic mass, and stromal desmoplasia is frequently absent. Examine
gastric biopsies with careful attention to areas of pallor, crush artifact, or slightly
increased cellularity. Make sure to account for every cell in the tissue and call out any
possible mimickers before moving on (e.g., crushed oxyntic glands, mucous neck cells,
xanthoma cells). If there is any uncertainty, a pancytokeratin immunostain can aid in
highlighting the architecture of epithelial cells (Figs. 2.142 and 2.152), and consultation
with a fellow surgical pathologist may clarify matters. Squamous cell carcinomas may
be either keratinizing or nonkeratinizing, and adenosquamous carcinomas should have at
least 25% squamous component by volume mixed with a glandular component. Biopsy
material may not be wholly representative, and final morphologic classification can be
either reserved for or revised following review of the resection specimen. The
carcinoma with lymphoid stroma (also referred to as medullary carcinoma or
lymphoepithelioma-like carcinoma) may resemble well differentiated tubular
adenocarcinomas or undifferentiated carcinomas with poorly formed sheets of tumor
cells, but in all instances it contains a prominent stromal lymphoid infiltrate, sometimes
nearly obscuring the carcinoma itself (Figs. 2.164–2.171). This tumor has reactivity for
Epstein-Barr virus–encoded RNA by in situ hybridization (EBER ISH) and is more
commonly seen in the proximal stomach, among men and in a younger age group. Some
observers have reported significantly better prognosis via longer disease-free survival
and overall cancer survival attributed to the younger age at presentation and less lymph
node metastasis.74,75 Another interesting variant, hepatoid adenocarcinoma,
morphologically resembles hepatocellular carcinoma with large polygonal cells
containing abundant eosinophilic granular cytoplasm (Figs. 2.172–2.175). These
hepatoid gastric adenocarcinomas can raise concern for metastatic hepatocellular
carcinoma and may even express immunoreactivity for alpha-fetoprotein (AFP) and
glypican-3, which are also reactive in hepatocellular carcinomas.76 SALL4
immunohistochemistry may help differentiate these tumors, as it is expressed in 89% of
AFP-positive hepatoid gastric adenocarcinomas and negative in most hepatocellular
carcinoma.76 However, we would caution that a subset of primary liver hepatocellular
carcinomas are SALL4 reactive; these latter tumors tend to be aggressive but, thankfully,
targetable.77 Tumors demonstrating features that are not classifiable into one of the other
WHO categories are considered undifferentiated carcinomas.
Adenocarcinoma
Carcinoma with lymphoid Poorly differentiated carcinoma with prominent lymphoid infiltrate
stroma Associated with Epstein-Barr virus
Better prognosis
Large cell NEC Poorly differentiated Cells are large and pleomorphic
High grade Moderate amount of cytoplasm
Marked nuclear atypia Prominent nucleoli
Synaptophysin+ or
Small cell NEC chromogranin+ Cells are small
May have: Finely granular chromatin (“salt
Focal necrosis and pepper”)
Ki-67 >20% Indistinct nucleoli
>20 mitoses per 10 HPF
Mixed adenoneuroendocrine >30% each of gland-forming and
carcinoma (MANEC) neuroendocrine areas
(Adenocarcinomas showing
immunoreactivity for
neuroendocrine markers is not
sufficient for diagnosis.)
Figure 2-135. Benign gastric mucous neck cells. Do not overcall carcinoma when benign
gastric mucous neck cells are seen. These mucous filled cells found in the neck of gastric
pits/glands are inconspicuous in normal tissues but can appear concerning when tissue is
crushed, fragmented, or reactive. Do not make a diagnosis of signet ring carcinoma unless
there is 100% certainty; suggest rebiopsy if the diagnosis is unclear.
Figure 2-136. Gastric adenocarcinoma, diffuse type. These discohesive cells are infiltrating
beneath an intact and benign surface epithelium.
Figure 2-137. Gastric adenocarcinoma, diffuse type. Single discohesive and non-gland-forming
cells infiltrate the wall of the stomach. The cytoplasm is filled with mucin and pushes the
nucleus off to one side, similar in profile to a signet ring.
Figure 2-138. Gastric adenocarcinoma, diffuse type, with signet ring cells. True signet ring cells
(arrow) have cytoplasm distended with a clear vacuole of mucin that displaces and
compresses the nucleus.
Figure 2-139. Metastatic lobular breast carcinoma. These tumor cells appear similar to signet
ring cells, but lobular breast carcinoma cells do not contain mucin. Instead, there is a single
round cytoplasmic vacuole with a sharply demarcated border, which sometimes contains a
dense hyaline eosinophilic body (arrow) imparting a targetoid appearance.
Figure 2-140. Gastric adenocarcinoma, diffuse type, with signet ring cells. Signet ring cells
have distended clear cytoplasm filled with mucin that displaces the nucleus to the periphery.
This example shows marked variation in size and shape among the signet ring cells.
Figure 2-141. Gastric adenocarcinoma, diffuse type. At low magnification, one can appreciate
how the malignant cells diffusely infiltrate the gastric mucosa without forming an obvious mass
lesion. The surface epithelium is intact, and the gastric pits remain relatively evenly spaced.
The absence of significant architectural disturbance makes the diffuse type of gastric
adenocarcinoma more difficult to spot than the intestinal mass-forming type.
Figure 2-142. Gastric adenocarcinoma, diffuse type, cytokeratin 7 immunohistochemistry of the
previous figure. A cytokeratin immunostain can be helpful to assess the architecture in difficult
cases. In this example, CK7 highlights a lack of normal gland formation and single infiltrating
cells at the base of the biopsy.
Figure 2-143. Gastric adenocarcinoma, diffuse type, higher magnification of the previous figure.
The cells in this example are markedly atypical with large, irregularly shaped nuclei that vary in
size.
Figure 2-144. Gastric adenocarcinoma, diffuse type. Take care to look closely at “busy” gastric
biopsies. At low magnification, the inflammatory cells and reactive gastropathy changes
obscure a focus of gastric adenocarcinoma (arrow). One good rule of thumb is to always
review gastric biopsies at high magnification and mentally account for all cells present.
Figure 2-145. Gastric adenocarcinoma, diffuse type, higher magnification of the previous figure.
Amid a background of acute and chronic inflammation, this cluster of signet ring cells (arrow)
could be easily overlooked. Do not forget to always look beyond the first (obvious) diagnosis.
Figure 2-146. Gastric adenocarcinoma, diffuse type. This low-power view shows a “busy”
gastric biopsy with surface erosion and abundant inflammation. Always look closer at areas
that appear abnormal at low magnification.
Figure 2-147. Gastric adenocarcinoma, diffuse type, higher magnification of the previous figure.
Even at midpower magnification, the adenocarcinoma hiding in this busy tissue fragment is not
clear. The malignant cells blend into the background of inflammation which is distracting one’s
eye.
Figure 2-148. Gastric adenocarcinoma, diffuse type, higher magnification of the previous figure.
These sneaky signet ring cell clusters (arrows) are only visible at high magnification,
underscoring the importance of reviewing each gastric biopsy at high power and mentally
registering each and every cell.
Figure 2-149. Gastric adenocarcinoma, diffuse type. This low-power view shows gastric
mucosa with an area of edema and lamina propria hemorrhage. It looks harmless at this power,
but any area of abnormality should always trigger a higher-power perusal.
Figure 2-150. Gastric adenocarcinoma, diffuse type, higher magnification of the previous figure.
Impossible to see at low magnification, these small malignant cells infiltrate between the benign
glands, leaving the architecture relatively intact.
Figure 2-151. Gastric adenocarcinoma, diffuse type. Areas of gastric erosion and chronic
inflammation should always trigger a closer examination. Inflammation can be particularly
devious in hiding gastric cancer.
Figure 2-152. Gastric adenocarcinoma, diffuse type, pancytokeratin immunostain of the
previous figure. A pancytokeratin stain highlights a broad area containing single infiltrating cells
(bracket). The extent of invasive carcinoma is frequently surprising when highlighted in this
manner and demonstrates how challenging it is to discern these cells on H&E. Contrast the
malignant area against the normal glandular architecture to the left.
Figure 2-153. Gastric adenocarcinoma, diffuse type, higher magnification of the previous figure.
The malignant cells are obscured by the inflammatory backdrop. Some remnant benign glands
add to the difficulty in diagnosis. Even at this magnification, it can be challenging to point out the
malignant cells. Always take a moment to go to high magnification and mentally account for
each cell.
Figure 2-154. Gastric adenocarcinoma, diffuse type, higher magnification of the previous figure
. High magnification reveals many malignant cells admixed with acute and chronic inflammatory
cells. At this magnification, it is possible to name each cell present as benign or malignant.
Figure 2-155. Gastric adenocarcinoma, diffuse type. These discohesive tumor cells are
characteristic of diffuse-type adenocarcinoma by Lauren classification, infiltrative type by Ming
classification, and poorly cohesive carcinoma by WHO classification. The cells lack clear
cytoplasmic mucin vacuoles and thus are not true signet ring cells.
Figure 2-156. Gastric adenocarcinoma, diffuse type. Another example of the infiltrative
discohesive tumor cells found in diffuse-type adenocarcinoma. These cells permeate the
gastric wall without forming a mass lesion and frequently have no background gastritis or other
gastric pathology.
Figure 2-157. Gastric adenocarcinoma, diffuse type, eosinophilic variant. The cells in this
unusual example of diffuse-type adenocarcinoma are deeply eosinophilic and lack
intracytoplasmic mucin. This is classified as a poorly cohesive carcinoma by the WHO
classification, which includes signet ring cell and other variants. The tumor contains <50%
glands and is therefore poorly differentiated.
Figure 2-158. Gastric adenocarcinoma, diffuse type, eosinophilic variant. The cells are
markedly atypical with eosinophilic cytoplasm and a myxoid backdrop. These are not signet ring
cells because they lack the characteristic clear vacuole of intracytoplasmic mucin.
Figure 2-159. Gastric adenocarcinoma, diffuse type, eosinophilic variant. This unusual variant
contains cells that have abundant eosinophilic cytoplasm without mucin. Some areas show
cohesive malignant cells arranged in cords.
Figure 2-160. Gastric adenocarcinoma, diffuse type, eosinophilic variant. The tumor cells
appear plasmacytoid with irregular nuclei containing conspicuous nucleoli. These should not be
mistaken for signet ring cells, as they lack the characteristic compressed nucleus and
cytoplasmic mucin vacuole.
Figure 2-161. Gastric adenocarcinoma, WHO mucinous type. These tumors contain >50%
extracellular mucin pools and are considered a diffuse type by Lauren classification and are
poorly differentiated by definition.
Figure 2-162. Gastric adenocarcinoma, WHO mucinous type, Lauren diffuse type. There is
abundant intracellular and extracellular mucin, the latter of which comprises >50% of the tumor.
Figure 2-163. Gastric adenocarcinoma, WHO mucinous type, Lauren diffuse type. Mucinous
adenocarcinoma may or may not contain signet ring cells (arrows).
Figure 2-164. Gastric adenocarcinoma with lymphoid stroma (EBV-associated gastric
adenocarcinoma). At low magnification, this mass-forming lesion shows a prominent lymphoid
infiltrate that obscures the glands. At first glance, one might consider a lymphoid neoplasm
such as MALT lymphoma.
Figure 2-165. Gastric adenocarcinoma with lymphoid stroma (EBV-associated gastric
adenocarcinoma). Higher magnification of the previous figure shows abnormal distorted and
abortive glands with prominent intraepithelial lymphocytes. These features are reminiscent of
lymphoepithelial lesions found in MALT lymphoma, but do not be fooled, and take a closer look
at higher power.
Figure 2-166. Gastric adenocarcinoma with lymphoid stroma (EBV-associated gastric
adenocarcinoma). The intact glands show abundant intraepithelial lymphocytes, but the
abnormal glands are composed of malignant cells, indicating this is in fact a carcinoma and not
a lymphoma.
Figure 2-167. Gastric adenocarcinoma with lymphoid stroma (EBV-associated gastric
adenocarcinoma), pancytokeratin immunostain. Immunohistochemistry highlights the poorly
formed glands, which are more abundant than visualized on H&E and infiltrative in architecture.
Figure 2-168. Gastric adenocarcinoma with lymphoid stroma (EBV-associated gastric
adenocarcinoma), in situ hybridization for Epstein-Barr virus encoded RNA (EBER ISH). A
subset of adenocarcinomas with lymphoid stroma will show positivity for EBER ISH localized to
the tumor nuclei, as in this example.
Figure 2-169. Gastric adenocarcinoma with lymphoid stroma (EBV-associated gastric
adenocarcinoma). Another example of a poorly differentiated gastric carcinoma with abundant
lymphoid cells. The lymphocytes in close association with these sheets of malignant cells are a
clue to the diagnosis of EBV-associated gastric cancer and a relatively better prognosis.
Figure 2-170. Gastric adenocarcinoma with lymphoid stroma (EBV-associated gastric
adenocarcinoma). Higher magnification of the previous figure shows syncytial sheets of tumor
cells with indistinct cell borders and numerous intratumoral lymphocytes. Previous names for
this tumor include medullary carcinoma or lymphoepitheliallike carcinoma. The tumor is
considered poorly differentiated because it has <50% gland formation.
Figure 2-171. Gastric adenocarcinoma with lymphoid stroma (EBV-associated gastric
adenocarcinoma). In situ hybridization for Epstein-Barr encoded RNA shows the tumor nuclei
contain EBV RNA. This finding is associated with a better prognosis.
Figure 2-172. Gastric adenocarcinoma, WHO hepatoid variant. This tumor resembles
hepatocellular carcinoma with large polygonal cells containing abundant granular cytoplasm.
Pseudoacini may be present, and this example even shows bile formation.
Figure 2-173. Gastric adenocarcinoma, WHO hepatoid variant. This gastric cancer shows
sheets of malignant cells with abundant eosinophilic granular cytoplasm and prominent nucleoli
arranged in sheets or wide trabeculae with fine endothelial spaces simulating hepatic
sinusoids. The cytology and architecture are hepatoid. Exclusion of metastatic hepatocellular
carcinoma to the stomach may require correlation with radiologic findings.
Figure 2-174. Gastric adenocarcinoma, WHO hepatoid variant. The tumor is composed of large
cells with abundant eosinophilic cytoplasm and round nuclei with prominent nucleoli arranged in
vague trabeculae. The delicate vasculature reinforces the similarity to widened hepatic plates
and sinusoids.
Figure 2-175. Gastric adenocarcinoma, WHO hepatoid variant, hepatocyte-specific antigen
(HSA). Hepatoid variant of primary gastric carcinoma may be reactive for HSA (pictured), AFP,
and glypican-3, all of which are also reactive in hepatocellular carcinoma. SALL-4 can help
differentiate these tumors, as it is positive in most gastric hepatoid carcinomas and negative in
hepatocellular carcinoma.
NECs are classified as such by their poorly differentiated high-grade cytology and
marked nuclear atypia, although this was not always the case. The now outdated WHO
2010 classification of neuroendocrine neoplasms defined three grades based purely on
mitotic rate and Ki-67 index, with the high grade (G3) category defined as >20 mitoses
per 10 HPF or Ki-67 proliferation index >20% without regard to histologic atypia.71
This older WHO 2010 classification regarded G3 neoplasms as synonymous with
poorly differentiated NECs. However, when investigators divided the G3 group into
morphologically well- and poorly differentiated tumors, the poorly differentiated tumors
had different etiologies, genetic alterations, and response to treatment with worse
survival outcomes.78-82 For these reasons, the updated classification of endocrine
tumors (WHO 2017) now relies first on morphologic features; a tumor should be
identified as well differentiated or poorly differentiated before further stratification by
mitotic count and Ki-67 proliferative index. In the case of poorly differentiated
morphology, these should always be classified as NECs and staged as carcinomas.83
This group can be further divided into (1) small cell type, in which the cells are small
with finely granular “salt and pepper” chromatin and inconspicuous nucleoli (Figs.
2.176–2.179); (2) large cell type, in which the cells are large with a moderate amount
of cytoplasm and prominent nucleoli (Figs. 2.180–2.181); and (3) mixed
neuroendocrine-nonneuroendocrine neoplasm (MiNEN; previously “mixed
adenoneuroendocrine carcinoma” or MANEC), in which the tumor is composed of at
least 30% both NET and adenocarcinoma or other high-grade carcinoma.
Immunoreactivity for neuroendocrine markers within an adenocarcinoma does not
indicate MiNEN.
FAQ: What does it mean when a signet ring cell carcinoma is not staining for
mucin?
Not all tumors with single infiltrating cells are signet ring cell carcinomas.
True signet ring cells have cytoplasm distended with a clear vacuole of mucin
that displaces and compresses the nucleus (Fig. 2.138). Other poorly cohesive
or diffuse type variants without intracytoplasmic mucin may have eosinophilic
cytoplasm instead (Figs. 2.157–2.160) and should be classified as diffuse type
gastric adenocarcinoma without the “signet ring” designation. Do not forget to
consider metastatic lobular breast carcinoma, which may also mimic signet ring
cells. Lobular breast carcinoma cells contain intracytoplasmic lumina, which can
mimic the mucin vacuole of signet ring cells. However, the single round vacuole
found in lobular breast carcinoma cells have thick sharply demarcated edges,
sometimes contain a dense hyaline eosinophilic body imparting a “targetoid”
appearance (Fig. 2.139), and are not mucicarminophilic. They can be
differentiated from gastric cancer with immunohistochemistry for breast
markers such as GCDFP or GATA3.
FAQ: What is H. pylori CagA and how does it relate to gastric cancer?
FAQ: What is the link between nitrates, nitrites, and nitrosamines and gastric
cancer? Which of these ingredients do I need to avoid, exactly?
Do not quit your hot dog habit just yet. High consumption of processed
meats, such as ham, bacon, sausages, and hot dogs, is linked to an increased
gastric cancer risk, and many attribute this to the food additives nitrates and
nitrites that retard microbial spoilage, preserve meat’s recognizable
appearance, and enhance flavor. As a group, these compounds containing an –
NO group are referred to as N-nitroso compounds, which all become the same
end product when consumed. Nitrates (NO3) are inert until they are reduced to
nitrites (NO2) by oral bacteria, are swallowed, and, upon hitting the acidic
gastric juices, are converted to nitrous acid (HNO2), which then binds to
amines, amides, and amino acids to form nitrosamines. Major sources of
human exposure to N-nitroso compounds include diet, occupational exposure,
and smoking, but in vivo formation accounts for up to 75% (that is right, your
body is making most of it!).90 Several studies have investigated the potential
association between dietary consumption of nitrates, nitrites, and nitrosamines
with gastric cancer. Increased consumption of nitrites and N
nitrosodimethylamine are linked to an increased risk for gastric cancer,91
whereas several studies have demonstrated a protective effect of nitrates,91-93
and this was found related to the high levels of nitrates found in green leafy
vegetables. Researchers conclude that higher intake of antioxidants relative to
nitrates offers protective effects. So, although the hype surrounding nitrate-free
products is not entirely a load of bologna, a few bites of broccoli may be kinder
to your GI tract.
Familial Predisposition
Most gastric cancers are sporadic, but about 10% of cases occur with aggregation
within families. Truly hereditary familial gastric cancer accounts for 1% to 3% of the
global burden of gastric cancer and comprises at least two major syndromes: hereditary
diffuse gastric cancer (HDGC) and GAPPS of the stomach (essentially a variant of
FAP).
Hereditary Diffuse Gastric Cancer
HDGC is inherited in an autosomal dominant pattern with high penetrance. Nearly 50%
of HDGC is associated with germline truncating mutations of the CDH1 gene, located
on chromosome 16q22.1, which was first identified in three Maori families from New
Zealand that were predisposed to diffuse gastric cancer. More than 75 families with
nearly 4,000 probands have since been identified, and we now know these mutations
are not concentrated in a single hotspot but are evenly distributed along the CDH1 gene
in several different exons and, to date, more than 155 different germline CDH1
mutations have been identified.94,95
Quick Fact: CDH1 is a tumor suppressor gene that requires a second hit for tumor
formation. The CDH1 gene provides instructions for making the E-cadherin protein,
which is a membrane-bound cell-adhesion molecule that also acts as a tumor suppressor
protein, preventing cells from growing or dividing too rapidly. Simply put, CDH1 and
E-cadherin are important in controlling cellular cohesion (cells sticking together) and
division. The mechanism by which the second allele of E-cadherin is inactivated is
diverse and includes promoter hypermethylation, mutation, and loss of heterozygosity,
any of which results in loss of E-cadherin expression. Functionally, the resulting loss of
cellular cohesion leads to an increased ability for tumor cells to invade and migrate, a
feature seen in diffuse gastric cancers and invasive lobular breast carcinomas.
The lifetime risk of gastric cancer in individuals from these families is 70% for men
and 56% for women, and the average age of onset is 38 years (range 14 to 82
years).94,95 Asymptomatic carriers of the mutation are recommended prophylactic total
gastrectomy generally between ages 20 and 30 years, during which the risk of gastric
cancer rises from <1% to 4%.95 Women in these affected families also have a 42%
cumulative risk of lobular breast carcinoma, with increased risk before age 30.94 Cases
of signet ring cell appendiceal and colorectal cancers have also been reported, but these
do not appear increased as compared with those in non-CDH1-mutated populations.94
Histologically, the tumors in these patients are identical to sporadic diffuse gastric
cancers and are similarly challenging to identify (Figs. 2.186–2.195). In situ lesions or
pagetoid spread of signet ring cells are described commonly in the literature with
CDH1-mutated diffuse gastric cancer, but identifying these nearly invisible lesions
requires skillful experience and, in most cases, prior knowledge of the patient’s history.
Consensus guidelines for CDH1 mutation testing include the following criteria:
1. Two gastric cancer cases in a family regardless of age (at least on
confirmed diffuse type)
2. Diffuse gastric cancer in an individual <40 years
3. Personal or family history (first- or second-degree relative) of diffuse gastric
cancer and lobular breast cancer, one diagnosed <50 years.95
Other patients in whom testing should be considered include:
1. Bilateral lobular breast cancer or family history (first- or second-degree
relative) of ≥2 cases of lobular breast cancer <50 years
2. Personal or family history (first- or second-degree relative) of cleft lip/palate
in a patient with diffuse gastric cancer
3. Any individual with in situ signet ring cells and/or pagetoid spread of signet
ring cell on a gastric biopsy
Figure 2-186. Hereditary diffuse gastric cancer, CDH1 mutated. A nearly invisible focus of early
gastric cancer (arrow) is found in this prophylactic gastrectomy specimen from a patient known
to carry a CDH1 germline mutation. Without prior knowledge of the patient’s history, such a
small focus could easily be missed.
Figure 2-187. Hereditary diffuse gastric cancer, CDH1 mutated, higher power of the previous
figure. The small cluster of signet ring cells (bracket) invade into the lamina propria without
disturbing the overall architecture or surface epithelium, making it extremely difficult to detect.
Figure 2-188. Hereditary diffuse gastric cancer, CDH1 mutated. Another example of early
gastric cancer in a patient with CDH1 gene mutation shows subepithelial signet ring cells
(brackets) infiltrating the lamina propria without disturbing the overall architecture or surface
mucosa, features that would normally alert the pathologist to take a closer look.
Figure 2-189. Hereditary diffuse gastric cancer, CDH1 mutated, higher magnification of the
previous figure. These pale signet ring cells (brackets) infiltrate the lamina propria and cause no
epithelial or stromal reaction. Note the absence of background gastritis or intestinal metaplasia.
Figure 2-190. Hereditary diffuse gastric cancer, CDH1 mutated. Single cells (arrowheads) are
so subtle as to be nearly invisible. Even with knowledge of the patient’s clinical history, finding
these foci on a prophylactic gastrectomy can be extremely challenging.
Figure 2-191. Hereditary diffuse gastric cancer, CDH1 mutated. Signet ring cells are easier to
identify when sufficient numbers cluster together. The absence of an expansile lesion, surface
change, or stromal changes is highly characteristic.
Figure 2-192. Hereditary diffuse gastric cancer, CDH1 mutated. This example has a backdrop
of inactive chronic gastritis that obscures an already difficult diagnosis. Single malignant signet
ring cells (arrows) are surrounded by chronic inflammatory cells.
Figure 2-193. Hereditary diffuse gastric cancer, CDH1 mutated, PAS stain. Some experts
suggest performing PAS stain in lieu of H&E to screen prophylactic gastrectomy specimens.
The PAS stain highlights tumor cells (arrow), providing a better contrast as compared with
H&E. This technique can also be helpful in biopsy material.
Figure 2-194. Hereditary diffuse gastric cancer, CDH1 mutated, PAS stain, higher magnification
of the previous figure. The mucin within the signet ring cells (arrow) are PAS positive.
Figure 2-195. Hereditary diffuse gastric cancer, CDH1 mutated, PAS stain, higher magnification
of the previous figure. Individual signet ring cells are highlighted by PAS. Although many cells
contain intracytoplasmic mucin, benign cells maintain normal architecture and gland formation.
By comparison, the signet ring cells are single, discohesive, and disorganized.
FAQ: Is there a role for endoscopic surveillance for patients with HDGC?
There are no reliable screening tests that allow for early detection of diffuse
gastric cancer. Prophylactic gastrectomy is the treatment of choice for CDH1
mutation carriers after age 20 years, but annual endoscopic surveillance may
be considered for patients under the age of 20 years, patients who refuse or
postpone gastrectomy (e.g., fertility concerns), and individuals who have
genetic variants of undetermined significance. Note, however, that because
diffuse gastric carcinomas do not form endoscopically visible lesions,
endoscopic surveillance is likely to have extremely low detection rates for
cancers in these patients. Random mapping biopsies may be sent to pathology,
but the estimated number of biopsies necessary to capture a single focus of
cancer (90% detection rate) is theoretically projected at 1768!96
Figure 2-196. Gastric adenocarcinoma and proximal polyposis syndrome, endoscopic view.
Diagnostic criteria include >100 gastric polyps limited to the gastric body/fundus (endoscopic
view pictured) without colorectal or duodenal polyposis and autosomal dominant inheritance.
Figure 2-197. Fundic gland polyps in a patient with GAPPS. The polyps in GAPPS are primarily
fundic gland polyps (pictured).
Figure 2-198. Fundic gland polyp in a patient with GAPPS. Most of the polyps found in GAPPS
are fundic gland polyps. This example shows cystically dilated glands amid oxyntic glands and
overlying normal foveolar epithelium.
Figure 2-199. Pyloric gland adenoma in a patient with GAPPS. This patient had a large gastric
pyloric gland adenoma (pictured) giving rise to adenocarcinoma, in addition to hundreds of
fundic gland polyps.
Figure 2-200. Pyloric gland adenoma (PGA) in a patient with GAPPS, higher magnification of
the previous figure. Architecturally, PGAs are composed of back-to-back tubules, which when
large can form papillary or frondlike extensions.
Figure 2-201. Pyloric gland adenoma (PGA) in a patient with GAPPS, higher magnification of
the previous figure. The cells of PGA can be columnar to cuboidal and have abundant clear to
eosinophilic cytoplasm with a ground glass or foamy appearance.
Figure 2-202. Pyloric gland adenoma in a patient with GAPPS. Other areas closely resemble
pyloric glands, with closely packed small glands composed of cuboidal cells with clear foamy
cytoplasm and nuclei pushed toward the basement membrane.
PEARLS & PITFALLS: PD-1 and PD-L1 Negative Results on Archived Material
If PD-L1 expression is not detected in an older archived gastric cancer
specimen, the FDA recommends assessing the feasibility of a fresh tumor
biopsy for repeat testing.
Phase I: Experimental treatment is tested on a small group of people (20 to 80) for the
first time. The purpose is to evaluate safety and identify side effects.
Phase II: Experimental treatment is tested on a larger group of people (100 to 300) to
determine effectiveness and further evaluate safety.
Phase III: Experimental treatment is administered to large groups of people (1000 to
3000) and compared against standard treatment. Effectiveness, side effects, and
safety are further assessed.
Phase IV: This phase follows FDA approval and availability of the treatment for the
public. Researchers continue to track safety and monitor information about risks,
benefits, and optimal use.
PEARLS & PITFALLS: Select the Tissue Block With the Lowest-Grade Tumor
Morphology!
As mentioned earlier, HER2 overexpression is seen more frequently in the
intestinal phenotype (24%) and mixed tumors (20%) as compared with diffuse
signet ring cell type (0% to 6%).109,110 Low-grade tumors show greater
frequency of HER2 positivity compared with high grade,111 and so block
selection should target the lowest-grade tumor and areas showing intestinal
differentiation to have the highest prospect of positive HER2 result. Other rare
morphologic variants of gastric adenocarcinoma (e.g., adenosquamous,
papillary, hepatoid) lack sufficient data for comment. More than one tissue
block may be tested if the tumor is morphologically heterogeneous.
Figure 2-203. HER2 negative, score 1+. A sufficient number of tumor cells are staining in this
biopsy specimen (one cancer cell cluster of ≥5 cells), and membranous staining is present.
However, the intensity of the stain is 1+, as it is barely perceptible, even at high magnification
(40x). A score of 1+ is considered negative, and there is no need to reflex to FISH testing.
Figure 2-204. HER2 negative, score 1+. In this biopsy example, the staining criteria are met,
with basal, lateral, and basolateral staining present in at least ≥5 cells of this tumor cell cluster.
However, the stain is faint and requires 40x magnification to be perceptible. This 1+ staining
intensity is considered negative, and there is no need to reflex to FISH testing.
Figure 2-205. HER2 equivocal, score 2+. Sufficient tumor cells are staining (at least one cancer
cell cluster defined as ≥5 cells in a biopsy) in a basolateral membranous pattern. However, the
staining intensity is weak to moderate and is best seen at 10–20x. This is scored as 2+ and
requires follow-up with FISH studies.
Figure 2-206. HER2 equivocal, score 2+. Complete or incomplete basolateral staining is
present in a sufficient number of tumor cells (at least one cancer cell cluster defined as ≥5 cells
in a biopsy). Compared with breast carcinoma, which requires circumferential membranous
staining, discontinuous basal/lateral membranous staining is acceptable with gastric cancers.
However, the staining intensity is weak to moderate and is best seen at 10–20x. This is scored
as 2+ and requires follow-up with FISH studies to determine whether the patient can benefit
from trastuzumab therapy.
Figure 2-207. HER2 positive, score 3+. Sufficient tumor cells are staining (at least one cancer
cell cluster defined as ≥5 cells in a biopsy) in a complete membranous pattern with strong
reactivity visible at low magnification (2–4x). A 3+ interpretation is positive and requires no
additional FISH testing. Although this example shows complete membranous reactivity, gastric
HER2 interpretation requires only basal, lateral, or basolateral staining.
Figure 2-208. HER2 positive, score 3+. At least 10% of tumor cells in a resection specimen
must show reactivity to be properly graded. This example shows strong and complete
membranous reactivity in the tumor cells visible at low magnification (2–4x). A score of 3+ is
considered positive, and patients may benefit from trastuzumab therapy.
Figure 2-209. HER2 negative, score 0. Beware of strong staining in areas of overlying dysplasia
(pictured) or intestinal metaplasia. Areas of metaplasia or regenerative changes near ulcers
may also show false-positive reactivity. Interpretation of HER2 should be limited to the invasive
carcinoma only, and it is helpful to have the H&E handy for correlation.
Figure 2-210. HER2 negative, score 0. The invasive cancer (bracket) is negative for HER2, but
the overlying dysplastic epithelium shows strong reactivity. This is a common false-positive
pitfall to avoid; always inspect well-preserved areas of invasive tumor only.
Figure 2-211. HER2 negative, score 0. Although there appears to be linear membranous
reactivity in at least five tumor cells, this shows an isolated luminal staining pattern (arrowhead
). True positive staining requires basal, lateral, or basolateral membranous reactivity.
Figure 2-212. HER2 negative, score 0. There is moderate to strong reactivity in at least five
tumor cells, but the staining patterns are nuclear, cytoplasmic, granular, and extracellular. True
linear membranous reactivity in a lateral/basolateral distribution is absent. This HER2 should be
interpreted as negative.
ISH criteria Single probe HER2 ≥6.0 signals per cell Same
copy number
No. Unlike breast carcinoma, there are no specific fixation requirements for
HER2 testing of gastric/gastroesophageal adenocarcinoma (see Table 2.6).
FAQ: Should clinicians wait for HER2 results before initiating therapy?
No. Many centers send out testing to reference laboratories, which may
increase turnaround time for HER2 results. Although most oncologists will be
anxious to receive these results, there is no need to know HER2 status before
starting chemotherapy. If HER2 results are subsequently positive, clinicians
may later add trastuzumab to the treatment plan. In addition, most patients with
gastric cancer are symptomatic at the time of diagnosis and will benefit from
immediate oncologic management. For pathologists, the turnaround time
benchmark is 90% of reports within 10 working days from the date of
procedure. For send outs, 90% of specimens should be sent within 3 days of
tissue processing.
FAQ: Why do scoring criteria for HER2 IHC differ between gastric and breast
cancers?
In gastric cancers, HER2 testing by FISH and IHC show only moderate
fidelity, as up to 20% of gastric cancers with negative IHC interpretation (0 or
1+) show positive amplification by FISH. However, no significant survival benefit
from trastuzumab is seen in these patients and, as a result, IHC is the first-line
test for HER2 overexpression.119 This is in contrast to the experience with
breast carcinoma whereby an extremely low threshold to perform FISH is
maintained and every attempt is sought to achieve a positive HER2 result.
Partially driving this practice is the high efficacy of trastuzumab in HER2+
breast carcinoma along with the low toxicity profile of the drug. This makes
trastuzumab low risk and potentially high yield for clinicians to include in their
arsenal against breast cancer. However, this does not hold true for gastric
cancers, and thus the testing algorithms diverge. Unlike in breast cancer, there
is no significant survival benefit for trastuzumab if IHC is 0 or 1+ even when
FISH shows HER2 amplification. On the other hand, if there is uncertainty over
IHC scoring of 1+ versus 2+, the best approach is to simply reflex to FISH.
No. There are no data to support repeat HER2 testing if initial testing is
negative, although some clinical colleagues may request this.
PEARLS & PITFALLS: The Path Report Should Specify the Antibodies and
Probes Used
Several HER2 antibodies are offered through various vendors, and there is no
specific recommended antibody. The ToGA trial used HercepTest, whereas
other studies have applied Ventana 4B5 or Thermo Fischer Scientific CB11, and
even more variations are available on the market. Although concordance among
antibodies is moderate to good, the ASCO/CAP/ASCP guidelines strongly
recommend laboratories specify the antibodies and probes used for the test.120
An example of this standard verbiage is included later. The guidelines also
emphasize that assays should be appropriately validated for HER2 IHC and
ISH on gastroesophageal adenocarcinoma specimens, although this is standard
practice for any laboratory test. As with any other test, 20 positive and 20
negative gastroesophageal adenocarcinomas should be verified for FDA-
approved tests, and 40 samples for laboratory-developed tests.
Example verbiage for inclusion in report:
Method: Testing is performed using FDA-approved Ventana Pathway HER2
(4B5) rabbit monoclonal primary antibody and a proprietary detection system.
No expression (HER2 score of 0), low expression (HER2 score of 1+), and high
expression (HER2 score of 3+) controls are used. All controls show
appropriate reactivity.
Scoring: Scoring is performed according to the following article: Ruschoff J,
Dietel M, Baretton G, et al. HER2 diagnostics in gastric cancer-guideline
validation and development of standardized immunohistochemical testing.
Virchows Arch. 2010; 457(3):299-307.
WELL-DIFFERENTIATED NEUROENDOCRINE
TUMORS (FORMERLY “CARCINOID”)
Based on WHO 2017 classification, well-differentiated neuroendocrine tumors (WD-
NETs) are distinguished from NECs by their morphologic features, independent of
mitotic count or Ki-67 proliferative index. This is a departure from previous WHO
2010 classification for neuroendocrine neoplasms, in which tumors were stratified
based solely on mitoses and Ki-67. The updated system not only more reliably indicates
prognosis and response to therapy but is also more intuitive to pathologists. For
example, tumors that are morphologically uniform are classified as WD-NETs, whereas
NECs are classified by their poorly differentiated high-grade cytology and marked
nuclear atypia. NEC is discussed separately in the gastric adenocarcinoma section
“Tumor Classification”, as its prognosis and staging more closely reflect those of
gastric adenocarcinoma.
Most gastric WD-NETs are composed of ECL cells, typically in the corpus and
fundus (90%),121 which express chromogranin A or synaptophysin by
immunohistochemistry. Endoscopically they appear as submucosal nodules or polyps
(Fig. 2.213), sometimes with ulcerations. Gastric WD-NETs are classified into three
groups, each arising in different clinical contexts (see Table 2.7), and each with
divergent prognoses and treatment protocols. In isolation, the tumors are histologically
indistinguishable and are composed of nests or trabeculae of small, uniform, polygonal,
or cuboidal cells with lightly eosinophilic and finely granular cytoplasm. The nuclei are
round or oval with smooth nuclear borders and stippled chromatin with indistinct
nucleoli. The key to differentiating the three types of gastric WD-NETs from one another
requires examination of the nonpolypoid background mucosa, yet again underscoring the
importance of background gastric biopsies with all gastric lesions.
Figure 2-213. Gastric well-differentiated neuroendocrine tumor. This pale endoscopic nodule
appears submucosal. The background gastric mucosa also shows a mosaic pattern and
patchy atrophy.
Figure 2-214. Gastric WD-NET, histology of the previous figure. At low magnification, this is an
expansile lesion composed of sheets of uniform cells with a slightly trabecular architecture. The
base of the lesion is invading into the muscularis mucosae. The prognosis of this lesion
depends upon the etiopathogenesis, which can be deduced from the changes found in the
background nonlesional sample.
Figure 2-215. Autoimmune metaplastic atrophic gastritis, background mucosa of the previous
figure. Background nonlesional gastric tissue is important when encountering a WD-NET in the
stomach. Type 1 WD-NETs have an excellent prognosis and arise in the setting of AMAG,
which shows a constellation of features. The oxyntic glands are absent in this image because
of complete oxyntic gland atrophy, and this is accompanied by background chronic
inflammation, intestinal metaplasia (arrowhead ), and pyloric metaplasia (arrow).
Figure 2-216. Autoimmune metaplastic atrophic gastritis. At lower magnification, one can
appreciate the complete absence of oxyntic glands, the presence of a low-lying lymphocytic
infiltrate, intestinal metaplasia (arrowhead ), and pyloric gland metaplasia (arrow). These
features, in conjunction with ECL-cell hyperplasia (not pictured), indicate AMAG.
Figure 2-217. Autoimmune metaplastic atrophic gastritis, nodular ECL-cell hyperplasia. ECL-
cell hyperplasia is not always visible on H&E stain, but these small nodular aggregates
(arrowheads) stain with chromogranin. To diagnose AMAG, one must find linear or nodular
ECL-cell hyperplasia (defined as at least five adjacent cells) in combination with oxyntic gland
atrophy (note the absence of the typical pink parietal cell and blue chief cells), low-lying
lymphocytic inflammation, intestinal metaplasia, and pyloric gland metaplasia.
Figure 2-218. Autoimmune metaplastic atrophic gastritis, nodular ECL-cell hyperplasia. Nodular
ECL-cell hyperplasia (arrowhead ) can be confirmed by chromogranin immunostain (not
pictured). The mucosa additionally shows a combination of complete atrophy of oxyntic glands,
low-lying lymphocytic inflammation, intestinal metaplasia, and pyloric metaplasia. The absence
of oxyntic glands leads to loss of acid secretion, which normally inhibits gastrin secretion. In the
absence of acid, gastrin levels increase and lead to ECL-cell hyperplasia, seen here.
Figure 2-219. Autoimmune metaplastic atrophic gastritis, nodular ECL-cell hyperplasia.
Uninhibited gastrin secretion results in nodular hyperplasia of ECL cells (arrowhead ), which
can become neuroendocrine tumors. The size cutoff varies by publication (0.5 vs. 0.5 cm).
However, because the metastatic rate of small lesions is negligible, one practical approach is to
report all small ECL-cell nodules as nodular hyperplasia and reserve the term WD-NET for
endoscopically visible lesions submitted as nodules or polyps.
Figure 2-220. WD-NET arising in AMAG, endoscopy. This endoscopically visible lesion is a
WD-NET, and the background gastric mucosa is nodular with atrophy. Biopsy samples of the
background mucosa (previous figures) show histologic features of AMAG, including numerous
areas containing nodular ECL-cell hyperplasia. Size cutoffs for differentiating hyperplasias from
NETs are arbitrary, and the authors take a practical approach in calling lesions WD-NETs only if
they correlate with an endoscopically visible lesion (arrow).
Figure 2-221. Quick tutorial on the interpretation of AMAG. AMAG is a body-predominant
disease. Biopsies of the gastric antrum (top row) are essentially unremarkable or may show
changes of chemical/reactive gastropathy. Gastrin and chromogranin stains in the antrum
highlight the normal band of stimulatory “G” cells that secrete gastrin. By comparison, the
gastric body and fundus (bottom row) show a constellation of features that can be identified by
H&E, including (1) partial or complete atrophy of oxyntic glands; (2) lymphocytic inflammation,
often low lying; (3) intestinal metaplasia; and (4) pyloric gland metaplasia. The features are
almost indistinguishable from gastric antral tissue containing intestinal metaplasia. Therefore, a
gastrin stain can be performed to confirm the absence of G cells (which reside only in the true
antrum), thus identifying the tissue as true body/fundus with atrophy. A chromogranin stain
highlights the final diagnostic feature of ECL-cell hyperplasia, either linear (arrowhead ) or
nodular (arrow).
Figure 2-222. Quick tutorial on the interpretation of AMAG. By H&E, the gastric antrum (top row)
is unremarkable or has chemical/reactive gastropathy, as seen here. The gastric body (bottom
row) contains red flags to further pursue an AMAG workup. The easiest red flag to spot is
intestinal metaplasia in a biopsy labeled as body/fundus. Other features include the absence of
normal oxyntic glands, a low-lying lymphocytic infiltrate, and pyloric gland metaplasia. Any
combination of these should prompt a basic AMAG workup, which includes gastrin and
chromogranin immunostains. In the antrum, gastrin and chromogranin both highlight the
horizontal band of gastrin-secreting G cells. In the body/fundus, the widespread absence of G
cells is expected and confirms the tissue source. Chromogranin highlights linear (arrow) and
nodular (arrowhead ) ECL-cell hyperplasia.
Figure 2-223. WD-NET arising in AMAG. At low magnification, this tumor is expansile and
composed of small nests of cells. The background mucosa appears atrophic with intestinal
metaplasia and pyloric-type glands.
Figure 2-224. WD-NET arising in AMAG. A chromogranin immunostain of the previous case
highlights the tumor cells, confirming neuroendocrine differentiation.
Figure 2-225. WD-NET arising in AMAG, higher magnification of the previous figure. The tumor
cells are uniform and arranged in small nests and cords.
Figure 2-226. WD-NET arising in AMAG. The mucosa adjacent to the tumor (right) provides
clues to the pathogenesis and prognosis. A specific combination of features offers a telltale
story of AMAG: complete atrophy of oxyntic glands, chronic inflammation, intestinal metaplasia,
and pyloric metaplasia. ECL-cell hyperplasia can be found on the chromogranin stain.
Figure 2-227. AMAG, background mucosa of the previous figure. The H&E features show a
complete absence of oxyntic glands (no chief cells or parietal cells), chronic inflammation,
intestinal metaplasia, and pyloric metaplasia. The last feature of AMAG (ECL-cell hyperplasia)
can be confirmed by chromogranin immunostain.
Figure 2-228. AMAG, linear and nodular ECL-cell hyperplasia, chromogranin stain. Linear and
nodular ECL-cell hyperplasia is defined as five or more adjacent chromogranin reactive cells.
Figure 2-229. AMAG, linear and nodular ECL-cell hyperplasia, chromogranin stain. Linear and
nodular ECL-cell hyperplasia is defined as five or more adjacent chromogranin reactive cells.
GASTRIC WELL-DIFFERENTIATED NEUROENDOCRINE
TUMOR, TYPE II
Type II NETs (Fig. 2.230) are rare and arise in the setting of ZE syndrome due to MEN1
syndrome or a gastrin-secreting tumor elsewhere in the GI tract. Similar to the
mechanism in AMAG, the uninhibited gastrin secretion stimulates ECL cells to
proliferate, resulting in WD-NETs (often multiple). These type II tumors have worse
prognosis than type I, with metastasis in about 30% of cases.122 However, type II tumors
behave distinctly better than type III tumors, again underscoring the importance of
differentiating WD-NETs, which can be achieved by reviewing tandem biopsies of the
nonpolypoid mucosa (Figs. 2.230–2.237). Biopsies of the background mucosa in ZE
syndrome show oxyntic gland hyperplasia (Fig. 2.231) (as compared with atrophy in
AMAG), and diffuse endocrine cell hyperplasia can be identified by
immunohistochemistry. Local resection of the NET, evaluation for metastatic disease,
and resection of the stimulatory gastrin-secreting tumor (usually found in the small
bowel) is the mainstay of therapy.122
Figure 2-230. Zollinger-Ellison syndrome in a patient with MEN1, endoscopic view. The gastric
folds are hypertrophic.
Figure 2-231. Gastric oxyntic mucosa of a patient with Zollinger-Ellison syndrome. These
patients have a gastrin-secreting tumor (gastrinoma), often found in the small bowel, and the
resulting hypergastrinemia causes direct stimulation of oxyntic mucosa to secrete copious
amounts of acid. Biopsies show hyperplastic oxyntic mucosa with proton pump inhibitor effect
(pictured); PPIs are prescribed to suppress acid secretion. Curative treatment relies on
identification and resection of the gastrinoma.
Figure 2-232. WD-NET arising in gastric oxyntic mucosa of a patient with Zollinger-Ellison
syndrome. Multiple nests of uniform cells are present between the oxyntic glands.
Figure 2-233. WD-NET arising in gastric oxyntic mucosa of a patient with Zollinger-Ellison
syndrome. Higher magnification of the previous figure shows uniform tumor cells without
prominent nucleoli.
Figure 2-234. WD-NET arising in gastric oxyntic mucosa of a patient with Zollinger-Ellison
syndrome. Biopsies submitted as gastric “nodules” show extensive involvement of the gastric
mucosa by WD-NET.
Figure 2-235. WD-NET arising in gastric oxyntic mucosa of a patient with Zollinger-Ellison
syndrome, chromogranin immunostain. The tumor cells are reactive for chromogranin.
Figure 2-236. WD-NET arising in gastric oxyntic mucosa of a patient with Zollinger-Ellison
syndrome. Another look at the tumor cells, which appear remarkably uniform.
Figure 2-237. WD-NET arising in gastric oxyntic mucosa of a patient with Zollinger-Ellison
syndrome, chromogranin immunostain.
Comment
Type I gastric WD-NETs arise in the setting of hypergastrinemia owing to AMAG and
are well-performing tumors. The rate of metastatic disease to lymph nodes or distant
sites is negligible and, if the lesion is amenable, conservative EMR is adequate
treatment. Patients with AMAG are at risk for pernicious anemia, dysplasia, and gastric
adenocarcinoma. Continued endoscopic surveillance is suggested, if clinically
appropriate.
FAQ: What is the size cutoff for a NET versus hyperplasia, and what is the
significance?
Size cutoffs for NETs vary based on published sources. For example:
College of American
Pathologists
PEARLS & PITFALLS: Grading of WD-NET Requires Both Mitotic Count and
Ki-67 Proliferative Index
WD-NETs are graded by both mitotic index and Ki-67 proliferation index (Figs.
2.242–2.244). The Ki-67 index frequently results in a higher grade than mitotic
count, and studies have shown these grade-discordant tumors more likely to
have metastases to lymph nodes and distant sites, perineural invasion, small
vessel invasion, and overall survival.125 In cases for which grade results are
discordant, assign the higher grade.
WHO 2017 Classification of
Neuroendocrine Neoplasms
FAQ: Can I eyeball the Ki-67 or does this require a manual count?
PEARLS & PITFALLS: Grading by Mitotic Rate Requires Counting 50 HPF But
Is Reported as per 10 HPF
The mitoses in 50 HPF should be counted to accurately grade the tumor. This
number is divided by five to report mitoses per 10 HPF. These minimum
requirements for grading (50 HPF for mitotic count and 5000 to 2000 cells for
Ki-67 index) presume there is enough tissue for accurate grading, but small
biopsy material may be insufficient in many cases. See the following sample
note.
Comment
There is insufficient tumor cell quantity to accurately grade this WD-NET (minimum
requirement 50 HPF for mitotic count and 500 cells for Ki-67 proliferation index).
Based on the available material, this tumor appears to be (G1, G2, G3). Final grading
will be revised following review of a larger sample (e.g., excision specimen).
PEARLS & PITFALLS: Both Size and Depth of Invasion Are Considered in
Staging of Gastric WD-NETs
Although the depth of invasion defines most staging criteria, gastric WD-NETs
are among the few that also take into account tumor size at the lower stages.
For example, staging criteria by depth is fairly typical, with invasion into the
lamina propria and submucosa staged as pT1, but only if the tumor is ≤1 cm
Any tumor >1 cm is automatically pT2 or higher, even if superficially invasive.
Once tumors invade at least into the muscularis propria, the tumors are staged
by depth regardless of size: pT2, involvement of muscularis propria, pT3,
involvement of subserosa, pT4, invasion of serosa or adjacent tissue/organs.
Figure 2-245. Manual count for Ki-67 proliferation index. A simple computer printout is quick and
economical if digital image analysis software is not available. Each cell can be marked off
during the 500–2000 cell count to avoid duplicate counts. To facilitate turnaround time, ancillary
staff can be trained in this method for reporting Ki-67 proliferation index.
MALT LYMPHOMA
GI lymphomas are challenging to recognize because the GI tract serves many
immunologic functions, and there is considerable histologic overlap between benign
inflammatory conditions and malignant lymphomas. In-depth coverage of lymphomas is
beyond the scope of this text and is left to our subspecialty hematopathology colleagues.
We encourage a low threshold to liberally share cases with such experts, but any
pathologist reviewing GI biopsies will be faced with gastric MALT extranodal marginal
zone lymphoma. The tools given in the following text are intended to ensure readers are
comfortable triaging these cases and are confident in recognizing features requiring
additional workup and consultation with hematopathology colleagues.
Gastric MALT lymphoma is driven by H. pylori infection, and eradication of the
organism is the first-line treatment of MALT lymphoma, resulting in remission in nearly
80% of cases.127 At low magnification, a robust and expansile deep chronic
inflammatory infiltrate is the first red flag to evaluate further for MALT lymphoma
(Figs. 2.246–2.250). At higher magnification, the infiltrate is typically composed of
monomorphic small lymphocytes with pericellular clearing or “halos” (Figs.
2.251–2.254). Features that serve as red flags to differentiate a malignant infiltrate from
benign gastritis include glandular destruction, in which lymphocytes (usually three or
more) invade the glandular epithelium and disrupt normal architecture (i.e.,
lymphoepithelial lesions) (Figs. 2.251–2.257) and the presence of dense lymphoid
infiltrates involving the muscularis mucosae (Figs. 2.246–2.248).128 These features
should trigger immunohistochemical workup, including, at minimum, CD3, CD20, and
CD43. This limited and economical panel can identify about half of MALT lymphomas,
which will show aberrant coexpression of CD43+ in the predominantly CD20+ B-cell
infiltrate (Fig. 2.258). The CD3 immunostain will provide a contrast by highlighting any
T cells, which normally express CD43. If this panel is insufficient to make a diagnosis,
expansion to a more comprehensive immunohistochemical panel will show the
following pattern in MALT lymphoma: CD20+, CD79a+, BCL2+, CD5−, CD10−,
cyclin D1−, CD23− CD43± (Figs. 2.259–2.264). This fundamental panel will also
differentiate other mature B-cell neoplasms, such as chronic lymphocytic
leukemia/small lymphocytic lymphoma (CD5+), follicular lymphoma (CD10+), and
mantle cell lymphoma (cyclin D1/BCL1+).
Figure 2-246. Gastric MALT lymphoma. Features that should trigger a MALT lymphoma workup
are seen here, including a deep monotonous lymphoid infiltrate that is gland destructive and
splaying out the muscularis mucosae.
Figure 2-247. Gastric MALT lymphoma. The infiltrate at low power is far more robust than
expected for a simple chronic gastritis. The lymphoid infiltrate is densely packed, gland
destructive, deep, and expanding the muscularis mucosae.
Figure 2-248. Gastric MALT lymphoma, higher magnification of the previous figure. The infiltrate
is deep, dense, and composed of a monotonous population of lymphocytes. The lymphoid cells
not only cross the muscularis mucosae but also spread the muscle bundles (arrow) apart.
Figure 2-249. Gastric MALT lymphoma. The infiltrate is more dense and monotonous than the
usual chronic gastritis. At low magnification, the expansile and gland-destructive quality should
trigger further workup for lymphoma.
Figure 2-250. Gastric MALT lymphoma, higher magnification of the previous figure. Gland
destruction is seen in the center of the field, as lymphocytes invade the glandular epithelium.
Figure 2-251. Gastric MALT lymphoma, higher magnification of the previous figure.
Lymphoepithelial lesions are characterized by lymphocytes (usually ≥3) invading the glandular
epithelium and disrupting the normal architecture (arrowhead ). Destroyed glands (arrow) leave
areas of drop-out which are filled in by the monotonous lymphocytes.
Figure 2-252. Lymphoepithelial lesion of gastric MALT lymphoma. The malignant lymphoid cells
invade the glandular epithelium and disrupt the normal architecture. A feature of the malignant
cells is the pericellular clearing or halo around each cell.
Figure 2-253. Gastric MALT lymphoma. These malignant lymphocytes are destroying areas of
glandular epithelium (arrowhead ) and muscularis mucosae (arrow). Features of glandular
destruction and muscularis mucosae abnormality are not seen in benign gastritis and should
prompt further workup for lymphoma.
Figure 2-254. Lymphoepithelial lesion of gastric MALT lymphoma. These lymphocytes have a
characteristic pericellular halo seen in MALT lymphoma cells. The presence of three or more
lymphocytes invading the glandular epithelium (arrow) is called a lymphoepithelial lesion (LEL).
Compared with benign intraepithelial lymphocytosis, which are T cells, these LELs are
composed of malignant B cells.
Figure 2-255. Gastric MALT lymphoma. At low magnification, this infiltrate differs from a benign
chronic gastritis because it is deep, dense, and monotonous with gland destruction.
Figure 2-256. Lymphoepithelial lesions in gastric MALT lymphoma, higher magnification of the
previous figure. Lymphoepithelial lesions are seen in various stages (arrows). On the far left,
the gland structure is still visible. The far right shows marked disruption of the glandular
architecture, but remnant epithelial cells are still visible. The center lesion is a nearly destroyed
gland and is barely visible.
Figure 2-257. Gastric MALT lymphoma, pancytokeratin stain. A pancytokeratin stain can be
helpful in highlighting residual glands and areas of lymphoepithelial lesions, which may be
obscured by the dense lymphocytic infiltrate on H&E.
Figure 2-258. Gastric MALT lymphoma, CD20+ with coexpression of CD43+. About half of
gastric MALT lymphomas can be identified by a limited immunopanel of CD3, CD20, and CD43.
These tumors show aberrant coexpression of CD43+ (normally found in T cells) in the
predominantly CD20+ B-cell infiltrate. The CD3 immunostain provides a contrast by highlighting
the T cells. Should this panel fail to solidify a diagnosis, an extended immunopanel can be
performed.
Figure 2-259. Gastric MALT lymphoma. This infiltrate is just a little too dense and too
monotonous to consider chronic gastritis. Some areas appear expansile, whereas others
appear gland destructive. In these instances, it is best to rule out lymphoma.
Figure 2-260. Gastric MALT lymphoma, CD43−, CD20+ with coexpression of BCL2. As noted
earlier, about half of MALT lymphomas do not express CD43. An extended panel of
immunostains will show reactivity for BCL2+ in the CD20-positive B cells.
Figure 2-261. Lymphoepithelial lesions in gastric MALT lymphoma. Lymphoepithelial lesions can
be found in various stages. Early lesions show ≥3 tumor lymphocytes invading the glandular
epithelium (arrow). More mature lesions show disruption of glandular architecture with
degenerating epithelial cells (arrowhead ).
Figure 2-262. Plasmacytoid variant of gastric MALT lymphoma. At low power, the architecture of
this gastric biopsy is abnormal. The lamina propria appears expanded and cellular, whereas the
glands are irregularly distributed. The pigment is incidental hemosiderin.
Figure 2-263. Plasmacytoid variant of gastric MALT lymphoma, higher magnification of the
previous figure. Do not be falsely reassured by the presence of a plasmacytic infiltrate in this
case. The plasma cells in the lamina propria are atypical with binucleate forms and marked
variation in size. A plasmacytic clone is found in 30% of MALT lymphomas. The pigment is
incidental hemosiderin.
Figure 2-264. Plasmacytoid variant of gastric MALT lymphoma, higher magnification of the
previous figure. These subtle MALT lymphomas can be CD20 negative but should express
CD79a and show kappa or lambda restriction. This example shows CD20−, CD79a+, and
lambda restriction.
FAQ: How does one handle biopsies for posttreatment assessment of MALT
lymphoma?
Reference:
Copie-Bergman C, Gaulard P, Lavergne-Slove A, et al. Proposal for a new histological
grading system for post-treatment evaluation of gastric MALT lymphoma. Gut.
2003;52(11):1656. PMID:14570741; PMCID:PMC1773845.
Figure 2-267. Probable minimal residual disease. Sometimes residual lymphoid cells are
present in posttreatment biopsies yet are insufficient in size to immunophenotype. These can
simply be reported as probable minimal residual disease, which is managed clinically as a
state of remission.
MESENCHYMAL LESIONS
Mesenchymal lesions cover a broad spectrum of mesodermally derived tumors, which
are covered more completely in “Mesenchymal Tumors” chapter. Select mesenchymal
polyps common to the stomach are briefly covered herein, including the inflammatory
fibroid polyp (IFP), gastrointestinal stromal tumor (GIST), leiomyoma, and granular
cell tumor (GCT).
Figure 2-268. Inflammatory fibroid polyp. The most common location for IFP is the gastric
antrum, as seen here. The epicenter of these lesions is submucosal, and they appear
endoscopically as a nodule.
Figure 2-269. Inflammatory fibroid polyp, CD34 immunohistochemistry of the previous figure. A
CD34 highlights the scope of this benign lesion, which is surprisingly more extensive than
appreciated on H&E.
Figure 2-270. Inflammatory fibroid polyp. The spindle cells extend from the submucosa and
percolate through the lamina propria toward the surface. However, the findings are subtle and
one can appreciate how a superficial biopsy might be challenging to interpret. Diagnostic clues
include unexplained bland spindle cells and the presence of eosinophils.
Figure 2-271. Inflammatory fibroid polyp. The spindle cells may swirl concentrically around
vessels in an onion skin pattern, as seen here. As the spindle cells extend upward, they
traverse the muscularis mucosae and splay the muscle fibers.
Figure 2-272. Inflammatory fibroid polyp, CD34 immunohistochemistry of the previous figure.
CD34 highlights the spindle cells of the IFP, which are more abundant than appreciated by
H&E.
Figure 2-273. Inflammatory fibroid polyp. Higher magnification shows the bland spindle cells
forming a concentric pattern around the artery. Intimately admixed are frequent eosinophils, an
extremely helpful diagnostic clue to this entity.
Figure 2-274. Inflammatory fibroid polyp. Some IFPs appear more edematous or myxoid. In
these areas, the spindle cells (arrows) may be sparse. Often the first clue to diagnosis is the
eye-catching eosinophils.
LEIOMYOMA
Leiomyomas are benign smooth muscle tumors, typically asymptomatic and found
incidentally.154-156 Endoscopically they appear as rounded submucosal lesions with
intact overlying mucosa (Fig. 2.281) and range in size from 0.5 to 20 cm.157 Both
leiomyomas and GISTs can grow inwardly and outwardly to form a dumbbell shape,
although leiomyomas are more likely to grow intraluminally (vs. GIST, which expands
predominantly in an extramural fashion). Histologically, the tumor is composed of
intersecting bundles of smooth muscle without atypia, frequent mitotic activity, or
necrosis (Figs. 2.282–2.284). The tumor can be differentiated from GIST, which is
CD117 or DOG-1 positive, whereas leiomyoma is smooth muscle actin and desmin
positive and negative for CD117/DOG-1.
Figure 2-275. Gastrointestinal stromal tumor (GIST). These lesions derive from the interstitial
cells of Cajal and arise almost exclusively from the myenteric (Auerbach) plexus, which is
located between the inner circular and outer longitudinal layers of the muscularis propria
(pictured). For this reason, a spindle cell lesion arising from the muscularis mucosae cannot be
a GIST.
Figure 2-276. Gastrointestinal stromal tumor (GIST), CD117 immunohistochemistry of the
previous figure. CD117 immunoreactivity confirms the diagnosis in 95% of cases. CD117-
negative GISTs can be stained for DOG1.
Figure 2-277. GIST, epithelioid type. The cells are round and fairly uniform. There is no
prognostic significance to the morphologic variant.
Figure 2-278. GIST, spindled type. These cells are elongated with cigar-shaped nuclei.
Figure 2-279. GIST. These tumors can take on many morphologic variations and are wonderful
mimickers of other tumors. This region shows features similar to Verocay bodies found in
schwannomas.
Figure 2-280. GIST. Perpendicular fascicles of spindle cells raise the differential for leiomyoma.
GISTs are excellent mimickers of other tumors.
Figure 2-281. Leiomyoma. This spindle cell neoplasm arises from the muscularis mucosae.
Because of this location, the diagnosis cannot be GIST.
Figure 2-282. Leiomyoma, higher magnification of the previous figure. The tumor arises from
the muscularis mucosae, and the cells are bland and spindled.
Figure 2-283. Leiomyoma, smooth muscle actin (SMA) immunohistochemistry. SMA confirms
the smooth muscle differentiation of this tumor.
Figure 2-284. Leiomyoma. The smooth muscle bundles characteristically intersect at
perpendicular angles.
Figure 2-285. Granular cell tumor. The tumor cells have indistinct cell borders and slightly
atypical angulated nuclei. There is abundant eosinophilic and granular cytoplasm.
Figure 2-286. Granular cell tumor. Oil immersion shows the granular quality of the cytoplasm,
which by electron microscopy is filled with lysosomes (not pictured).
NEAR MISS
METASTATIC LOBULAR BREAST CARCINOMA
New diagnoses of diffuse-type gastric cancer in women should include at least one
immunohistochemical marker to exclude metastatic lobular breast carcinoma, such as
GATA3. Although breast cancer metastasis to the GI tract is a rare occurrence, the
stomach is the most common location aside from the liver, and the discohesive
infiltrating cells of lobular breast carcinoma can be easily mistaken for a primary
diffuse-type gastric cancer (Figs. 2.287–2.294). Differentiation of these tumors relies
almost entirely upon immunohistochemical confirmation, an important step because the
treatment of these tumors diverge. A diagnosis of primary gastric cancer leads to
surgical management, whereas metastatic breast cancer may benefit from
chemotherapeutic options depending on the hormone receptor status. Histologically,
metastatic lobular breast cancer cells infiltrate the lamina propria individually or in
single-file cords (Fig. 2.288). These uniform small discohesive cells characteristically
lack E-cadherin (Fig. 2.289), a surface cohesion molecule. Because diffuse-type gastric
cancer lacks a precursor lesion, this metastatic pattern is nearly indistinguishable from a
primary gastric tumor. A PAS stain is negative for cytoplasmic PAS staining or
intracytoplasmic mucin (Fig. 2.290), whereas most gastric signet ring cell carcinomas
will show PAS staining. One helpful clue to metastatic lobular breast carcinoma is the
presence of intracytoplasmic lumina, which have a sharply demarcated edge and may
contain a hyaline globule imparting a targetoid appearance (Fig. 2.294).
GASTRIC XANTHOMA
These subepithelial aggregates of histiocytes are submitted as endoscopic nodules,
polyps, or plaques and cause no diagnostic difficulty for pathologists when encountered
in the gallbladder (i.e., cholesterolosis) but can prove tricky when seen in the stomach
(Figs. 2.295–2.302). The most common concern among extramural consultations is
exclusion of diffuse-type gastric cancer (Figs. 2.295–2.297). At low magnification, an
area of pallor is eye-catching as the foamy histiocytes expand the lamina propria (Fig.
2.298). Collections of bland macrophages with abundant foamy cytoplasm are usually
subepithelial but can be found anywhere within the tissue (Fig. 2.299 and 2.301). Small
and crushed biopsies provide the most challenging material, but application of CD68
immunohistochemistry is almost always helpful (Figs. 2.297 and 2.302). PAS stain is
negative for intracytoplasmic mucin (Fig. 2.300).
Figure 2-295. Gastric xanthoma in a crushed and suboptimal biopsy. By H&E, several cells in
this biopsy are hard to name (arrows). They are subepithelial in the lamina propria and contain
clear cytoplasm. Small and crushed biopsies are always difficult to interpret, and diffuse-type
gastric cancer raises the stakes even further. Do not hesitate to request repeat biopsy if the
diagnosis is unclear.
Figure 2-296. Gastric xanthoma, pancytokeratin immunostain of the previous figure. A
pancytokeratin stain can highlight the gastric foveolar and glandular architecture and provide
reassurance that the cells are not invasive carcinoma cells.
Figure 2-297. Gastric xanthoma, CD68 immunostain. A CD68 immunostain in this example
highlights the scant crushed cells, confirming they are foamy macrophages and not signet ring
cells.
Figure 2-298. Gastric xanthoma. Gastric xanthoma cells can mimic a diffuse-type gastric
cancer at low magnification. All areas of increased cellularity or pallor (arrow) should be
reviewed more closely to exclude a sneaky diffuse-type gastric cancer.
Figure 2-299. Gastric xanthoma, higher magnification of the previous figure. Gastric xanthomas
are composed of collections of foamy macrophages. At high magnification, they have bland
uniform nuclei and abundant foamy cytoplasm that makes them, in most cases, easy to
distinguish from diffuse gastric cancer by H&E alone.
Figure 2-300. Gastric xanthoma, PAS stain. The cytoplasm of foamy macrophages is not PAS
positive and does not contain mucin.
Figure 2-301. Gastric xanthoma. This example is more challenging, as the foamy macrophages
(arrow) are embedded within the muscularis mucosa and raise concern for a sneaky invasive
diffuse-type gastric cancer.
Figure 2-302. Gastric xanthoma, CD68 immunostain of the previous figure.
Immunohistochemistry confirms histiocytic differentiation and reassures that these are benign
cells.
References
1. Carmack SW, Genta RM, Schuler CM, Saboorian MH. The current spectrum of
gastric polyps: a 1-year national study of over 120,000 patients. Am J
Gastroenterol. 2009;104(6):1524-1532.
2. Jhala NC, Montemor M, Jhala D, et al. Pancreatic acinar cell metaplasia in
autoimmune gastritis. Arch Pathol Lab Med. 2003;127(7):854-857.
3. Lam-Himlin D, Park JY, Cornish TC, Shi C, Montgomery E. Morphologic
characterization of syndromic gastric polyps. Am J Surg Pathol.
2010;34(11):1656-1662.
4. Beggs AD, Latchford AR, Vasen HF, et al. Peutz-Jeghers syndrome: a systematic
review and recommendations for management. Gut. 2010;59(7):975-986.
5. Hemminki A, Markie D, Tomlinson I, et al. A serine/threonine kinase gene
defective in Peutz-Jeghers syndrome. Nature. 1998;391(6663):184-187.
6. Volikos E, Robinson J, Aittomaki K, et al. LKB1 exonic and whole gene deletions
are a common cause of Peutz-Jeghers syndrome. J Med Genet. 2006;43(5):e18.
7. Giardiello FM, Welsh SB, Hamilton SR, et al. Increased risk of cancer in the
Peutz-Jeghers syndrome. N Engl J Med. 1987;316(24):1511-1514.
8. Hearle N, Schumacher V, Menko FH, et al. Frequency and spectrum of cancers in
the Peutz-Jeghers syndrome. Clin Cancer Res. 2006;12(10):3209-3215.
9. Jeghers H, Mc KV, Katz KH. Generalized intestinal polyposis and melanin spots of
the oral mucosa, lips and digits; a syndrome of diagnostic significance. N Engl J
Med. 1949;241(26):1031-1036.
10. Offerhaus GJA BM, Gruber SB. Peutz-Jeghers syndrome. In: Bosman FT, Carneiro
F, Hruban RH, et al, eds. Tumors of the Digestive System. Lyon, France: IACR;
2010:160-170.
11. Latchford AR, Phillips RK. Gastrointestinal polyps and cancer in Peutz-Jeghers
syndrome: clinical aspects. Fam Cancer. 2011;10(3):455-461.
12. Tse JY, Wu S, Shinagare SA, et al. Peutz-Jeghers syndrome: a critical look at
colonic Peutz-Jeghers polyps. Mod Pathol. 2013;26(9):1235-1240.
13. Giardiello FM, Brensinger JD, Tersmette AC, et al. Very high risk of cancer in
familial Peutz-Jeghers syndrome. Gastroenterology. 2000;119(6):1447-1453.
14. Burkart AL, Sheridan T, Lewin M, Fenton H, Ali NJ, Montgomery E. Do sporadic
Peutz-Jeghers polyps exist? Experience of a large teaching hospital. Am J Surg
Pathol. 2007;31(8):1209-1214.
15. Burt RW, Bishop DT, Lynch HT, Rozen P, Winawer SJ. Risk and surveillance of
individuals with heritable factors for colorectal cancer. WHO Collaborating
Centre for the Prevention of Colorectal Cancer. Bull World Health Organ.
1990;68(5):655-665.
16. Howe JR, Roth S, Ringold JC, et al. Mutations in the SMAD4/DPC4 gene in
juvenile polyposis. Science. 1998;280(5366):1086-1088.
17. Sweet K, Willis J, Zhou XP, et al. Molecular classification of patients with
unexplained hamartomatous and hyperplastic polyposis. JAMA.
2005;294(19):2465-2473.
18. Howe JR, Mitros FA, Summers RW. The risk of gastrointestinal carcinoma in
familial juvenile polyposis. Ann Surg Oncol. 1998;5(8):751-756.
19. Giardiello FM, Hamilton SR, Kern SE, et al. Colorectal neoplasia in juvenile
polyposis or juvenile polyps. Arch Dis Child. 1991;66(8):971-975.
20. Offerhaus GJA HJ. Juvenile polyposis. In: Bosman FT, Carneiro F, Hruban RH, et
al, eds. WHO Classification of Tumors of the Digestive System. Lyon, France:
IARC; 2010:166-167.
21. Cronkhite LW Jr, Canada WJ. Generalized gastrointestinal polyposis; an unusual
syndrome of polyposis, pigmentation, alopecia and onychotrophia. N Engl J Med.
1955;252(24):1011-1015.
22. Nakamura M, Kobashikawa K, Tamura J, et al. Cronkhite-Canada syndrome. Intern
Med. 2009;48(17):1561-1562.
23. Daniel ES, Ludwig SL, Lewin KJ, Ruprecht RM, Rajacich GM, Schwabe AD. The
Cronkhite-Canada syndrome. An analysis of clinical and pathologic features and
therapy in 55 patients. Medicine (Baltim). 1982;61(5):293-309.
24. Heald B, Mester J, Rybicki L, Orloff MS, Burke CA, Eng C. Frequent
gastrointestinal polyps and colorectal adenocarcinomas in a prospective series of
PTEN mutation carriers. Gastroenterology. 2010;139(6):1927-1933.
25. Kato M, Mizuki A, Hayashi T, et al. Cowden’s disease diagnosed through
mucocutaneous lesions and gastrointestinal polyposis with recurrent hematochezia,
unrevealed by initial diagnosis. Intern Med. 2000;39(7):559-563.
26. Levi Z, Baris HN, Kedar I, et al. Upper and lower gastrointestinal findings in
PTEN mutation-positive Cowden syndrome patients participating in an active
surveillance program. Clin Transl Gastroenterol. 2011;2:e5.
27. Tan MH, Mester JL, Ngeow J, Rybicki LA, Orloff MS, Eng C. Lifetime cancer
risks in individuals with germline PTEN mutations. Clin Canc Res.
2012;18(2):400-407.
28. Abraham SC, Singh VK, Yardley JH, Wu TT. Hyperplastic polyps of the stomach:
associations with histologic patterns of gastritis and gastric atrophy. Am J Surg
Pathol. 2001;25(4):500-507.
29. Koch HK, Lesch R, Cremer M, Oehlert W. Polyps and polypoid foveolar
hyperplasia in gastric biopsy specimens and their precancerous prevalence. Front
Gastrointest Res. 1979;4:183-191.
30. Koga S, Watanabe H, Enjoji M. Stomal polypoid hypertrophic gastritis: a polypoid
gastric lesion at gastroenterostomy site. Cancer. 1979;43(2):647-657.
31. Zea-Iriarte WL, Sekine I, Itsuno M, et al. Carcinoma in gastric hyperplastic polyps.
A phenotypic study. Dig Dis Sci. 1996;41(2):377-386.
32. Ginsberg GG, Al-Kawas FH, Fleischer DE, Reilly HF, Benjamin SB. Gastric
polyps: relationship of size and histology to cancer risk. Am J Gastroenterol.
1996;91(4):714-717.
33. Orlowska J, Jarosz D, Pachlewski J, Butruk E. Malignant transformation of benign
epithelial gastric polyps. Am J Gastroenterol. 1995;90(12):2152-2159.
34. Genta RM, Schuler CM, Robiou CI, Lash RH. No association between gastric
fundic gland polyps and gastrointestinal neoplasia in a study of over 100,000
patients. Clin Gastroenterol Hepatol. 2009;7(8):849-854.
35. Zelter A, Fernandez JL, Bilder C, et al. Fundic gland polyps and association with
proton pump inhibitor intake: a prospective study in 1,780 endoscopies. Dig Dis
Sci. 2011;56(6):1743-1748.
36. Vieth M, Stolte M. Fundic gland polyps are not induced by proton pump inhibitor
therapy. Am J Clin Pathol. 2001;116(5):716-720.
37. Bianchi LK, Burke CA, Bennett AE, Lopez R, Hasson H, Church JM. Fundic gland
polyp dysplasia is common in familial adenomatous polyposis. Clin Gastroenterol
Hepatol. 2008;6(2):180-185.
38. Garrean S, Hering J, Saied A, Jani J, Espat NJ. Gastric adenocarcinoma arising
from fundic gland polyps in a patient with familial adenomatous polyposis
syndrome. Am Surg. 2008;74(1):79-83.
39. Zwick A, Munir M, Ryan CK, et al. Gastric adenocarcinoma and dysplasia in
fundic gland polyps of a patient with attenuated adenomatous polyposis coli.
Gastroenterology. 1997;113(2):659-663.
40. Wu TT, Kornacki S, Rashid A, Yardley JH, Hamilton SR. Dysplasia and
dysregulation of proliferation in foveolar and surface epithelia of fundic gland
polyps from patients with familial adenomatous polyposis. Am J Surg Pathol.
1998;22(3):293-298.
41. Offerhaus GJ, Giardiello FM, Krush AJ, et al. The risk of upper gastrointestinal
cancer in familial adenomatous polyposis. Gastroenterology. 1992;102(6):1980-
1982.
42. Torbenson M, Lee JH, Cruz-Correa M, et al. Sporadic fundic gland polyposis: a
clinical, histological, and molecular analysis. Mod Pathol. 2002;15(7):718-723.
43. Li J, Woods SL, Healey S, et al. Point mutations in exon 1B of APC reveal gastric
adenocarcinoma and proximal polyposis of the stomach as a familial adenomatous
polyposis variant. Am J Hum Genet. 2016;98(5):830-842.
44. Abraham SC, Nobukawa B, Giardiello FM, Hamilton SR, Wu TT. Sporadic fundic
gland polyps: common gastric polyps arising through activating mutations in the
beta-catenin gene. Am J Pathol. 2001;158(3):1005-1010.
45. Lauwers GY, Shimizu M, Correa P, et al. Evaluation of gastric biopsies for
neoplasia: differences between Japanese and Western pathologists. Am J Surg
Pathol. 1999;23(5):511-518.
46. Schlemper RJ, Riddell RH, Kato Y, et al. The Vienna classification of
gastrointestinal epithelial neoplasia. Gut. 2000;47(2):251-255.
47. Singhi AD, Lazenby AJ, Montgomery EA. Gastric adenocarcinoma with chief cell
differentiation: a proposal for reclassification as oxyntic gland polyp/adenoma.
Am J Surg Pathol. 2012;36(7):1030-1035.
48. Vieth M, Kushima R, Borchard F, Stolte M. Pyloric gland adenoma: a clinico-
pathological analysis of 90 cases. Virchows Arch. 2003;442(4):317-321.
49. Abraham SC, Montgomery EA, Singh VK, Yardley JH, Wu TT. Gastric adenomas:
intestinal-type and gastric-type adenomas differ in the risk of adenocarcinoma and
presence of background mucosal pathology. Am J Surg Pathol. 2002;26(10):1276-
1285.
50. Chen ZM, Scudiere JR, Abraham SC, Montgomery E. Pyloric gland adenoma: an
entity distinct from gastric foveolar type adenoma. Am J Surg Pathol.
2009;33(2):186-193.
51. Abraham SC, Park SJ, Lee JH, Mugartegui L, Wu TT. Genetic alterations in gastric
adenomas of intestinal and foveolar phenotypes. Mod Pathol. 2003;16(8):786-795.
52. Spigelman AD, Williams CB, Talbot IC, Domizio P, Phillips RK. Upper
gastrointestinal cancer in patients with familial adenomatous polyposis. Lancet.
1989;2(8666):783-785.
53. Bakotic BW, Robinson MJ, Sturm PD, Hruban RH, Offerhaus GJ, Albores-
Saavedra J. Pyloric gland adenoma of the main pancreatic duct. Am J Surg Pathol.
1999;23(2):227-231.
54. Hackeng WM, Montgomery EA, Giardiello FM, et al. Morphology and genetics of
pyloric gland adenomas in familial adenomatous polyposis. Histopathology.
2017;70(4):549-557.
55. Hashimoto T, Ogawa R, Matsubara A, et al. Familial adenomatous polyposis-
associated and sporadic pyloric gland adenomas of the upper gastrointestinal tract
share common genetic features. Histopathology. 2015;67(5):689-698.
56. Choi WT, Brown I, Ushiku T, et al. Gastric pyloric gland adenoma: a multicentre
clinicopathological study of 67 cases. Histopathology. 2018;72(6):1007-1014.
57. Wood LD, Salaria SN, Cruise MW, Giardiello FM, Montgomery EA. Upper GI
tract lesions in familial adenomatous polyposis (FAP): enrichment of pyloric gland
adenomas and other gastric and duodenal neoplasms. Am J Surg Pathol.
2014;38(3):389-393.
58. Ueyama H, Yao T, Nakashima Y, et al. Gastric adenocarcinoma of fundic gland
type (chief cell predominant type): proposal for a new entity of gastric
adenocarcinoma. Am J Surg Pathol. 2010;34(5):609-619.
59. Matsukawa A, Kurano R, Takemoto T, Kagayama M, Ito T. Chief cell hyperplasia
with structural and nuclear atypia: a variant of fundic gland polyp. Pathol Res
Pract. 2005;200(11-12):817-821.
60. Muller-Hocker J, Rellecke P. Chief cell proliferation of the gastric mucosa
mimicking early gastric cancer: an unusual variant of fundic gland polyp. Virchows
Arch. 2003;442(5):496-500.
61. Kushima R, Sekine S, Matsubara A, Taniguchi H, Ikegami M, Tsuda H. Gastric
adenocarcinoma of the fundic gland type shares common genetic and phenotypic
features with pyloric gland adenoma. Pathol Int. 2013;63(6):318-325.
62. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J
Clin. 2005;55(2):74-108.
63. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer
statistics. CA Cancer J Clin. 2011;61(2):69-90.
64. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin.
2017;67(1):7-30.
65. Ahn HS, Lee HJ, Yoo MW, et al. Changes in clinicopathological features and
survival after gastrectomy for gastric cancer over a 20-year period. Br J Surg.
2011;98(2):255-260.
66. Brown LM, Devesa SS. Epidemiologic trends in esophageal and gastric cancer in
the United States. Surg Oncol Clin N Am. 2002;11(2):235-256.
67. Amin MB, Greene FL, Edge SB, et al. The Eighth Edition AJCC Cancer Staging
Manual: continuing to build a bridge from a population-based to a more
“personalized” approach to cancer staging. CA Cancer J Clin. 2017;67(2):93-99.
68. Park JC, Lee YK, Kim SY, et al. Long-term outcomes of endoscopic submucosal
dissection in comparison to surgery in undifferentiated-type intramucosal gastric
cancer using propensity score analysis. Surg Endosc. 2018;32(4):2046-2057.
69. Lauren P. The two histological main types of gastric carcinoma: diffuse and so-
called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta
Pathol Microbiol Scand. 1965;64:31-49.
70. Cimerman M, Repse S, Jelenc F, Omejc M, Bitenc M, Lamovec J. Comparison of
Lauren’s, Ming’s and WHO histological classifications of gastric cancer as a
prognostic factor for operated patients. Int Surg. 1994;79(1):27-32.
71. Bosman FT, Carneiro F, Hruban RH, et al., ed WHO Classification of Tumours of
the Digestive System. 4th ed. Lyon: WHO Press; 2010.
72. Talamonti MS, Kim SP, Yao KA, et al. Surgical outcomes of patients with gastric
carcinoma: the importance of primary tumor location and microvessel invasion.
Surgery. 2003;134(4):720-727; discussion 727-729.
73. Luebke T, Baldus SE, Grass G, et al. Histological grading in gastric cancer by
Ming classification: correlation with histopathological subtypes, metastasis, and
prognosis. World J Surg. 2005;29(11):1422-1427; discussion 1428.
74. van Beek J, zur Hausen A, Klein Kranenbarg E, et al. EBV-positive gastric
adenocarcinomas: a distinct clinicopathologic entity with a low frequency of
lymph node involvement. J Clin Oncol. 2004;22(4):664-670.
75. Zur Hausen A, van Rees BP, van Beek J, et al. Epstein-Barr virus in gastric
carcinomas and gastric stump carcinomas: a late event in gastric carcinogenesis. J
Clin Pathol. 2004;57(5):487-491.
76. Ushiku T, Shinozaki A, Shibahara J, et al. SALL4 represents fetal gut
differentiation of gastric cancer, and is diagnostically useful in distinguishing
hepatoid gastric carcinoma from hepatocellular carcinoma. Am J Surg Pathol.
2010;34(4):533-540.
77. Yong KJ, Gao C, Lim JS, et al. Oncofetal gene SALL4 in aggressive hepatocellular
carcinoma. N Engl J Med. 2013;368(24):2266-2276.
78. Tang LH, Untch BR, Reidy DL, et al. Well-differentiated neuroendocrine tumors
with a morphologically apparent high-grade component: a pathway distinct from
poorly differentiated neuroendocrine carcinomas. Clin Cancer Res.
2016;22(4):1011-1017.
79. Yachida S, Vakiani E, White CM, et al. Small cell and large cell neuroendocrine
carcinomas of the pancreas are genetically similar and distinct from well-
differentiated pancreatic neuroendocrine tumors. Am J Surg Pathol.
2012;36(2):173-184.
80. Basturk O, Yang Z, Tang LH, et al. The high-grade (WHO G3) pancreatic
neuroendocrine tumor category is morphologically and biologically heterogenous
and includes both well differentiated and poorly differentiated neoplasms. Am J
Surg Pathol. 2015;39(5):683-690.
81. Raj N, Valentino E, Capanu M, et al. Treatment response and outcomes of grade 3
pancreatic neuroendocrine neoplasms based on morphology: well differentiated
versus poorly differentiated. Pancreas. 2017;46(3):296-301.
82. Sorbye H, Welin S, Langer SW, et al. Predictive and prognostic factors for
treatment and survival in 305 patients with advanced gastrointestinal
neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Ann Oncol.
2013;24(1):152-160.
83. Lloyd RV OR, Kloppel G, Rosai J. WHO Classification of Tumours of Endocrine
Organs. Vol 10. 4th ed. Lyon: IARC; 2017.
84. Fennerty MB. Gastric intestinal metaplasia on routine endoscopic biopsy.
Gastroenterology. 2003;125(2):586-590.
85. Correa P. Human gastric carcinogenesis: a multistep and multifactorial process–
first American Cancer Society Award Lecture on cancer epidemiology and
prevention. Cancer Res. 1992;52(24):6735-6740.
86. Correa P, Haenszel W, Cuello C, Tannenbaum S, Archer M. A model for gastric
cancer epidemiology. Lancet. 1975;2(7924):58-60.
87. Park JY, Cornish TC, Lam-Himlin D, Shi C, Montgomery E. Gastric lesions in
patients with autoimmune metaplastic atrophic gastritis (AMAG) in a tertiary care
setting. Am J Surg Pathol. 2010;34(11):1591-1598.
88. Coggon D, Barker DJ, Cole RB, Nelson M. Stomach cancer and food storage. J
Natl Cancer Inst. 1989;81(15):1178-1182.
89. La Vecchia C, Negri E, D’Avanzo B, Franceschi S. Electric refrigerator use and
gastric cancer risk. Br J Cancer. 1990;62(1):136-137.
90. Tricker AR. N-nitroso compounds and man: sources of exposure, endogenous
formation and occurrence in body fluids. Eur J Cancer Prev. 1997;6(3):226-268.
91. Song P, Wu L, Guan W. Dietary nitrates, nitrites, and nitrosamines intake and the
risk of gastric cancer: a meta-analysis. Nutrients. 2015;7(12):9872-9895.
92. La Vecchia C, Ferraroni M, D’Avanzo B, Decarli A, Franceschi S. Selected
micronutrient intake and the risk of gastric cancer. Cancer Epidemiol Biomarkers
Prev. 1994;3(5):393-398.
93. Palli D, Russo A, Decarli A. Dietary patterns, nutrient intake and gastric cancer in
a high-risk area of Italy. Cancer Causes Control. 2001;12(2):163-172.
94. Hansford S, Kaurah P, Li-Chang H, et al. Hereditary diffuse gastric cancer
syndrome: CDH1 mutations and beyond. JAMA Oncol. 2015;1(1):23-32.
95. van der Post RS, Vogelaar IP, Carneiro F, et al. Hereditary diffuse gastric cancer:
updated clinical guidelines with an emphasis on germline CDH1 mutation carriers.
J Med Genet. 2015;52(6):361-374.
96. Fujita H, Lennerz JK, Chung DC, et al. Endoscopic surveillance of patients with
hereditary diffuse gastric cancer: biopsy recommendations after topographic
distribution of cancer foci in a series of 10 CDH1-mutated gastrectomies. Am J
Surg Pathol. 2012;36(11):1709-1717.
97. Lee AF, Rees H, Owen DA, Huntsman DG. Periodic acid-schiff is superior to
hematoxylin and eosin for screening prophylactic gastrectomies from CDH1
mutation carriers. Am J Surg Pathol. 2010;34(7):1007-1013.
98. Barber ME, Save V, Carneiro F, et al. Histopathological and molecular analysis of
gastrectomy specimens from hereditary diffuse gastric cancer patients has
implications for endoscopic surveillance of individuals at risk. J Pathol.
2008;216(3):286-294.
99. Worthley DL, Phillips KD, Wayte N, et al. Gastric adenocarcinoma and proximal
polyposis of the stomach (GAPPS): a new autosomal dominant syndrome. Gut.
2012;61(5):774-779.
100. Yanaru-Fujisawa R, Nakamura S, Moriyama T, et al. Familial fundic gland
polyposis with gastric cancer. Gut. 2012;61(7):1103-1104.
101. McDuffie LA, Sabesan A, Allgaeuer M, et al. beta-Catenin activation in fundic
gland polyps, gastric cancer and colonic polyps in families afflicted by ‘gastric
adenocarcinoma and proximal polyposis of the stomach’ (GAPPS). J Clin Pathol.
2016;69(9):826-833.
102. Repak R, Kohoutova D, Podhola M, et al. The first European family with gastric
adenocarcinoma and proximal polyposis of the stomach: case report and review of
the literature. Gastrointest Endosc. 2016;84(4):718-725.
103. Guan J, Lim KS, Mekhail T, Chang CC. Programmed death Ligand-1 (PD-L1)
expression in the programmed death receptor-1 (PD-1)/PD-L1 blockade: a key
player against various cancers. Arch Pathol Lab Med. 2017;141(6):851-861.
104. Akiyama T, Sudo C, Ogawara H, Toyoshima K, Yamamoto T. The product of the
human c-erbB-2 gene: a 185-kilodalton glycoprotein with tyrosine kinase activity.
Science. 1986;232(4758):1644-1646.
105. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human
breast cancer: correlation of relapse and survival with amplification of the HER-
2/neu oncogene. Science. 1987;235(4785):177-182.
106. Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z, Kurzrock R. HER2
expression status in diverse cancers: review of results from 37,992 patients.
Cancer Metastasis Rev. 2015;34(1):157-164.
107. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with
chemotherapy versus chemotherapy alone for treatment of HER2-positive
advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-
label, randomised controlled trial. Lancet. 2010;376(9742):687-697.
108. Bartley AN, Washington MK, Ventura CB, et al. HER2 testing and clinical decision
making in gastroesophageal adenocarcinoma: guideline from the College of
American Pathologists, American Society for Clinical Pathology, and American
Society of Clinical Oncology. Arch Pathol Lab Med. 2016;140(12):1345-1363.
109. Cruz-Reyes C, Gamboa-Dominguez A. HER2 amplification in gastric cancer is a
rare event restricted to the intestinal phenotype. Int J Surg Pathol. 2013;21(3):240-
246.
110. Kunz PL, Mojtahed A, Fisher GA, et al. HER2 expression in gastric and
gastroesophageal junction adenocarcinoma in a US population: clinicopathologic
analysis with proposed approach to HER2 assessment. Appl Immunohistochem
Mol Morphol. 2012;20(1):13-24.
111. Park JS, Rha SY, Chung HC, et al. Clinicopathological features and prognostic
significance of HER2 expression in gastric cancer. Oncology. 2015;88(3):147-
156.
112. Okines AF, Thompson LC, Cunningham D, et al. Effect of HER2 on prognosis and
benefit from peri-operative chemotherapy in early oesophago-gastric
adenocarcinoma in the MAGIC trial. Ann Oncol. 2013;24(5):1253-1261.
113. Janjigian YY, Werner D, Pauligk C, et al. Prognosis of metastatic gastric and
gastroesophageal junction cancer by HER2 status: a European and USA
International collaborative analysis. Ann Oncol. 2012;23(10):2656-2662.
114. Pirrelli M, Caruso ML, Di Maggio M, Armentano R, Valentini AM. Are biopsy
specimens predictive of HER2 status in gastric cancer patients? Dig Dis Sci.
2013;58(2):397-404.
115. Qiu Z, Sun W, Zhou C, Zhang J. HER2 expression variability between primary
gastric cancers and corresponding lymph node metastases.
Hepatogastroenterology. 2015;62(137):231-233.
116. Bozzetti C, Negri FV, Lagrasta CA, et al. Comparison of HER2 status in primary
and paired metastatic sites of gastric carcinoma. Br J Cancer. 2011;104(9):1372-
1376.
117. Ruschoff J, Dietel M, Baretton G, et al. HER2 diagnostics in gastric cancer-
guideline validation and development of standardized immunohistochemical
testing. Virchows Arch. 2010;457(3):299-307.
118. Gullo I, Grillo F, Molinaro L, et al. Minimum biopsy set for HER2 evaluation in
gastric and gastro-esophageal junction cancer. Endosc Int Open. 2015;3(2):E165-
E170.
119. Bang YJ. Advances in the management of HER2-positive advanced gastric and
gastroesophageal junction cancer. J Clin Gastroenterol. 2012;46(8):637-648.
120. Abrahao-Machado LF, Jacome AA, Wohnrath DR, et al. HER2 in gastric cancer:
comparative analysis of three different antibodies using whole-tissue sections and
tissue microarrays. World J Gastroenterol. 2013;19(38):6438-6446.
121. Stolte M, Sticht T, Eidt S, Ebert D, Finkenzeller G. Frequency, location, and age
and sex distribution of various types of gastric polyp. Endoscopy. 1994;26(8):659-
665.
122. Borch K, Ahren B, Ahlman H, Falkmer S, Granerus G, Grimelius L. Gastric
carcinoids: biologic behavior and prognosis after differentiated treatment in
relation to type. Ann Surg. 2005;242(1):64-73.
123. Burkitt MD, Pritchard DM. Review article: pathogenesis and management of
gastric carcinoid tumours. Aliment Pharmacol Ther. 2006;24(9):1305-1320.
124. Rorstad O. Prognostic indicators for carcinoid neuroendocrine tumors of the
gastrointestinal tract. J Surg Oncol. 2005;89(3):151-160.
125. McCall CM, Shi C, Cornish TC, et al. Grading of well-differentiated pancreatic
neuroendocrine tumors is improved by the inclusion of both Ki67 proliferative
index and mitotic rate. Am J Surg Pathol. 2013;37(11):1671-1677.
126. Chen WC, Warner RR, Ward SC, et al. Management and disease outcome of type I
gastric neuroendocrine tumors: the Mount Sinai experience. Dig Dis Sci.
2015;60(4):996-1003.
127. Raderer M, Kiesewetter B, Ferreri AJ. Clinicopathologic characteristics and
treatment of marginal zone lymphoma of mucosa-associated lymphoid tissue
(MALT lymphoma). CA Cancer J Clin. 2016;66(2):153-171.
128. Isaacson PG, Spencer J. Malignant lymphoma of mucosa-associated lymphoid
tissue. Histopathology. 1987;11(5):445-462.
129. Ye H, Liu H, Raderer M, et al. High incidence of t(11;18)(q21;q21) in
Helicobacter pylori-negative gastric MALT lymphoma. Blood. 2003;101(7):2547-
2550.
130. Bacon CM, Du MQ, Dogan A. Mucosa-associated lymphoid tissue (MALT)
lymphoma: a practical guide for pathologists. J Clin Pathol. 2007;60(4):361-372.
131. Copie-Bergman C, Gaulard P, Lavergne-Slove A, et al. Proposal for a new
histological grading system for post-treatment evaluation of gastric MALT
lymphoma. Gut. 2003;52(11):1656.
132. Vanek J. Gastric submucosal granuloma with eosinophilic infiltration. Am J Pathol.
1949;25(3):397-411.
133. Carmack SW, Vemulapalli R, Spechler SJ, Genta RM. Esophagitis dissecans
superficialis (“sloughing esophagitis”): a clinicopathologic study of 12 cases. Am
J Surg Pathol. 2009;33(12):1789-1794.
134. Rossi P, Montuori M, Balassone V, et al. Inflammatory fibroid polyp. A case report
and review of the literature. Ann Ital Chir. 2012;83(4):347-351.
135. Rittershaus AC, Appelman HD. Benign gastrointestinal mesenchymal BUMPS: a
brief review of some spindle cell polyps with published names. Arch Pathol Lab
Med. 2011;135(10):1311-1319.
136. Morandi E, Pisoni L, Castoldi M, Tavani E, Trabucchi E. Gastric outlet obstruction
due to inflammatory fibroid polyp. Ann Ital Chir. 2006;77(1):59-61.
137. Stolte M. Clinical consequences of the endoscopic diagnosis of gastric polyps.
Endoscopy. 1995;27(1):32-37; discussion 59-60.
138. Cassier PA, Ducimetiere F, Lurkin A, et al. A prospective epidemiological study of
new incident GISTs during two consecutive years in Rhone Alpes region:
incidence and molecular distribution of GIST in a European region. Br J Cancer.
2010;103(2):165-170.
139. Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal stromal
tumors: a consensus approach. Hum Pathol. 2002;33(5):459-465.
140. Miettinen M, El-Rifai W, H L Sobin L, Lasota J. Evaluation of malignancy and
prognosis of gastrointestinal stromal tumors: a review. Hum Pathol.
2002;33(5):478-483.
141. Miettinen M, Sarlomo-Rikala M, Lasota J. Gastrointestinal stromal tumors: recent
advances in understanding of their biology. Hum Pathol. 1999;30(10):1213-1220.
142. Goddard AF, Badreldin R, Pritchard DM, Walker MM, Warren B. The management
of gastric polyps. Gut. 2010;59(9):1270-1276.
143. Dei Tos AP, Laurino L, Bearzi I, Messerini L, Farinati F. Gastrointestinal stromal
tumors: the histology report. Dig Liver Dis. 2011;43 Suppl 4:S304-S309.
144. Liegl-Atzwanger B, Fletcher JA, Fletcher CD. Gastrointestinal stromal tumors.
Virchows Arch. 2010;456(2):111-127.
145. Caletti G, Deviere J, Fockens P, et al. Guidelines of the European Society of
Gastrointestinal Endoscopy (ESGE) Part II: retroperitoneum and large bowel,
training. The European Endosonography Club working party. Endoscopy.
1996;28(7):626-628.
146. Van Dam J, Brady PG, Freeman M, et al. Guidelines for training in electronic
ultrasound: guidelines for clinical application. From the ASGE. American Society
for Gastrointestinal Endoscopy. Gastrointest Endosc. 1999;49(6):829-833.
147. Nesje LB, Laerum OD, Svanes K, Odegaard S. Subepithelial masses of the
gastrointestinal tract evaluated by endoscopic ultrasonography. Eur J Ultrasound.
2002;15(1-2):45-54.
148. Casali PG, Jost L, Reichardt P, Schlemmer M, Blay JY. Gastrointestinal stromal
tumours: ESMO clinical recommendations for diagnosis, treatment and follow-up.
Ann Oncol. 2009;20 Suppl 4:64-67.
149. Casali PG VJ, Kotasek D, LeCesne A, Reichardt P, Blay JY. Imatinib mesylate in
advanced gastrointestinal stromal tumors (GIST): survival analysis of the
intergroup EORTC/ISG/AGITG randomized trial in 946 patients. Eur J Cancer.
2005;3:201; abstract 711.
150. Debiec-Rychter M, Sciot R, Le Cesne A, et al. KIT mutations and dose selection
for imatinib in patients with advanced gastrointestinal stromal tumours. Eur J
Cancer. 2006;42(8):1093-1103.
151. Carney JA, Stratakis CA. Familial paraganglioma and gastric stromal sarcoma: a
new syndrome distinct from the Carney triad. Am J Med Genet. 2002;108(2):132-
139.
152. Pasini B, McWhinney SR, Bei T, et al. Clinical and molecular genetics of patients
with the Carney-Stratakis syndrome and germline mutations of the genes coding for
the succinate dehydrogenase subunits SDHB, SDHC, and SDHD. Eur J Hum Genet.
2008;16(1):79-88.
153. Gaal J, Stratakis CA, Carney JA, et al. SDHB immunohistochemistry: a useful tool
in the diagnosis of Carney-Stratakis and Carney triad gastrointestinal stromal
tumors. Mod Pathol. 2011;24(1):147-151.
154. Miettinen M, Sarlomo-Rikala M, Sobin LH, Lasota J. Esophageal stromal tumors:
a clinicopathologic, immunohistochemical, and molecular genetic study of 17 cases
and comparison with esophageal leiomyomas and leiomyosarcomas. Am J Surg
Pathol. 2000;24(2):211-222.
155. Hyun JH, Jeen YT, C