Sei sulla pagina 1di 247

Survival Guide to Gastrointestinal

Mucosal Biopsies.

Pathology Survival Guides

hil novatiw·e

Pathology
Press
PATHOLOGY SURVIVAL GUIDES

First Series
Volume 1

Survival Guide
to Gastrointestinal Mucosal Biopsies
by
Elizabeth A. Montgomery, MD
Professor of Pathology, Oncology, and Orthopedic Surgery
The Johns Hopkins Medical Institutions
Baltimore, Maryland

Kiyoko Oshima, MD, Dr. Se.


Director of Clinical Hepatic Pathology
Assistant Professor of Pathology
Johns Hopkins Medical Institutions

Lysandra Voltaggio, MD
Director, Gastrointestinal Pathology Fellowship
Assistant Professor of Pathology
Johns Hopkins Medical Institutions

Published by the
Innovative Pathology Press
2017

I Innovative
~)' Pathology
...;.s;.. Press
Pathology Survival Guides

EDITOR

Elizabeth A. Montgomery, MD
Professor of Pathology, Oncology, and Orthopedic Surgery
The Johns Hopkins Medical Inst itutions
Baltimore, Maryland

EDITORIAL BOARD

Jerad M. Gardner, MD
Associate Professor of Pathology and Dermatology
Dermatopathology, Bone & Soft Tissue Pathology
Program Director, Dermatopathology Fellowship
University of Arkansas for Medical Sciences

Kiyoko Oshima, MD, Dr. Se.


Director of Clinical Hepatic Pathology
Assistant Professor of Pathology
Johns Hopkins Medical Institutions

Lisa M. Rooper, MD
Assistant Professor of Pathology
Johns Hopkins Medical Institutions

Lysandra Voltaggio, MD
Director, Gastrointestinal Pathology Fellowship
Assistant Professor of Pathology
Johns Hopkins Medical Institutions

Available from the Innovative Pathology Press


Virginia
www.innovativepathologypress.com
ISBN 1-933477-62-8
978-1-933477-62-6

Copyright © 2017 The Innovative Pathology Press

Printed in South Korea


Preface

With this slim volume, we introduce the Pathology Survival Guide series from Innovative
Pathology Press. Although there are many excellent sources for learning·diagnostic pathology, those
directed at medical students are too simplified to be practical for daily work with diagnostic pathology,
and many other texts assume a foundation of more knowledge than many of us have as we begin to
learn about a new area of diagnostic pathology. This series is intended to help residents and colleagues
who begin to tackle an organ system that is unfamiliar to them. Further, in the interest of making the
volumes affordable to trainees and those beginning, we have chosen a soft cover format to contain
costs but not at the expense of high quality images, which we believe will enhance the availability of the
books to those who will benefit most from them.
We begin with mucosal gastrointestinal pathology with the basics and a framework to diagnose
the common lesions together with a few exotic lesions that are important. In some sections, the focus is
on patterns of injury whereas in others, the key is paying attention to the specific type of mucosa that is
injured. We have tried to point out a few pitfalls in each section. We will not be able to create experts
with the material presented, but a road map is provided to set the stage to learn more, and we hope to
pique interest in the topic.
Upcoming volumes will cover breast, liver, prostate, head and neck, frozen sections, skin,
pancreas, bone, soft tissue pathology, hematopathology, and cytopathology, and feature a host of
authors who are superb educators. On behalf of ourselves and the rest of the Editorial Board, we hope
that many colleagues and residents will find the series useful and enjoyable to read.

Elizabeth A. Montgomery, MD
Kiyoko Oshima, MD, Dr. Se.
Lysandra Voltaggio, MD

Acknowledgments and dedication

We acknowledge the team at Innovative Pathology Press and the editorial board members for
their expertise and advice.

We dedicate this volume to the residents, fellows, and colleagues who love diagnostic
pathology.
CONTENTS

1. Non-Neoplastic Esophagus. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Intraepithelial Eosinophils. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Intraepithelial Lymphocytes with or without Apoptotic Bodies. . . . . . . . . . . . . . . . 8
Ulcers and Erosions (Neutrophils) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Apoptotic Injury. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Esophagitis Dissecans Pattern . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Granulomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Hyperkeratosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . 15
2. Barrett's Esophagus and Esophageal Neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
Dysplasia in Barrett's Esophagus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Barrett's Esophagus: Negative for Dysplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Barrett's Esophagus: Indefinite for Dysplasia. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Barrett's Esophagus: Low-Grade Dysplasia. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Barrett's Esophagus: High-Grade Dysplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Intramucosal Carcinoma ......................................... : . . 31
Variant Dysplasia and Carcinoma Patterns. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
Treatment Issues. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
Endoscopic Mucosal Resection (EMR) Specimens . . . . . . . . . . . . . . . . . . . . . . . . . 36
Adenocarcinoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
Squamous Precursor Lesions and Squamous Cell Carcinoma . . . . . . . . . . . . . . . . . . 37
Precursor Lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
Squamous Cell Carcinoma. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
Other Esophageal Lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
Granular Cell Tumor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
Leiomyoma. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
3. Non-Neoplastic Stomach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
What Type of Gastric Mucosa is Present? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
Is the Mucosa Inflamed? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
H. Pylori Gastritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
Autoimmune Gastritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
Lymphocytic Gastritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
Is there Prominent Pit Apoptosis? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
Other Conditions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
4. Gastric Polyps and Neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
Polyps.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
Fundic Gland Polyps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
Hyperplastic Polyps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
Swvival Guide to Gastrointestinal Mucosal Biopsies

Adenomas ...... . . . ........ .......... ...... . .. .. ... , . . . . . . . . . . . . . . 70


Adenocarcinoma and Mimics ........... ·. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74
Metastatic Carcinomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77 ·
Neuroendocrine Tumos .................................. ·. . . . . . . . . . . . . 81
Lymphomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Mucosa-Associated Lymphoid Tissue Lymphomas.. . . . . . . . . . . . . . . . . . . . . . . 85
Inflammatory Fibroid Polyp. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
Gastrointestinal Stromal Tumors (GISTs) of the Stomach. . . . . . . . . . . . . . . . . . . . . 88
5. Non-Neoplastic Small Intestine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
Normal and Reactive Conditions.. ..... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
Infectious Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
Celiac Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
Autoimmune Enteropathy and Common Variable Immunodeficiency . . . . . . . . . . 109
Medication-Associated Injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
6. Small Bowel Neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Polyps. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Tubular/Tubulovillous Adenoma. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Pyloric Gland Adenoma. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
Brunner Gland Lesions- and Pancreatic Heterotopia . . . . . . . . . . . . . . . . . . . . . . . 124
Peutz-Jeghers Polyps. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
Adenocarcinoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
Ampullary Carcinomas.... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Neuroendocrine Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132
Gangliocytic Paraganglioma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Lymphomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Follicular Lymphoma. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 7
Mantle Cell Lymphoma. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
T-Cell Lymphomas. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
Mesenchymal Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
Gastrointestinal Stromal Tumors (GISTs) of the Small Bowel. . . . . . . . . . . . . . . . 140
Gastrointestinal Tumors of Duodenum. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
Gastrointestinal Tumors of]ejunum and Ileum . . . . . . . . . . . . . . . . . . . . . . . . . . 140
Vascular Lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141
7. Non-Neoplastic Colon . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
Normal Colonic Mucosa . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146
Infections with Characteristic Features . . . ....... ..... .. . . ... . . ...... . ... . 148
Acute Colitis. . . . . . . . . . . . . . . . . . . . . . . . ....... ..... .. . . ... . . ...... . ... . 148
Ischemic Colitis Pattern . . . . . . . . . . . . . . . ....... ..... .. . . ... . . ...... . ... . 148
Active Chronic Colitis . . . . . . . . . . . . . . . . ....... ..... .. . . ... . . ...... . ... . 153
Active Chronic Colitis Mimickers of Inflammatory Bowel Disease. . . . . . . . . . . . . . 156

ii
Contents

Lymphocytic and Collagenous Colitis............... . . . . . . . . . . . . . . . . . . . . . 157


Apoptotic Colopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161
8. Colon Polyps and Neoplasms. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
Benign Findings that Can Mimic Neoplasms or Other Diseases. . . . . . . . . . . . . . . . 167
Mucosal Prolapse Polyps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
Polyps: Conventional Adenomas, Hyperplastic Polyps, and Other Serrated Polyps. 174
Tubular Adenomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17 4
High-Grade Dysplasia and Villous Features in Adenomas . . . . . . . . . . . . . . . . . . 17 6
Pseudoinvasion (Mucosal Prolapse Change) in Adenomas. . . . . . . . . . . . . . . . . . 178
Evaluation of Adenomas that Contain Carcinomas in Polypectomy Specimens
("Malignant Polyp"). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 180
Serrated Polyps. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183
Colorectal Cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189
Colitis-Associated Neoplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
Juvenile Polyps. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
Neuroendocrine Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
Other Colorectal Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197
9. Non-Neoplastic Anus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
He1norrhoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
Papillary Endothelial Hyperplasia (Vegetant Intravascular Hemangioendo-
thelioma) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206
Infectious Proctitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208
Herpes Simples Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208
Cytomegalovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208
Syphilis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208
Lymphogranuloma Venereum.. . ... .... .... . ......... . . .... .. . ....... 211
Crohn's Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
10. Anal Polyps and Neoplasms. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
Hidradenoma Papilliferum. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
Polyps........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
Inflammatory Cloacogenic Polyp . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
Fibroepithelial Polyps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
Anal Squamous Intraepithelial Neoplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 217
Anal Squamous Cell Carcinoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220
Paget Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 225
Adenocarcinoma and Anal Gland Adenocarcinomas . . . . . . . . . . . . . . . . . . . . . . . . 228
Other Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229
Granular Cell Tumor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229
Melanoma and Nevi. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 235

iii
1 NON -NEOPLASTIC ESOPHAGUS

Although it takes many years and constant Esophageal biopsies often lack lamina pro-
evaluation of the literature to develop a deep pria because the normal esophagus is slippery,
~xpertise in gastrointestinal pathology, it is rel- strong and difficult to biopsy (fig. 1-1). If lamina
atively easy to master the foundations of dealing propria is present, don't mistake it for submu-
with gastrointestinal biopsies. The objective is to cosa (figs. 1-2-1-5). This can lead to errors in
offer tips for getting started as well as examples determining depth of invasion when confront-
of some of the pitfalls that can be encountered. ed with an early invasive carcinoma. In general,
We will begin in the esophagus and review topics lamina propria is only present on biopsies of
from proximal to distal beginning in the esoph- esophageal squamous mucosa that is dam-
agus and ending with the anal canal. aged. There is often only epithelium present

Figure 1-1
NORMAL ESOPHAGEAL
SQUAMOUS EPITHELIUM
The basal cells are darker and
have less cytoplasm than the
more superficial mature squa-
mous cells, which are oriented
with their cytop lasm parallel
to the basement membrane.
The indicated vascular papilla
extends for less than half the
thickness of the epithelium.

Figure 1-2
LAYERS OF THE ESOPHAGUS
The important feature to note
is that the lamina propria lies
beneath the epithelium, between
the epithelium and muscularis
mucosae. This layer is often
misinterpreted as submucosa.
This is from a resection specimen.
Note that the tissue at the left of
the image has been crushed. At
the right part of the image, the
ti ssue has curled, creating an
impression that the lateral edge
is a deep one.

1
Survival Guide to Gastrointestinal Mucosal Biopsies

I '':-: "'

Figure 1-3 -. ..
ESOPHAGEAL BIOPSY WITH
ATTACHED LAMINA PROPRIA
AND SUBMUCOSAL
GLAND AND DUCT
It is unusual to see submucosal
glands (lower right) or ducts
(left) in biopsies unless there has
been prior damage to the tissue.
Ducts are typically surrounded
by a brisk lymphoplasmacytic
infiltrate. .
~.(
~~~:tP«;·,.:.tl~-,
~ ;, .,
':;'\. '1.; ..
.~r'~;:-.-
rf ·'
~f-1~;·t;-, ~ · . .... ..-·-
1. .i-,v~: "}). . ,. ~ ~
- . ·-·~·:-
·.:.;". ''1\-::.~'\!fl:~' /, .: . ,., _..,-..~. ~~-"H ;.,.pr,);:-.,.,~;.. ,.

·~t~:!~~z~~~1~~~~~~5~ ~:·:i~~~:~~~~(~~:~. ..,


•\\.(/ .. )~ ~: . • .:: ... !\l · · • •' ...·?"~ J:~~i."' ;~:\~{ I
'·".t..:...-.:3.~·· :: -~~~

( .'
··. -~: ... _. . . .

.,
,~ . ....
\

Figure 1-4
ESOPHAGEAL BIOPSY WITH ATTACHED LAMINA PROPRIA AND SUBMUCOSAL GLAND
Left: The submucosal gland (lower right) is present beneath muscularis mucosae.
Right: In this high magnification, there is small duct at the left of the glands (center). These glands produce mucin to
lubricate the esophagus.

in biopsies. The vascular papillae should only patterns that offer clues to inquire further. If
extend halfway up to the surface. The basal normal versus "reactive epithelial changes"
cells should be only a few layers (2 to 4layers). is reported, no one will call with queries, so
Most of the time the consequences of what is most of these samples can be handled quickly.
written in pathology reports of squamous mu- Although reflux disease has traditionally been
cosal biopsies are minimal but there are several associated with basal layer proliferation and

2
Non-Neoplastic Esophagus

.....' • •
I

- · ~··"' '
, . ! "
,.' I
....
t' "' . ., .. .

·\
'
- ., ,, ,
'
-..
-· \.
• . .... ~)

••l",., • •
. -·-.-
. .... . •
• •
. ."
-.

• -~
'
( t .•.•
' t ~
J
•, •
.' . ....
.' '... . •'
~ "' .'


Figure 1-5
ESOPHAGEAL BIOPSY FROM A PATIENT WITH AMYLOIDOSIS
Left: The presence of lamina propria and muscularis mucosae indicate prior damage. There is prominent chronic
inflammation in the lamina propria investing the cardiac glands at the right side of the image. On careful examination thick
vessels laden with amyloid are apparent.
Right: High magnification of the muscularis mucosae from the sample is shown on the left. Note the waxy appearance
of the amyloid that thickens the vessels.

extension of the vascular papillae with scat- The gastric cardia does not produce hor-
tered eosinophils, a thickness of more than 430 mones of interest in clinical diagnosis, so, for
microns has been better correlated with reflux example, gastrin stains are negative in the gas-
disease than these traditional features (1). This tric cardia. An example of a damaged gastro-
is equivalent to 0.43 mm. It is easy to estimate esophageal junctional mucosa is seen in figure
1 mm on a glass slide, so finding well oriented 1-6. The most likely etiology for the damage is
squamous epithelium measuring 1 mm or more reflux disease.
is a great clue that the patient has reflux disease The esophagus and the gastric cardiac area is
before the slide is even reviewed. frequently damaged in adults, most commonly
The gastric cardia lies between the esophagus as a result of reflux. Repeated cycles of damage
and stomach. It is lined by foveolar epithelium and repair produce disordered anatomy and
and cardiac glands are found underneath in a the most important of these ana to mic disorders
generous lamina propria. Note that the lamina is the duplication or general expansion of the
propria in the stomach, small intestine, and co- muscularis mucosae. This change is seen in most
lon consists of loose connective tissue by which examples of Barrett's esophagus but it can also
glands are invested, whereas it sits beneath the be found in the adjoining squamous mucosa
epithelium and above the muscularis mucosae (figs. 1-6, 1-7). It is important to be aware of
in the esophagus. this anatomic alteration as it becomes an issue

3
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 1-6
INFLAMED GASTROESOPHAGEAL
]UNCTIONAL MUCOSA
There is inflamed squamous
epithelium on the left, together with
lamina propria (the edematous tissue
containing inflammatory cells just
beneath the epithelium; this layer
is NOT the submucosa). Note that
the basal cells involve about half the
thickness of the squamous epithelium.
On the right is reactive gastric cardiac
mucosa, with mucin-containing
epithelium on top of lamina propria.

Duplicated disorganized Figure 1-7


ENDOSCOPIC MUCOSAL RESECTION
SAMPLE TO DEMONSTRATE LAYERS
OF THE MUCOSAL/SUBMUCOSA
An early adenocarcinoma is present
at the right part of the field and the
,\. · · ' '• original and duplicated muscularis
\ •1 . mucosae are indicated. Since the tissue
•' •••
>
has curled, the lateral margin at the
right has the appearance of a deep
margin. The lateral (mucosal) margins,
consisting of epithelium, lamina
propria, and muscularis mucosae, are
indicated by the thick black lines,
Submucosa whereas the deep (submucosal) margin
is indicated by the orange line.

Figure 1-8
DUPLICATED MUSCULARIS
MUCOSAE IN PREVIOUSLY
DAMAGED SQUAMOUS MUCOSA
ADJOINING BARRETT MUCOSA
It is easy to mistake the original
muscularis mucosae for muscularis
propria.

4
Non-Neoplastic Esophagus

Figure 1-9
GASTRIC CARDIAC MUCOSA
In this image, four layers/lines are
present. The first consists of the apical
mucin cap of the foveolar epithelium,
the second consists of the base of
this cap, the third of the remaining
cytoplasm, and the last of the row
of nuclei.

in evaluating mucosal resection samples or even


esophagectomy specimens (fig. 1-8).
If gastric mucosa is encountered in the upper
esophagus, it is an embryologic rest termed inlet
patch. It is an incidental finding although, very
rarely carcinomas are believed to arise within
this tissue.
Reflux disease sets the stage for columnar
metaplasia in the esophagus with or without
goblet cells. In figure 1-9, the concept of four
layers in surface cardiac mucosa is demonstrat-
ed, a concept that we can use when we evaluate
Banett's dysplasia later in this volume. There are
several patterns of injury to the esophagus that
can be seen: 1) intraepithelial eosinophils; 2) in-
traepitheliallymphocytes with or without apop-
totic bodies; 3) ulcers and erosions (neutrophils);
4) apoptotic injury; 5) esophagitis dissecans
pattern; 6) granulomas; and 7) hyperkeratosis.
Figure 1-10
INTRAEPITHELIAL EOSINOPHIL$
FELINE ESOPHAGUS
In theory there should be no eosinophils in This pattern of ridges is reminiscent of the appearance
esophageal epithelium or lamina propria. They of the esophagus in cats and is characteristic of eosinophilic
are found in the setting of reflux disease and esophagitis.
allergic disease (eosinophilic esophagitis) . It is
currently believed that there is frequent over- ologic exercise. Some cases are, however, slam
lap in these diseases (2,3), but some cases are dunks! If there is striking basal cell hyperplasia at
readily attributable to one or the other. We are low magnification and eosinophils are numerous
often asked to determine whether biopsies show (no need to count them), the patient has eosin-
eosinophilic (allergic) esophagitis versus reflux ophilic esophagitis (figs. 1-10-1-12). If there is
esophagitis. We are only part of the equation and only slight basal cell hyperplasia and a search
these conditions are separated as a clinicopath- is required to find eosinophils, the patient has

5
Survival Guide to Gastrointestinal Mucosal Biopsies

,..
' T •

,._ J •
,t'fi ~t;, ·,·. J
:......~ ~.

. .. . . . ..
,.;: ,.· ; ..... •• c

Figure 1-11
EOSINOPHILIC ESOPHAGITIS
A: There is striking basal cell hyperplasia although the intraepithelial eosinophils are not particularly conspicuous at this
magnification . The attached lamina propria is scarred/fibrotic.
B: There is striking basal cell hyperplasia and eosinophils are plentiful.
C: There is no need to count to see that there are far too many eosinophils. Report a large number thereof!
D: There is striking basal cell hyperplasia and prominent edema between the epithelial cells such that the intercellular
bridges are easy to see.

6
Non-Neoplastic Esophagus

••

Figure 1-12
EOSINOPHILIC ESOPHAGITIS
Left: This example features an eosinophilic microabscess.
Right: Food impaction is a classic presentation. Note the adjoining skeletal muscle fibe rs in keeping with ingested
impacted meat.

Figure 1-13
REFLUX ESOPHAGITIS
Left: The basal cell h yperplasia is far less striking than that associated with eosinophilic gastritis.
Right: Scattered eosinophils are present.

7
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 1-14
PROMINENT INTRAEPITHELIAL LYMPHOCYTES, A MARKER FOR CROHN'S DISEASE IN CHILDREN
Left: This is a nonspecific finding in adults but very characteristic of Crohn's disease in children .
Right: High magnification of left image.

reflux disease (fig. 1-13). If the endoscopist has in white male children with food impaction
sampled both the mid esophagus and the distal (fig. 1-12, right) but there are many exceptions.
esophagus and there are loads of eosinophils in Reflux is treated with proton pump inhibitors
the mid esophagus, the patient probably has and other medications intended to reduce the
allergic/eosinophilic esophagitis. In truth there mucosal damage from the gastric contents that
is no need to count the eosinophils-an estimate bathe the esophagus. In contrast, the idea for
will do as it is desired by colleagues even though eosinophilic esophagitis is to reduce dietary
in some respects it is meaningless because there is allergens but the diets are inedible for some
tremendous variability of the eosinophil counts in such that steroids are used. The so-called six
different biopsy fragments from the same patient food elimination diet is quite unpalatable. Few
at the same time. Society guidelines suggest a 15 adults among us could follow it but it can be
eosinophils per high power field cutoff for eosino- forced on small children who don't know better:
philic esophagitis in the correct clinical setting. Of
course if there is a history of eosinophilic esopha- INTRAEPITHELIAL LYMPHOCYTES W ITH
gitis that has been treated, the counts go down. OR W ITHOUT APOPTOTIC BODIES
Finding scarring in the lamina propria (fig. Finding lymphocytes in esophageal squa-
1-llA,B) is very typical of eosinophilic esoph- mous epithelium is often a nonspecific finding
agitis and the scarring, which itself contains and it is attributed to reflux disease in adults but
eosinophils, tends to persist after the epithelial there are a few situations for which this finding
eosinophil counts decrease. For biopsies in offers a clue.
which the history and microscopic findings If intraepithelial lymphocytes without asso-
are not clear cut, the following report style is ciated apoptotic squamous epithelial cells are
acceptable: "Squamous mucosa with reactive found in biopsies from children, this is very
epithelial changes with up to X eosinophils per strong evidence that the child has Crohn 's
high power field." disease (fig. 1-14). It cannot be reported un-
Reflux disease is classically encountered in equivocally as such but should prompt a note
obese white males and eosinophilic esophagitis indicating that the patient should be further

8
Non-Neoplastic Esophagus

Figure 1-15
LICHEN PLANUS ESOPHAGITIS
A: There is prominent lamina propria
chronic inflammation, marked intra-
epithelial lymphocytosis, and so m e
hyperke ratosis at the surface of the
epithelium. Diagnosing esophageal
lichen planus can require correlation
with a history of skin involvement or
oral involvement or even immuno-
fluorescence studies since it is indistin-
guishable from lichenoid esophagitis.
B: Note the apoptotic epithelial cell
in the center of the field (Civatte body).
C: High magnification of an apoptotic
epithelial cell.



c
evaluated for Crohn's disease. Of course, ications and viral hepatitis (4). If the patient has a
occasional lymphocytes are acceptable in the known history of skin lichen planus, the esopha-
esophageal squamous epithelium but if they geal disease can be assumed to be a component
are readily noted at 4x, there are too many. In but otherwise the diagnosis must be descriptive.
adults, esophageal intraepitheliallymphocyto- Esophageal lichen planus tends to be encoun-
sis is wholly nonspecific. tered in middle aged to elderly women. The
If intraepitheliallymphocytosis accompanied esophagus lacks a granular layer so the findings
by apoptotic bodies is encountered, the patient of esophageallichen planus do not fully mirror
will nearly invariably be an adult (figs. 1-15, 1-16). those of skin disease. When a lichenoid pattern
This is the pattern encountered in esophageal or is encountered, especially if there are a few neu-
oral lichen planus but it is indistinguishable from a trophils, it is always worthwhile to also assess for
pattern of injury that has been termed "lichenoid Candida, which may be superimposed. In general
esophagitis," which is associated with vaiious med- Candida alone does not cause this type of injury.

9
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 1-16
LICHENOID ESOPHAGITIS
Left: This example was seen in a patient taking a monoclonal immunomodulatory medication. There is lymphocytosis
and an apoptotic epithelial cell at the bottom of the field.
Right: High magnification of the apoptotic epithelial cell on the left. The findings are identical to those associated with
lichen planus.

Similarly, if there are numerous monocytic cells/ However, ulcers are also likely to result from
macrophages, infection with herpes simplex virus reflux disease and from medications (figs. 1-19-1-
and cytomegalovirus should be sought, even if 23). The list of medications that can result in
immunolabeling is required (5,6). esophageal injury is long but some produce
characteristic patterns of injury and sometimes
ULCERS AND EROSIONS (NEUTROPHIL$) pill fragments (polarizable material) can be found
Ulcers and erosions merit a search for in- embedded in the ulcers or erosions. Sodium poly-
fectious agents (figs. 1-17, 1-18). An erosion styrene sulfonate (Kayexalate), an exchange resin
destroys part of the mucosa whereas an ulcer used to reduce serum potassium in renal failure
destroys it all, down to the submucosa or deeper. patients results in deposition of characteristic
Of course the key viruses to consider are herpes crystals (fig. 1-19) (7). Doxycycline often results
simplex virus (HSV) and cytomegalovirus (CMV) in nonspecific ulcers, but a characteristic pattern
whereas in general Candida esophagitis that is of vascular injury can also be encountered (fig.
encountered on biopsies results in parakeratosis 1-20) (8,9). Iron pills can also cause esophageal
(below) rather than ulcers. Remember that HSV damage or potentiate existing damage (fig.
viral cytopathic change is found in epithelial 1-21) (10). Agents to treat osteoporosis (the bi-
cells and CMV is almost always detected in en- sphosphonates) are well-known for resulting in
dothelial cells. As such, endoscopists are taught esophageal damage (11) but after a spate of legal
to sample the edges of ulcer beds tohave both actions, patients are currently instructed to take
types of "hunting ground" in their samples. these with copious amounts of water and not to

10
Non-Neoplastic Esophagus

Figure 1-17
CYTOMEGALOVIRUS ESOPHAGITIS
An infected cell is in the center of the
image and it is an endothelial cell. Note
the monohistiocytic background, a clue
to search for the viral cytopathic effect.
In this case, the viral cytopathic effect is
somewhat subtle.

Figure 1-18 Figure 1-19


HERPES SIMPLEX VIRUS ESOPHAGITIS KAYEXALATE CRY STAL
Note that epithelial cells are affected. This kayexalate crystal found in an ulcer bed has a
cracked appearance and purple calor.

lie down shortly after taking them, so this type of are vastly more common than sarcomas or sar-
injury, which lacks a characteristic morphology, comatoid carcinomas involving the esophagus.
is encountered less frequently than in the past.
Of course, ulcers initiated by one form of injmy APOPTOTIC INJURY
can always be complicated by another, so ulcers Esophageal apoptotic injury has fewer as-
and erosions should always be studied carefully. sociations than such injury in the stomach
Lastly, ulcers can be associated with striking and intestines, but they are important. If there
reactive stromal changes (fig. 1-24) (12), which is associated lymphocytosis, as noted above,

11
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 1-20
EROSIVE ESOPHAGITIS ASSOCIATED WITH DOXYCYCLINE
Left: The appea rance of the lamina propria vessel is a clue. Review of the chart indicated that the patient was taking
doxycycline (a history of severe acne is common). Note that there are Candida organisms embedded in the epithelium at
the upper right. It is not clear which of the two initiated the mucosal in jury, but probably the candidiasis is a superinfection .
Right: High magnification of the lesfon shown on the left. Note the fibrinoid necrosis of the vessel.

Figure 1-21
IRON PILL ESOPHAGITIS
Left: The iron has oxidized, imparting a blue to black tint. The key is to note the reactive epithelial cha nges, which can
be mistaken for dysplasia/intra-epithelial neoplasia.
Right: This is an iron stain.

12
Non-Neoplastic Esophagus

Figure 1-22
EROSIVE ESOPHAGITIS
WITH REACTIVE CHANGES
A: This erosion was found at the gastro-
esophageal junction in a patient without
eosinophilic esophagitis in undamaged mucosa, so
the significance of the eosinophils is not clear; it is
perhaps allergic or a florid response to the refluxate.
Note the epithelial changes, which are reparative.
B: This high power image shows the lamina
propria scarring and inflammation.
C: Note that the regenerating squamous
epithelium features squamous cells that are clearly
separated from one another and the intercellular
bridges are readily apparent. This is a clue that the
cells are reactive rather than dysplastic, although
occasional squamous dysplasia cases display
prominent intercellular bridges.

·v_,,,
•I '
'j I

Figure 1-23
PILL ESOPHAGITIS
Note the pill fragments . Note also the
striking reactive changes and that the upper
half of the epithelium has become necrotic
in an esophagitis dissecans pattern.

13
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 1-24
PSEUDOSARCOMATOUS CHANGES
ASSOCIATED WITH ULCER
A: There is Barrett's mucosa in the center, ulcer exudate at
the upper left, and another fragment of interest at top center.
B: Note the fragment at the upper portion of the image,
which contains exudate and plump spindle cells.
C: These are not sarcoma cells but reactive fibroblasts
deprived of oxygen. They generally have a low nuclear
to cytoplasmic ratio and are found in ulcer beds at the
interface between exudate and fully viable tissue. When the
fragments are tangentially embedded, these cells can suggest
sarcoma but they disappear once ulcers heal.

this differs from apoptotic injury without it is important to address viral infections since
lymphocytosis. Viral infections can result in they themselves can result in apoptosis. If they
apoptosis, but there is predominantly an ero- are seen, all bets are off in diagnosing graft ver-
sive or ulcerative appearance as the diagnostic sus host disease AND a viral infection, but the
clue. The main issue is graft-versus-host disease, patient's infection must be treated before giving
which can be diagnosed in the setting of bone steroids to treat the graft versus host disease so
marrow transplantation, often accompanied by the uncertainty is not of clinical importance.
skin changes. It is diagnosed simply by finding Mycophenolate (Cellcept), administered
apoptotic bodies in the squamous mucosa in the to reduce host immunologic damage to solid
correct clinical setting (fig. 1-25). The epithelial organ transplant patients by blocking nucleic
apoptosis in graft versus host disease is usually acid synthesis in lymphocytes, can result in
unaccompanied by much inflammation unless it striking injury in the duodenum or stomach
is severe and there are ulcers. If that is the case, but usually not much injury to the esophagus

14
Non-Neoplastic Esophagus

Figure 1-25 Figure 1-26


GRAFT VERSUS HOST DISEASE ESOPHAGITIS DISSECANS/ SLOUGHING ESOPHAGITIS
Note the striking basal zone apoptosis unaccompanied The epithelium sloughs off in strips.
by inflammation.

since esophageal squamous cells are less depen- initiated by mechanical or thermal (hot beverages)
dent on the blocked pathway than enterocytes esophageal injury. It is generally self-limited.
(13, 14). Thus if a bone marrow transplant A cleft is seen in the middle of the squamous
patient has been taking mycophenolate and epithelium and results in sloughing of the upper
esophageal squamous apoptosis is seen, the half of the epithelium as strips of squamous
damage is probably attributable to graft versus tissue with nuclear coagulative necrosis. Some-
host disease. times inflammatory cells are seen in the cleft but
no fungal elements are seen (figs. 1-26-1-28).
ESOPHAGITIS DISSECANS PATTERN
This pattern is also termed "sloughing GRANULOMAS
esophagitis" because large strips of epithelium Although in theory, esophageal granulomas
are seen sloughing into the lumen at the time might be associated with fungal infections and
of endoscopy (15,16). It is sometimes report- mycobacterial disease, in practice they are usu-
ed incorrectly as esophagitis "dissecans." The ally encountered in children as a manifestation
surface is dissecting, not dry. Endoscopically it of Crohn's disease (fig. 1-29).
can be mistaken for Candida esophagitis and
HYPERKERATOSIS
for bullous skin disease. It is more common
that bullous skin disease and the crack in the There are three main benign associations
epithelium is in a different location. with hyperkeratosis, namely reflux, candidiasis,
This pattern is associated with debilitation, and a rarer presumed precursor to squamous cell
alcoholism and polypharmacy but it is poorly carcinoma termed epidermoid metaplasia. The
understood. There may be some evidence that it is hyperkeratosis associated with reflux is brightly

15
Survival Guide to Gastrointestinal Mucosal Biopsies

. _-...-- .:

:-
__- .. __- _ -- ___ _ ...,. :. - _r"
- -
_ J _ - :-_

... -. -
- -~- ~ -
-
--- -~ - : -:::, --::.,-::--- ... - -- ... _-_.....
... ..--
Figure 1-27 -..'
ESOPHAGITIS DISSECANS/ ·-
SLOUGHING ESOPHAGITIS
In this image, the upper half of the
epithelium is nonviable but has yet to ' -~·~ c:,•
slough off the surface. .-

::- .'
·. : ~

--> ..: ~: ~

Figure 1-28
ESOPHAGITIS DISSECANS/SLOUGHING ESOPHAGITIS
Left: The cleft is in the middle of the epithelium rather than at the base as in pemphigus vulgaris.
Right: At high magnification, there are some inflammatory cells in the cleft but no fungal elements.

eosinophilic compared to the squamous cells be- magnification allows the pathologist to focus
neath it and adheres tightly to the surface, some- on where to search for organisms.
times associated with neutrophils (fig. 1-30). Epidermoid metaplasia simply means that
The hyperkeratosis associated with Candida the esophagus acquires an abnormal granular
is the same calor as the other squamous epithe- layer akin to that in skin (figs. 1-32, 1-33). It is
lium and flakes off the surface into the lumen easy to overlook. Epidermoid metaplasia has
(fig. 1-31). Scanning for this pattern at low the appearance of leukoplakia endoscopically

16
Non-Neoplastic Esophagus

Figure 1-29 Figure 1-30


CROHN'S DISEASE MILD PARAKERATOSIS ASSOCIATED WITH REFLUX
There is a granuloma in the lamina propria. The lamina The squamous cells are plump and more or less adherent
propria is patt of the biopsy since the tissue has been damaged. to the surface.

1 ·.f
··,··'
• : 1'-
..~ . ,t!.

'•
...
,.. ... '
. / t..
..
0

.
c; ,'\ ~r- .

.' .
~

., ·' •
..
Figure 1-31
PARAKERATOSIS ASSOCIATED WITH CANDIDIASIS
Left: The parakeratotic cells flake off the surface. The organisms are found in the flaked zones.
Right: Note the pseudohyphae.

17
Survival Guide to Gastroin.testin.al Mucosal Biopsies

.r
Figure 1-32
EPIDERMOID METAPLASIA
This manifests as a white plaque.

Figure 1-33
EPIDERMOID METAPLASIA
Left: A granular layer, normally absent in esophageal squamous epithelium, is present. The epithelium is otherwise
unremarkable. This seems to be a precursor to squamous neoplasia .
Right: Hyperkeratosis and a prominent granular layer are shown in this example.

and has been termed as such. It is probably a ageal squamous cell carcinoma such as smok-
precursor to squamous cell carcinoma but the ing and alcohol use (17). This contrasts with
rate of progression is not known. It is associated sebaceous heterotopia, which is an incidental
with the same risk factors as those for esoph- finding (fig. 1-34).

18
Non-Neoplastic Esophagus

Figure 1-34
SEBACEOUS HETEROTOPIA
A: Note the oily appearing nodules.
B: Sebaceous glands identical to those in skin are found
in the lamina propria . Note that there is no granular layer
in the overlying epithelium such that one can exclude a
sample switch.
C: High magnification.

19
Swvival Guide to Gastrointestinal Mucosal Biopsies

REFERENCES

1. Vieth M, Mastracci L, Vakil N, et al. Epith eli- 9. Xiao SY, Zhao L, Hart J, Semrad CE. Gastric
a! Thickness is a marker of gastroesophageal mucosal necrosis with vascular degeneration
reflux disease. Clin Gastroenterol Hepatol induced by doxycycline. Am J Surg Pathol
2016;14):1544-1551. 2013;37:259-263.
2. Cheng E, Souza RF, Spechler SJ. Eosinophilic 10. Abraham SC, Yardley JH, Wu TT. Erosive injury
esophagitis: interactions with gastroesophageal to the upper gastrointestinal tract in patients
reflux disease. Gastroenterol Clin North Am receiving iron medication: an underrecognized
2014;43:243-256. entity. Am] Surg Pathol1999;23:1241-1247.
3. Molina-Infante}, Bredenoord A], Cheng E, et al. 11. Abraham SC, Cruz-Correa M, Lee LA, Yardley
Proton pump inhibitor-responsive oesophageal ]H, Wu TT. Alendronate-associated esophageal
eosinophilia: an entity challenging current di- injury: pathologic and endoscopic features . Mod
agnostic criteria for eosinophilic oesophagitis. Pathol1999;12:1152-1157.
Gut 2016;65:524-531. 12. Shekitka KM, Helwig EB. Deceptive bizarre
4. Salaria SN, Abu Alfa AK, Cruise MW, Wood LD, stromal cells in polyps and ulcers of the gastro-
Montgomery EA. Lichenoid esophagitis: clinico- intestinal tract. Cancer 1991;67:2111-2117.
pathologic overlap with established esophageal 13. Nguyen T, Park ]Y, Scudiere JR, Montgomery
lichen planus. Am J Surg Pathol 2013;37:1889- E. Mycophenolic acid (cellcept and myofortic)
1894. induced injury of the upper GI tract. Am J Surg
5. Greenson}K. Macrophage aggregates in cytomega- Pathol 2009;33:1355-1363.
lovirus esophagitis. Hum Pathol 1997;28:375- 14. Vieth M, Montgomery EA. Medication-associat-
378. ed gastrointestinal tract injury. Virchows Arch
6. Greenson}K, Beschorner WE, Boitnott}K, Yard- 2017;470:245-266.
ley ]H . Prominent mononuclear cell infiltrate is 15. Cannack SW, Vemulapalli R, Spechler SJ, Genta
characteristic of herpes esophagitis. Hum Pathol RM. Esophagitis dissecans superficialis ("slough-
1991;22:541-549. ing esophagitis"): a clinicopathologic study of
7. Abraham SC, Bhagavan BS, Lee LA, Rashid A, 12 cases. Am] Surg Pathol2009;33:1789-1794.
Wu TT. Upper gastrointestinal tract injury in 16. Purdy ]K, Appelman HD, McKenna B]. Sloughing
patients receiving kayexalate (sodium polysty- esophagitis is associated with chronic debilita-
rene sulfonate) in sorbitol: clinical, endoscopic, tion and medications that injure the esophageal
and histopathologic findings. Am J Surg Pathol mucosa. Mod Pathol 2012;25 :767-775 .
2001;25:637-644. 17. Singhi AD, Amold CA, Crowder CD, Lam-Himlin
8. Xiao SY, Zhao L, Hart ], Semrad CE. Doxycy- DM, Voltaggio L, Montgomery EA. Esophageal
dine-induced gastric and esophageal mucosal leukoplakia or epidermoid metaplasia: a clinico-
injuries with vascular degeneration. Am J Surg pathological study of 18 patients. Mod Pathol
Pathol2013;37:1115-1116. 2014;27:38-43.

20
2 BARRETT'S ESOPHAGUS AND
ESOPHAGEAL NEOPlASMS

Many biopsies from the esophagus are taken you need once you recognize the findings (2).
to address Barrett's esophagus or other colum- Knowing the endoscopic appearance is import-
nar epithelium found in the esophagus. The ant but recording precisely which epithelial
risk factors for Barrett's esophagus are chronic types are encountered is the main issue. There
gastroesophageal reflux (more than 5 years), age are a number of types of mucosa that can be
over 50 years, male gender, smoking, central encountered: 1) oxyntic mucosa (fig. 2-1); 2)
obesity and Caucasian race. Alcohol use is not cardiac mucosa (fig. 2-2); 3) pancreatic het-
a significant factor and some data suggest a pro- erotopia/metaplasia (fig. 2-3); 4) multilayered
tective effect for wine drinking (1) . Bottoms up! epithelium (figs. 2-4, 2-5) (3); 5) squamous ep-
Many of the issues surround the source of ithelium; and 6) esophageal ducts (see chapter
the biopsy (proximal stomach versus tubular 1, fig. 1-3).
esophagus). There are many articles concern- Finding either multilayered epithelium or
ing special stains to address these issues but a ducts on a biopsy confirms that a sample is from
good hematoxylin and eosin (H&E) stain is all the esophagus rather than the stomach.

Figure 2-1 Figure 2-2


OXYNTIC MUCOSA GASTRIC CARDIAC MUCOSA
This is gastric type mucosa of the type found in the gastric There are no parietal cells and there is lamina propria
body and fundus. The gastric pits are convoluted tubules chronic inflammation , a common finding in patients
that connect to the surface such that a two dimensional who undergo biopsies after presenting reflux symptoms.
section shows crowded glands lined by parietal cells (the Since reflux results in cycles of damage and repair, there is
pink ones in the center of the field). The deep portion of disorga nized smooth muscle in the lamina propria between
the mucosa contains bluish chief cells. The surface foveolar cardiac type glands, which produce mucin. The surface has
cells each have an apical mucin cap. foveolar cells just as the oxyntic mucosa does. Some samples
show a combination ("cardia-oxyntic mucosa").

21
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-4
MULTILAYERED EPITHELIUM
This type of epithelium is common in persons with reflux
Figure 2-3 an d is fou nd in the tubular esophagus. The appearance
PANCREATIC AC INAR CELL
is reminiscent of that of immature squamous metaplasia
HETEROTOPIA IN CARDIAC TYPE MUCOSA of the uterine cervix. The surface appea rs co lumnar
whereas the base is squamous. Note that this finding is in
Th e tubules are closely packed, arranged in lobules, a background of cardiac type mucosa. There is no need to
and the cells formi ng them are more amphophilic (neither report this finding; it is simply important to know abo ut it
eosinophilic nor basophilic) than parietal cells. This is a and not diagnose it as dysplasia. The presence of this type
common finding. of epith elium proves that the sample was taken from the
esoph agus despite the presence of gastric mucosa.

Figure 2-5
MULTILAYERED EPITHELIUM
Left: Note th e mucin in the surface cells. These cells can be shown to demonstrate MUC2 and CDX2 in a subset of
examples; there is no reason to perform these studies in daily practice.
Right: This is a PAS/Aician blue stain. Some of the cells have a bluish tint but they lack th e morphologic features of goble t
cells. There is no reason to perform this staining in daily practice to identify these cells.

22
Barrett's Esophagus and Esophageal Neoplasms

Much of the evaluation concerns whether could be expected but that after 20 years, a 9.1
goblet cells are present in any columnar epithe- to 9.5 percent cumulative cancer incidence was
lium sampled from the esophagus. Although the predicted, suggesting that intensified surveil-
British criteria for Banett's esophagus do not re- lance is indicated for patients with longstanding
quire goblet cells, they do have a segment length Barrett's esophagus (8). Presently identification
requirement (1 cm) (4). This length requirement of the key patients for screening and surveil-
was adopted by the American College of Gastro- lance is poor (9) such that most patients present
enterologists in 2016 (1) although the American with symptoms of advanced incurable disease.
Gastroenterological Association has yet to do so
as of this writing (5). The objective is to collect DYSPLASIA IN BARRETT'S ESOPHAGUS
the patients at highest risk for the development There are four main features to study when
of adenocarcinoma for screening. evaluating Barrett's esophagus for dysplasia (10),
The recommendation by the ACG to intro- although there can be some variations in mor-
duce a disease length criterion is problematic phology (11): 1) surface maturation compared to
for the pathologist since endoscopic findings underlying glands; 2) architecture of the glands;
are often unknown. When the pathologist has 3) cytologic features; and 4) inflammation, espe-
documentation that the segment is less than 1 cially erosions/ulcers.
cm, the term specialized intestinal metaplasia of the Surface Maturation. This refers to the obser-
esophagogastric junction has been suggested (1). vation that, in nondysplastic Barrett's esophagus,
When samples are received that are labeled the nuclei in the basal glands are larger and more
"esophagus" and contain goblet cells with- hyperchromatic than those at the surface. Surface
out documentation of segment length, the nuclei are generally arranged in a monolayer
following comment can be added to reports: featuring polarized basal nuclei. In contrast, the
"Note: The above diagnosis of Barrett's esophagus glands in Banett's esophagus are mildly atypical
is made due to presence of goblet cells (intestinal compared to nearby gastric cardiac or fundic
metaplasia) with the assumption that the biopsies glands, gradually maturing at the surface.
were obtained from columnar mucosa in the distal Architecture. The glandular architecture is
esophagus located at least 1 cm proximal to the top the relationship between glands and the lamina
of the gastric folds as per 2016 American College of propria, and also encompasses the shapes of the
Gastroenterology (ACG) guidelines" (1). glands. Increased numbers of glands and changes
If a sample is labeled "gastroesophageal junc- in their shape are architectural alterations. In
tion" and goblet cells are present, an alternate nondysplastic mucosa, glands are round with
comment can be added: "Note: This biopsy shows minimal budding set in abundant lamina pro-
gastric-type mucosa with scattered goblet cells. The pria. Crowding of normal-appearing glands is a
diagnosis in this case depends on the location of this mild architectural abnormality and a reparative
biopsy. If this biopsy was taken from the tubular change. Crowding of abnormal glands is a hall-
esophagus at least 1 cm above the gas tric folds, it mark of dysplasia or early carcinoma. Cribriform
shows Barrett mucosa of the distinctive type. If this glands, cystic dilation, and necrotic luminal
biopsy was taken from the gastric cardia, it shows debris are severe architectural abnormalities.
intestinal metaplasia of the gastric cardia" (1) . Cytologic Features. These are the key com-
Barrett's esophagus (with goblet cells) can ponent of diagnosis. Some degree of nuclear
be documented in about 1 to 2 percent of the alterations is inherent in nondysplastic Banett
general population (6) and in up to about 15 mucosa, especially in the basal zone and in the
percent of people with reflux disease. Banett's columnar epithelium adjoining squamous mu-
esophagus does not alter life expectancy (7). A cosa. Cytologic atypia can indicate dysplasia,
large Danish epidemiologic study found that reparative changes, and inherent changes in the
the annual incidence of progression to cancer deeper glands of Banett's esophagus .
in Banett's esophagus was only 0.12 percent. As Determining whether dysplasia is low- or
a follow-up, the AGA analyzed a large data set high-grade is somewhat subjective since altera-
and noted that in the first 5 years of follow-up, tions arise along a morphologic continuum, a
a mean 0.19 percent annual rate of progression point noted years ago (12). An important

23
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-6
BARRED'S ESOPHAGUS, NEGATIVE FOR DYSPLASIA
A: There are well formed goblet cells that are crisply
delineated, separated by foveolar cells. This pattern
is termed incomplete intestinal metaplasia because
the tissue has incompletely transformed from gastric
foveolar type epithelium to intestinal type epithelium
that features a brush border. This is the characteristic
appearance of mucosa in Barrett's esophagus.
B: The presence of the four lines/levels in areas
that are well oriented provides a clue that there is no
dysplasia. The first line/row is formed by the neutral
mucin cap of the foveolar cells, the second by the base
of the surface mucin cap, the third by the cytoplasm and
the fourth by the row of stratified nuclei.
C: Note the lines that are demonstrated at low
magnification in "B". These helpful lines vanish in zones
that are tangentially embedded such as the area at the
left part of the field.

component of assessment of cytologic features than that of the deeper glands. The architecture
is evaluation of the alignment of one nucleus is normal, and thus there is abundant lamina
compared to others: nuclear polarity. When propria. The cytologic features are normal;
"normal polarity" is present, the long axes of mitoses may be present in deeper glands. Indi-
the nuclei are perpendicular to the basement vidual nuclei have smooth nuclear membranes,
membrane, and the nuclei are aligned paral- and nucleoli, if present, are small with smooth
lel to one another. "Loss of nuclear polarity" outlines. Nuclear polarity is maintained.
indicates loss of this orientation and a random A characteristic feature of nondysplastic Bar-
array of the nuclei in relation to the basement rett mucosa, even reactive changes, is the pres-
membrane and one another. ence of four lines/tiers. The first tier is formed by
the surface gastric foveolar type mucin droplet,
Barrett's Esophagus: Negative for Dysplasia
the second by the base of the foveolar mucin vac-
Nondysplastic Barrett's esophagus (fig. 2-6) uole, the third by the cytoplasm below the mucin
shows surface maturation such that the nucle- vacuole, and the last by the row of nuclei. This
ar-to-cytoplasmic ratio of surface cells is lower pattern is fourid in both gastric cardiac mucosa

24
Barrett's Esophagus and Esophageal Neoplasms

Figure 2-7
BARRED'S ESOPHAGUS, INDEFINITE FOR DYSPLASIA
Left: Given that the term "indefinite" is used, this can be a subjective interpretation. In this sample, there is poor orientation
and plentiful inflammation such that the surface epithelial changes are concerning but could be reparative.
Right: This is a high magnification of the lesion seen on the left. The surface lines are absent but the surface nuclei are
not particularly enlarged and the lamina propria is quite inflamed.

(see chapter 1, fig. 1-11) and nondysplastic Some examples of low-grade dysplasia are
Barrett mucosa (fig. 2-6B). Using this criterion essentially indistinguishable from tubular ad-
helps reduce the number of lesions regarded as enomas (fig. 2-IOC). Do not use the term tubular
"indefinite for dysplasia" as below. adenoma. Polypoid low-grade columnar epithelial
dysplasia is preferable.
Barrett's Esophagus: Indefinite for Dysplasia
Barrett's Esophagus: High-Grade Dysplasia
The indefinite category describes cases that
have deeper cytologic changes suggestive of Surface maturation is lacking in high-grade
dysplasia, but that show surface maturation but dysplasia (figs. 2-11-2-16). Architecturally, there
are often cases in which inflammation obscures may be crowding of cytologically abnormal
the findings (figs. 2-7, 2-8). glands, but many cases show normal amounts of
lamina propria. Prominent nucleoli are usually
Barrett's Esophagus: Low-Grade Dysplasia
absent. They tend to be present in the setting
The surface appears similar to the underlying of either reactive changes or invasive carcino-
glands at low magnification in low-grade dyspla- ma, both of which are associated with ulcers.
sia (figs. 2-9, 2-10), or displays only slight matu- Markedly enlarged hyperchromatic cells that
ration. Lamina propria should be identifiable extend to the surface characterize high-grade
between glands. The cytologic changes should dysplasia (HGD). Loss of nuclear polarity is a
extend at least focally to the surface. Finding foci key feature of HGD.
with abrupt transition between non-dysplastic HGD associated with certain features should
and dysplastic epithelium is a clue that the suggest early carcinoma (figs. 2-17-2-19). Con-
changes are indeed neoplastic as opposed to reac- cerning features are: 1) cribriform architecture,
tive. Superficially located nuclei can be irregular, 2) dilated tubules containing necrotic debris,
hyperchromatic, mildly enlarged, and may show 3) associated ulceration, 4) neutrophils within
some degree of stratification and mucin loss. dysplastic glands, and 5) extension of single
Imp01tantly, the four lines/four tiers are lost. neoplastic cells into the overlying squamous

25
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-8
BARRETT'S ESOPHAGUS, INDEFINITE FOR DYSPLASIA
Left: The gland in the center co uld be dysplastic but there is ample inflammation and some surface maturation. The
surface lines are lost but the surface nuclei are smaller tha n the nuclei in the deeper glands.
Right: This is a higher magnification of the left figure .

.,, . (

. ..

r:
:t·
: I \

Figure 2-9
BARRETT'S ESOPHAGUS, LOW-GRADE DYSPLAS IA
Left: The surface lines are absent and there is minimal inflammation. The surface nuclei have similar appearances to
th e deep ones. There is no loss of nuclear polarity in that the long axes of the nuclei are perpendicular to the basement
membrane. This lesion h as overt intestinal type differentiation.
Right: Note the sharp abrupt transition between the gastri c foveo lar type epithelium on the top and the dysplasia at the
left and bottom. A few dysplastic glands are also present at the lower right.

26
Banett's Esophagus and Esophageal Neoplasms

.
r •
0 ~
·~ •4

Figure 2-10

BARRETT'S ESOPHAGUS, LOW-GRADE DYSPLASIA


A: A striking abrupt change is seen in the epithelium at the right.
B: This is a high magnification image of the surface. The surface lines are absent in the well oriented zone on the right
but the long axes of the nuclei are perpendicu lar to the basement membrane, which supports an interpretation of low-grade
dysplasia rather than high-grade dysplasia.
C: This lesion resembles a colorectal tubular adenoma.

27
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-11
BARRETT'S ESOPHAGUS, HIGH-GRADE DYSPLASIA
Left: In this example, the glands are pressed together ("back to back") but it is easy to trace the outline of each tubule.
Even at this magnification, it is apparent that surface nuclear polarity is lost and many of the nuclei are round and detached
from the basement membrane.
Right: This is a high magnification view of the surface showing rounded nuclei.

r
,._
..
.. - •
0 •
'
• • •• •
.. •

0

• ,. • ~


"•
' 0
• (I

• '
!,
~
..
• '

q

'

Figure 2-13
SQUAMOUS EPITHELIUM, p53 STAIN
It is entirely normal to find labeling in the basal
layer of squamous epithelium, which is the proliferative
compartment of the squamous epithelium. In this layer,
Figure 2-12 the TP53 gene is "turned on" and performing its duties of
abundant production of normal p5 3 (a tumor suppressor
BARRETT'S ESOPHAGUS, HIGH-GRADE DYSPLASIA molecule) rather than mutated and producing an aberrant
This is a p53 immunostain. There is strong nuclear gene product (protein) that is not degraded and thus
labeling in most of the cells . accumulates in the nuclei.

28
BaiTett's Esophagus and Esophageal Neoplasms

Figure 2-14 Figure 2-15


HIGH-GRADE DYSPLASIA AND BARRED'S ESOPHAGUS, HIGH-GRADE DYSPLASIA
INTRAMUCOSAL CARCINOMA Note the nuclear membrane alterations.
There is high-grade dysplasia at the upper right but the
area in the center has poorly formed tubules with luminal
necrosis, feature of early lamina propria invasion. Note the
cardiac glands at the upper left.

Figure 2-16
BARRED'S ESOPHAGUS,
HIGH-GRADE DYSPLASIA,
p53 "NULL" PADERN
The cardiac glands at the lower
right show weak labeling in th eir
proliferative compartment in keeping
with physiologic activity of the
p53 protein. However, in the high-
grade dysplasia at the left , there
is complete absence of nuclear
labeling, supporting biallelic loss of
the TP53 gene. This complete loss,
when encountered, is a wonderful
confirmation of an interpretation of
high-grade dysplasia or carcinoma.

29
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-17
BARRETT'S ESOPHAGUS, HIGH-GRADE DYSPLASIA AND INTRAMUCOSAL CARCINOMA
Left: This lesion shows at least high-grade dysplasia but areas in the center in which the glands are smeared and the presen ce
of glands with neutrophils suggest that there is early lamina propria invasion. Separating the two is of little importance in
the era of endoscopic treatment and indeed this is an area of interobserver variability.
Right: This is a PAS/Alcian blue stain of the lesion seen at left. Note the presence of both acid (blue) and neutral (pink) mucin.

Figure 2-18
BARRETT"S ESOPHAGUS,
HIGH-GRADE DYSPLASIA AND
INTRAMUCOSAL CARCINOMA
This lesion has a pS3 "null" pat-
tern. Note the internal control of weak
labeling in the proliferative com-
partm ent of a few entrapped cardiac
glands at the right.

30
Barrett's Esophagus and Esophageal Neoplasms

Figure 2-19
ESOPHAGEAL ADENOCARCINOMA, PAGETOID SPREAD INTO
SQUAMOUS EPITHELIUM BY A DEEPLY INVASIVE CARCINOMA
Left: The intraepithelial cells are seen in clusters and singly and have mucin vacuoles. Encountering such cells on a
mucosal sample is evidence that a deeply invasive adenocarcinoma is present.
Right: This is a PAS/Alcian blue stain from the adenocarcinoma depicted on the left.

epithelium in a Pagetoid pattern. A Pagetoid managed identically (with mucosal ablation/


pattern is always associated with deeply invasive endoscopic resection) (1,5).
carcinomas whereas the others overlap with fea-
Variant Dysplasia and Carcinoma Patterns
tures ofintramucosal carcinoma (fig. 2-19) (13).
Some lesions show basal pit alterations
lntramucosal Carcinoma
with surface maturation. The term basal oypt
Distinguishing HGD from the earliest carcino- dysplasia (BCD) has been used to describe this
mas (invasion through the basement membrane finding (note that the term oypt is inaccurate
into the lamina propria or muscularis mucosae; since the cardia has pits, but it could be argued
Tla) is difficult. Such cases show effacement that, since the tissue has intestinal metaplasia,
of lamina propria architecture and a syncytial then the pits have become crypts) (figs. 2-22,
growth pattern, back-to-back microglands, cells 2-23) (14). In some cases of BCD, the nuclear
with prominent nucleoli, and an intermingling alterations are identical to those of HGD. The
of single cells and small clusters within the lami- term "indefinite for dysplasia" can be applied to
na propria (figs. 2-20, 2-21). Desmoplasia is either these cases and regarded as an interim diagnosis
absent or minimal. Some observers believe that until conventional dysplasia can be confirmed
the earliest sign of intramucosal carcinoma is or excluded and resampling can be suggested in
the presence of glands with abnormal cytology a note. Although the use of the p53 staining is
(round hyperchromatic nuclei) that have begun not warranted for all cases (2), BCD often shows
to "grow sideways" instead of perpendicular to nuclear labeling in the affected pits.
the surface. Interobserver variability is a factor Other examples of dysplasia lack classic
in diagnosis of intra mucosal carcinoma in small intestinal type differentiation, showing dif-
biopsies. However, this distinction is less im- ferentiation along gastric foveolar or pyloric/
portant than it was in the past since both high cardiac type lines (figs. 2-24-2-26) . Reports
grade dysplasia and intramucosal carcinoma are of "non-adenomatous" (15) or "gastric" (16)

31
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-20
INTRAMUCOSAL CARCINOMA
A: Note the syncytial growth pattern in this example.
B: Although this lesion proliferates as angulated tubules invading into the muscularis mucosae, there is no desmoplasia.
C: Note the lack of desmoplastic response to the glands that invade the muscularis mucosae.
D: This lesion consists of back-to-back glands, some with luminal necrosis. This case also features promin ent Russell
bodies (plasma cells with ab undant intracytoplasmic globules of immunoglobulin) at the top left. This phenomenon can be
mistaken for signet ring cell carcinoma but note that th e nuclei are ti ny.

32
Banett's Esophagus and Esophageal Neoplasms

Figure 2-21
INTRAMUCOSAL CARCINOMA
Left: Many of the glands are oriented parallel to the surface, an abnormal architectural pattern.
Right: Note the nucleoli in the gland in the center of the image and the truncated gland at the left that is oriented parallel
rather than perpendicular to the surface.

Figure 2-22 Figure 2-23


INTRAMUCOSAL CARCINOMA BASAL PATTERN DYSPLASIA, p53 STAIN
This lesion is easy to diagn ose as neoplas tic but notice Diffuse strong staining in a basal lesion supports an
that the surface is uninvolved. This is an unusual pattern. interpretation of dysplasia. In this case, there is striking
Often it is unclear whether atypical glands that mature at nuclear enlargement. There are no good criteria for grading
the surface are neoplastic or reparative. Sometimes recuts but whether this pattern is interpreted as low or high
resolve th e issue but, in many cases, an interpretation of grade dysplasia, the interpretation will prompt additional
indefinite for dysplasia is in order. sampling.

33
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-24
LOW -GRADE DYSPLASIA,
GASTRIC FOVEOLAR PATTERN
A: This is an uncommon pattern with
pure gastric foveolar differentiation but
hyperchromatic nuclei at the surface. The
nuclei are slightly stratified.
B: There are no goblet cells and each
lesional cell has an apical mucin cap in the
manner of gastric foveolar cells.
C: This is a PAS/ Alcian blue stain. Note
the prominent foveolar type mucin at the
left. There are no well-formed goblet cells .

•B

describe these lesions that lack the classic grade dysplasia) there are closely packed tubules.
stratification of intestinal type dysplasia. Such Glandular crowding is far less prominent in
cases seem to account for 10 to 15 percent of intestinal-type dysplasia. Rare lesions have pure
dysplastic lesions. Lesions with cardiac/pylor- gastric foveolar differentiation (fig. 2-24), but
ic differentiation consist of numerous, tiny many lesions have combinations of the various
glands that appear bland at low magnification, types of epithelium. Occasionally dysplasia spreads
but that display nuclear alterations at high laterally over the surface of non-neoplastic cardiac
magnification. Criteria to grade such lesions or cardia-oxyntic glands (fig. 2-27).
are not well-established (11), but essentially,
Treatment Issues
in dysplasia with gastric type differentiation,
since the nuclei are round and in a monolayer In the past esophagectomy was the sug-
rather than stratified, the criteria for HGD simply gested treatment for HGD since there were no
involve loss of polarity whereas at baseline (low- better alternatives. As techniques have evolved,

34
Barrett's Esophagus and Esophageal Neoplasms

Figure 2-25
HIGH-GRADE DYSPLASIA, GASTRIC TYPE DIFFERENTIATION
Left: There are tiny tubules lined by a monolayer of hyperchromatic cells with round nuclei.
Right: This is a high magnification view of the lesion seen on the left.

Figure 2-26
HIGH-GRADE DYSPLASIA, PYLORIC/CARDIAC TYPE DIFFERENTIATION
Left: The lesion is polypoid and complex, consisting of closely packed tubules.
Right: This is a MUC6 immunostain, performed strictly for interest.

35
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-27
HIGH-GRADE DYSPLASIA EXTENDING ONTO THE SURFACE OF CARDIAC TYPE MUCOSA
Left: This pattern can also be seen extending onto the surface of oxyntic mucosa.
Right: High magnification of the case seen on the left.

endoscopic treatment is now the standard carcinoma are the most likely to have recurrent
of care both for HGD and early carcinomas and persistent lesions (22) .
(1 ,5,17). Once occult, more deeply invasive
Endoscopic Mucosal
carcinoma is excluded, endoscopic resection for
Resection (EMR) Specimens
visible lesions followed by ablation using radiof-
requency ablation (RFA) is the current protocol. To perform an EMR, the endoscopist injects
Other available endoscopic treatments in- the submucosa with fluid and then applies a
clude multipolar electrocoagulation, argon cap to the lesion followed by suction through
plasma coagulation, photodynamic therapy, the endoscope, thereby creating an artificial
and cryotherapy. RFA has become the preferred "polyp" that is then resected. The EMR speci-
technique since there are fewer complications men allows for good characterization of dys-
associated with it than the others (18,19), but plasia and neoplasia but there are pitfalls: The
some patients require other ablation methods. surface epithelial layer of the EMR sample may
Although in theory these techniques could be damaged by application of the suction cup
fail to ablate dysplastic mucosa underneath such that dysplasia must be evaluated in the
squamous mucosa (buried BE) this is a minor absence of an intact surface.
issue in practice (20). Endoscopic submucosal Duplicated muscularis mucosae is very com-
dissection, a more time-consuming method mon in Barrett's esophagus (see chapter 1, figs.
of removal of macroscopic lesions, results in a 1-5, right; 1-6). The original muscularis mucosae
greater likelihood of achieving negative mar- is accompanied by a second delicate smooth
gins but is really not necessary for esophageal muscle layer closer to the luminal surface, a fea-
lesions, for which EMR is adequate (21) . ture found in over 90 percent of resection sam-
Recurrences of Barrett mucosa following ples (24) and nearly 70 percent of EMR samples
ablation are common although recurrence of (25) . Duplicated muscularis mucosae creates a
neoplasia is unusual. Regardless, continued pitfall when examining superficial biopsies since
surveillance is indicated (22,23). Not surprising- lamina propria that underlies the more superfi-
ly, those patients with extensive intramucosal cial duplicated layer of muscularis mucosae is

36
Banett's Esophagus and Esophageal Neoplasms

Figure 2-28
SQUAMOUS DYSPLASIA
Left: The alterations involve less than half the thickness of the epithelium in low grade displacia
Right: The alterations involve the entire thickness of the epithelium in high grade displacia.

readily mistaken for submucosa. Awareness of this /

phenomenon prevents diagnosis of submucosal SQUAMOUS PRECURSOR LESIONS AND


invasion (Tl b) in patients whose invasive carcino- SQUAMOUS CELL CARCINOMA
ma is restricted to the lamina propria (Tla). This
Precursor Lesions
is a critical distinction because T1a lesions can be
treated endoscopically whereas submucosal inva- Epithelial changes confined the bottom half
sive lesions (T1b) often require esophagectomy. of the epithelium are graded as low-grade dys-
Cautery results in contraction of the muscu- plasia (intra-epithelial neoplasia terminology
laris mucosae, pulling the lateral edges of the is preferred in Europe) and into the top half
sample together (see chapter 1, figs. 1-5, right; as high-grade dysplasia (fig. 2-28). It should be
1-6) resulting in the false impression that lateral noted, however, that severe epithelial changes
margins are instead deep margins. in the bottom half of the epithelium often sug-
An attempt should be made to characterize gest an adjoining unsampled carcinoma that has
the depth of any invasion in EMR samples in ad- spread in the space between the basement mem-
dition to assessing the margins. It is our practice brane and the normal squamous epithelium.
to provide a description, such as "invading the A pattern of hyperkeratosis and hypergranu-
lamina propria" or "invading the space between losis has been termed "esophagealleukoplakia"
the original and duplicated muscularis mucosae, (white plaques are encountered at endoscopy).
T1a"; reporting the precise depth seems of value This pattern is associated with squamous dys-
to facilitate dialogue when patients are referred plasia and carcinoma (26,27) and also termed
between institutions. epidermoid metaplasia. Essentially a granular
layer is seen, whereas it is normally absent
Adenocarcinoma
in esophageal squamous mucosa (fig. 2-29).
Generally, adenocarcinomas are not difficult Genetic alterations detected in epidermoid
to diagnose; desmoplasia is well developed on bi- metaplasia mirror those in associated dysplasias
opsies as soon as submucosal invasion is present. and carcinomas (28).

37
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 2-29
EPIDERMOID METAPLASIA
A: At first glance, this biopsy simply shows the junction of the squamous and cardiac mucosa but the pattern of
hyperkeratosis is unusual.
B: This is high magnification of the lesion seen in "A." An abnormal granular layer is present just beneath the surface
hyperkeratosis.
C: In this case, epidermoid metaplasia is seen on top of high-grade squamous dysplasia.
D: High magnification of the lesion depicted in "C."

son syndrome (dysphagia, iron-deficiency ane-


Squamous Cell Carcinoma
mia, and esophageal webs), are becoming rare
Squamous cell carcinoma (in contrast to ad- in the developed world with the improvement
enocarcinoma) is associated with low socioeco- of overall nutrition. A role for human papillo-
nomic status. Happily, some of the deficiency ma virus (HPV) in esophageal squamous cell
syndromes associated with esophageal squa- carcinoma is debatable. HPV DNA detection
mous cell carcinoma, such as the Plummer-Vin- rates are insignificant (0 to 2 percent) in some

38
Barrett's Esophagus and Esophageal Neoplasms

Figure 2-30
SQUAMOUS CELL CARCINOMA
Left: The appearances are those of squamous cell carcinoma throughout the body.
Right: Keratin pearls are present.

studies from low incidence areas (29,30). On the carcinomas tend to present as a polypoid masses.
other hand, in a study from Mexico, an area of Squamous cell carcinomas of the esophagus,
low tumor incidence, high risk HPV DNA was like squamous carcinomas in other sites, express
detected in 25 percent of esophageal squamous p63, p40, and CKS/6. When these carcinomas
cell carcinomas (31) and rates of up to 50 per- are spindled, melanoma and sarcomas must be
cent are reported. The rate in the United States excluded but it is important to remember that
is on the order of 10 percent (32). most spindle cell lesions of the esophagus are sar-
Squamous carcinomas are often found in comatoid squamous cell carcinomas. Sampling
the middle third of the esophagus. On imaging as much of the overlying squamous mucosa
studies, the presence of an esophageal mass in as possible can sometimes disclose an in situ
the middle third of the esophagus is formed component or a zone of conventional-appearing
by squamous cell carcinoma until proven squamous cell carcinoma.
otherwise, but there are no specific radiologic
features of this type of tumor. Multifocality is OTHER ESOPHAGEAL LESIONS
not uncommon.
Granular Cell Tumor
On biopsies, esophageal squamous carcino-
mas of the esophagus appear similar to squamous Granular cell tumors are rare. The most com-
carcinomas elsewhere (fig. 2-30). They are usually mon sites for them are the tongue and skin (33).
well differentiated with prominent keratiniza- A subset is multicentric and malignant histolog-
tion but can be basaloid, spindled, papillary or ic features and behavior are sometimes reported
verrucous; the latter is virtually impossible to (34). Those of the esophagus account for about 1
diagnose on superficial samples and based only to 2 percent of all granular cell tumors (33), and
on histologic features. Spindle cell squamous cell the esophagus is the commonest gastrointestinal

39
Survival Guide to Gastrointestinal Mucosal Biopsies

•, :·;
'
• ......... I
. ~
' \o
...·'·,••... ......-
'v • ~

' '
''
... · ".
' '
.•
': ..... ;, .....
'I
.~~~ .. '
~ ;:· ,.; ""

..
"'

. ~' _. -' "' - •t, , I I


' ~ ~~~ V
' .~ .
' ,;: .·.
t T ' ...
;· • '
•\· ,, I
'
' ..
.• -
...._ \ )
:
1 " ' ''

Figure 2-31
GRANULAR CELL TUMOR
Left: There is a proliferation of squamous epithelium overlying the lesion.
Right: The cells have granular cytoplasm and small pyknotic nuclei. Compare their sizes to those of the overlying
squamous basal cell nuclei.

site (35). Most arise in the distal esophagus and it is prudent to scan the lamina propria in bi-
about 5 to 10 percent are multicentric. There is opsy samples showing hyperplastic squamous
a female predominance and these tumors are epithelium.
overrepresented in Mrican-Americans compared Granular cell tumors express S-100 protein,
to whites. Rare cases require radical surgery but calretinin, inhibin alpha, myelin basic protein,
many are managed endoscopically. Rare histo- and CD68/Kp1, but not muscle markers and
logically malignant examples have been report- melanoma markers. Unfortunately, like alveolar
ed in the esophagus (36,37); there is a report of soft part sarcoma (ASPS), they can also react
a large cytologically bland example that spread with TFE3 antibodies.
to the liver (38).
Leiomyoma
The microscopic features of granular cell
tumor in the esophagus are identical to those Leiomyoma is the most common esophageal
at other sites. Esophageal granular cell tumors spindle cell tumor of the esophagus, arising in
are centered in the submucosa, with extensions a relatively young population (median age, 35
into mucosa and muscularis propria. They are years) (39) with a male predominance. Leiomy-
well-marginated but not encapsulated. They omas of the esophagus are, by definition, benign.
consist of plump closely packed cells with abun- They consist of cells with eosinophilic cytoplasm,
dant amphophilic granular cytoplasm (fig. 2-31). and are positive for desmin and alpha-smooth
Nuclei are small to pyknotic, with occasional muscle actin (alpha-SMA), and negative for CD117
nucleoli. Associated squamous ("pseudoepitheli- and CD34. The key pitfall in diagnosing esopha-
omatous") hyperplasia in the overlying mucosa geal leiomyomas is that if immunolabeling for
is common, which can raise the possibility of CD 117 and DOG 1 is performed, it highlights
squamous cell carcinoma on superficial biopsies. Cajal cells that are either entrapped or prolifer-
Often, since the squamous epithelium is thick, ate with the lesion (fig. 2-32). For this reason, a
only a few lesional cells are present on biopsies; confident morphologist eschews these stains.

40
Barrett's Esophagus and Esophageal Neoplasms

.. .( .I
!•

..
I •

·. . I·
'
~
I
/

i'

..
'!
,
' .

. I • -·
f
" ' I .,
... I
'
/
~ :
. r

' .
. li) .·
Figure 2-32

ESOPHAGEAL LEIOMYOMA
A: The lesion is not particularly cellular, and consists of cells with small nuclei and brightly eosinophilic cytoplasm.
B: Note that the cytoplasm contains delicate longitudinal striations typical of smooth muscle differentiation.
C: This is a desmin stain of the lesion seen in "A" and "B."
D: This is a CD117 stain of the lesion seen in figures "A" and "B." Ignore the mast cells and incorporated Cajal cells.
With this morphology, there is no need to stain. Esophageal gastrointestinal stromal tumors are vanishingly rare and usually
overtly malignant.

41
Survival Guide to Gastrointestinal Mucosal Biopsies

REFERENCES

1. Shaheen N], Falk GW, Iyer PG, Gerson LB, histopathologic features and near-universal as-
American College of Gastroenterology. ACG sociation with underlying esophageal adenocar-
clinical guideline: diagnosis and management cinoma. Am] Surg Pathol2008;32:1068-1074.
ofBarrett's esophagus. Am] Gastroenterol2016; 14. Lomo LC, Blount PL, Sanchez CA, et aL Crypt
111:30-50; quiz 51. dysplasia with surface maturation: a clinical, patho-
2. Panarelli NC, Yantiss RK Do ancillary studies aid logic, and molecular study of a Barrett's esophagus
detection and classification of Banett's esopha- cohmt. Am] Surg Pathol2006;30:423-435.
gus? Am] Surg Pathol 2016;40:e83-93. 15. Rucker-Schmidt RL, Sanchez CA, Blount PL, et al.
3. Glickman ]N, Chen YY, Wang HH, Antonioli Nonadenomatous dysplasia in Barrett's esophagus:
DA, Odze RD. Phenotypic characteristics of a a clinical, pathologic, and DNA content flow cyto-
distinctive multilayered epithelium suggests that metric study. Am] Surg Pathol2009;33:886-893.
it is a precursor in the development of Barrett's 16. Mahajan D, Bennett AE, Liu X, Bena], Bronner
esophagus. Am] Surg Pathol2001;25:569-578. MP. Grading of gastric foveolar-type dysplasia in
4. Fitzgerald RC, di Pietro M, Ragunath K, et al. Banett's esophagus. Mod Pathol 2010;23:1-11.
British Society of Gastroenterology guidelines 17. Pech 0, May A, Manner H, et al. Long-term effica-
on the diagnosis and management of Barrett's cy and safety of endoscopic resection for patients
esophagus. Gut 2014;63:7-42. with mucosal adenocarcinoma of the esophagus.
5. American Gastroenterological Association, Gastroenterology 2014; 146:652-660.
Spechler S], Sharma P, Souza RF, Inadomi ]M, 18. Shaheen NJ, Sharma P, Overholt BF, et al. Radio-
Shaheen N]. American Gastroenterological As- frequency ablation in Barrett's esophagus with
sociation medical position statement on the dysplasia. N Engl] Med 2009;360:2277 -2288.
management of Barrett's esophagus. Gastroen- 19. Spechler S], Fitzgerald RC, Prasad GA, Wang KK.
terology 2011;140:1084-1091. . History, molecular mechanisms, and endoscopic
6. Ronkainen], Aro P, Storskrubb T, et al. Prevalence treatment of Banett's esophagus. Gastroenterol-
of Barrett's esophagus in the general popula- ogy 2010;138:854-869.
tion: an endoscopic study. Gastroenterology 20. Bronner MP, Overholt BF, Taylor SL, et al. Squa-
2005;129: 1825-1831. mous overgrowth is not a safety concern for
7. Sikkema M, de ]onge P], Steyerberg EW, Kui- photodynamic therapy for Barrett's esophagus
pers EJ. Risk of esophageal adenocarcinoma and with high-grade dysplasia . Gastroenterology
mortality in patients with Banett's esophagus: a 2009;136:56-64; quiz 351-352.
systematic review and meta-analysis. Clin Gas- 21. Terheggen G, Horn EM, Vieth M, et al. A ran-
troenterol Hepatol 2010;8:235-244; quiz e232. domised trial of endoscopic submucosal dissec-
8. Kroep S, Lansdorp-Vogelaar I, Rubenstein JH, tion versus endoscopic mucosal resection for
et al. An accurate cancer incidence in Barrett's early Barrett's neoplasia. Gut 2017;66:783-793.
esophagus: a best estimate using published data 22. Agoston AT, Strauss AC, Dulai PS, et al. Predictors of
and modeling. Gastroenterology 2015;149:577- treatment failure after radiofrequency ablation for
585; quiz e514-575. intramucosal adenocarcinoma in Barrett's esoph-
9. Vaughan TL, Fitzgerald RC. Precision preven- agus: a multi-institutional retrospective cohort
tion of oesophageal adenocarcinoma. Nat Rev study. Am] Surg Pathol2016;40(4):554-562.
Gastroenterol Hepatol 2015;12:243-248. 23 . Krishnamomthi R, Singh S, Ragunathan K, A Katzka
10. Montgomery EA, Bronner MP, Goldblum JR, et D, KWangK, G Iyer P. Risk ofrecunence ofBanett's
al. Reproducibility of the diagnosis of dysplasia esophagus after successful endoscopic therapy.
in Barrett's esophagus: a reaffirmation. Hum Gastrointest Endosc 2016;83:1090-1106.
Pathol 2001;32:368-3 78. 24. Abraham SC, Krasinskas AM, Correa AM, et al.
11. Vieth M, Montgomery EA, Riddell RH. Observa- Duplication of the muscularis mucosae in Bar-
tions of different patterns of dysplasia in Barrett's rett's esophagus: an undenecognized feature and
esophagus-a first step to harmonize grading. its implication for staging of adenocarcinoma.
Cesk Patol2016;52:154-163. Am] Surg Pathol 2007;31:1719-1725.
12. Reid B], Haggitt RC, Rubin CE, et al. Observer 25 . Lewis]T, Wang KK, Abraham SC. Muscularis mu-
variation in the diagnosis of dysplasia in Banett's cosae duplication and the musculo-fibrous anom-
esophagus. Hum Pathol 1988;19:166-178. aly in endoscopic mucosal resections for Barrett's
13. Abraham SC, Wang H, Wang KK, Wu TT. Paget esophagus: implications for staging of adenocarci-
cells in the esophagus: assessment of their noma. Am] Surg Pathol2008;32:566-571.

42
Barrett's Esophagus and Esophageal Neoplasms

26. Singhi AD, Arnold CA, Crowder CD, Lam-Himlin 33 . Lack EE, Worsham GF, Callihan MD, et al. Granu-
DM, Voltaggio L, Montgomery EA. Esophageal lar cell tumor: a clinicopathologic study of 110
leukoplakia or epidermoid metaplasia: a clinico- patients. J Surg Oncol1980;13 :301-316.
pathological study of 18 patients. Mod Pathol. 34. Fanburg-Smith JC, Meis-Kindblom JM, Fante R,
2014;27(1):38-43 . Kindblom LG. Malignant granular cell tumor of
27. Taggart MW, Rashid A, Ross WA, Abraham SC. soft tissue: diagnostic criteria and clinicopathologic
Oesophageal hyperkeratosis: clinicopathological as- correlation. Am] Surg Pathol1998;22:779-794.
sociations. Histopathology. 2013;63(4):463-473. 35. Johnston ], Helwig EB. Granular cell tumors of
28. Singhi AD, Montgomery EA. Genetic alterations the gastrointestinal tract and perianal region: a
in epodermoid metaplasia of the esophagus and study of 74 cases. Dig Dis Sci 1981;26:807-816.
the associated neoplasms. JOURNAL?? 2017. 36. Ohmori T, Arita N, Uraga N, Tabei R, Tani M,
29·. Turner JR, Shen LH, Crum CP, Dean PJ, Odze Okamura H. Malignant granular cell tumor of
RD. Low prevalence of human papillomavirus the esophagus. A case report with light and
infection in esophageal squamous cell carcino- electron microscopic, histochemical, and im-
mas from North America: analysis by a highly munohistochemical study. Acta Pathol Jpn
sensitive and specific polymerase chain reaction- 1987;37:775-783.
based approach. Hum Pathol1997;28:174-178. 37. Yoshizawa A, Ota H, Sakaguchi N, et al. Ma-
30. Poljak M, Cerar A, Seme K. Human papillomavi- lignant granular cell tumor of the esophagus.
rus infection in esophageal carcinomas: a study Virchows Arch 2004;444:304-306.
of 121 lesions using multiple broad-spectrum 38. Christopher PR, Kingsley PA, Singh Bedi H,
polymerase chain reactions and literature review. Singh Kwatra K, Rathore S, Das KC. Large Mid-
Hum Pathol 1998;29:266-271. esophageal granular cell tumor: benign versus
31. Herrera-Goepfert R, Lizano M, Akiba S, Car- malignant. Rare Tumors 2015;7:5772.
rillo-Garcia A, Becker-D' Acosta M. Human 39 . Miettinen M, Sarlomo-Rikala M, Sobin LH, La-
papilloma virus and esophageal carcinoma in sota]. Esophageal stromal tumors: a clinicopath-
a Latin-American region . World J Gastroenterol ologic, immunohistochemical, and molecular
2009;15:3142-3147. genetic study of 17 cases and comparison with
32. Syrjanen K. Geographic origin is a significant esophageal leiomyomas and leiomyosarcomas.
determinant of human papillomavirus preva- Am J Surg Pathol 2000;24:211 -222.
lence in oesophageal squamous cell carcinoma:
systematic review and meta-analysis. Scand J
Infect Dis 2013;45:1-18.

43
3 NON-NEOPLASTIC STOMACH

There are many obscure causes of gastritis, but has a more bubbly appearance compared to
most stomach biopsies can be addressed by pay- the crisply delineated apical mucin caps that
ing attention to just a few features in each and characterizes surface foveolar cells throughout
every case. In many centers in the United States, the stomach. Overall, the antrum appears very
the majority of gastric biopsies are essentially similar to the cardia but differs by having a more
normal. For example, in the population having organized architecture and by the endocrine cell
gastric biopsies at]ohns Hopkins, H. pylori is only constituents (figs. 3-1, 3-2). Most importantly,
detected in 6 percent (1) and autoimmune gastri- antral mucosa produces gastrin via the G cells
tis in about 2 to 3 percent (2,3). There are a few and the cardia does not. Gastrin from the antrum
unusual types of gastritis also encountered but stimulates enterochromaffin-like (ECL) cells in
most samples show normal histologic features the gastric body and fundu s to produce hista-
or minimal alterations. All biopsies, however, mine, which in turn stimulates parietal cell to
should be scanned carefully since it is easy to secrete acid for digestion. Gastrin also has a tro-
overlook tiny foci of signet ring cell carcinoma. phic effect on parietal cells and is necessary to
There are several patterns of gastritis and gas- maintain them in good working order. Some of
tropathy that can be recognized but the first step the antral endocrine cells secrete somatostatin
is to understand the difference between the mu- (D cells), but this is not of much interest for the
cosa of the body and fundus (oxyntic mucosa) purpose of interpreting gastric biopsies. There
and that of the antrum, which appears similar to should be only sparse lymphocytes, plasma
that of the cardia but differs in important ways. cells, and eosinophils in the lamina propria of
The questions that should be posed as a gastric the stomach and a few intraepitheliallympho-
biopsy is evaluated are as follows: cytes. It should be difficult to find apoptotic
bodies in the pits. There should only be 1 or
WHAT TYPE OF GASTRIC possibly 2 in a pinch biopsy (4).
MUCOSA IS PRESENT? As noted in chapter 2, mucosa from the gas-
Normal antral mucosa consists of surface tric body has a surface with foveolar cells and
foveolar cells and antral glands that are re- complex pits. The pits contain parietal and chief
plete with neutral mucin but their cytoplasm cells. Between the pits and the surface are mucus

Figure 3-1
ANTRAL MUCOSA
At low magnification, there
is only minimal inflammation
in the lamina propria. Th ere
are surface fov eolar cells and
mucin-producing antral glands
in the bottom of the im age.
This exa mple has slight reactive
changes but it lacks significant
findings. There is no muscularis
'· •• ,,. mucosae in the sample.
•.
.:._:,..•
. :. ·-,.:~"-.

45
Survival Guide to Gastrointestinal Mucosal Biopsies

•• • I

. ... ,.
. , ·: .:... ·.··:.·. ....
. .,
·.>

' ' \I ,. I ..
. _. ' • ' 1, i- : .
, , I\

Figure 3-2
ANTRAL MUCOSA
Left: This is a chromogranin stain. It labels G cells, which secrete gastrin and D cells, which secrete somatostatin.
Right: This is a gastrin stain. It labels cells situated in between the antral glands at the base and the surface foveolar cells.

neck cells, which have a proclivity to resemble


signet ring cell carcinoma in crushed biopsies
(fig. 3-3). The endocrine cells, as above, secrete
histamine in order to entice the parietal cells
to secrete acid. A common finding in biopsies
of oxyntic mucosa is the alterations associated
with proton pump inhibitor therapy (fig. 3-4).
Similarly, sometimes there are unusual vacu-
olated cell changes that are probably also related
to these medications (fig. 3-5) (5). These effects
need not be reported, but can be striking, con-
sisting of plump apocrine like parietal cells and
dilated oxyntic glands.
The transitional mucosa is found where the
antral and oxyntic mucosa meet, and features
parietal and chief cells as well as a few antral
type glands (fig. 3-6). The architecture should
be orderly. Knowing these normal features is
important because when stomach biopsies are
examined, it is important to decide if parietal
Figure 3-3 cells have been lost if a sample is derived from
CRUSHED OXYNTIC MUCOSA
the gastric body or fundus.
The crushed parietal cells have formed an eosinophilic IS THE MUCOSA INFLAMED?
sludge in the midst of which crushed mucus neck cells
reside. In the clusters at the upper right and center, Uninflamed Mucosa. If the mucosa shows
their appearance is reminiscent of that of signet ring cell scarcely any lamina propria inflammation at
carcinoma cells but they are smaller and not in intact
lamina propria.
low magnification, it is probably normal if it is

46
Non-Neoplastic Stomach

l'\f
..'' ~-·.,'.' ~j

Figure 3-4 Figure 3-5


OXYNTIC MUCOSA WITH OXYNTIC MUCOSA WITH VACUOLATED
PROTON PUMP INHIBITOR EFFECT CELL CHANGE IN PARIETAL CELLS
The cytoplasm of the pink parietal cells has an apocrine This appearance can be alarming. This finding may
appearance. also be associated with proton pump inhibitors.

Figure 3-6
TRANSITIONAL MUCOSA
Left: There are foveolar cells at the surface and antral glands at the base, but scattered parietal and chief cells can be seen
in the zone between the base and surface of the mucosa. The overall architecture of the mucosa is well organized and there
is minimal lamina propria inflammation .
Right: This image allows brightly eosinophilic parietal cells to be readily recognized but there are antral glands composed
of cells with bubbly mucinous cytoplasm at the base. The surface cells each have an apical mucin cap. Knowledge of the
appearances of these cells types is useful when gastric neoplasms are encountered.

47
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-7
CHEMICAL GASTROPATHY
A: The basic architecture of the antral
mucosa is maintained but the foveolar
type glands at the surface have a corkscrew
configuration.
B: This example is particularly striking.
The mucin loss in the foveolar cells results
in a high nuclear to cytoplasmic ratio but
the architecture is maintained. There is
only mild lamina propria inflammation
and many of the nuclei seen at low
magnification are stromal cell nuclei.
C: This is the surface of the lesion seen
in "B." The nuclei are enlarged but their
nuclear membranes are smooth. Another
name for this type of injury is reactive
gastropathy.

oxyntic mucosa. If it is antral mucosa, the most consist of foveolar cell mucin loss, proliferation
likely findings is so called reactive gastropathy of the foveolar glands imparting a corkscrew
or chemical gastropathy (figs. 3-7, 3-8). Some configuration to them, and the presence of
observers refer to this pattern as chemical gastritis smooth muscle cells in the lamina propria.
even though there is minimal inflammation. Some patients with portal hypertension
Reactive gastropathy is associated with nonste- display this pattern associated with vascular
roidal anti-inflammatory drug (NSAID) use (6) ectasia (fig. 3-9) without fibrin thrombi. If fibrin
and bile reflux. Persons with Barrett's esophagus thrombi are present, the key consideration is
tend to reflux their duodenal contents into their so-called watermelon stomach (7), based on the
stomachs, and this refluxate in turn passes into endoscopic appearance that is reminiscent of
the esophagus. A reactive gastropathy (chemi- the outside of a watermelon (stripes) (figs. 3-10).
cal gastropathy/gastritis pattern) is observed in This condition is probably related to mucosal
these people and is quite common. The findings prolapse but patients present with anemia and

48
Non-Neoplastic Stomach

. - . _-·...
...

-
' -' ... - - . '• ,· - ~··..:~~
Figure 3-8
EROSIVE CHEMICAL GASTROPATHY
Left: An erosion refers to necrosis that involves part of the mucosa (the mucosa consists of the epithelium, lamina propria
and muscularis mucosae) whereas an ulcer describes injury that extends into the submucosa or deeper. This example shows
striking reactive epithelial changes.
Right: This is a higher magnification of the lesion seen on the left. The degree of cytologic atypia raises the possibility
of malignancy but note that the architecture is intact. The nuclear to cytoplasmic ratio of the cells is only mildly increased .

Figure 3-9
PORTAL GASTROPATHY
Left: At a glance this process is very much like that of chemical gastropathy but the presence of the dilated lamina propria
vessels suggests portal hypertension.
Right: The lamina propria smooth muscle is a feature of chemical gastropathy but the dilated capillary to th e left of the
smooth muscle is a feature of portal gastropathy.

49
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-10
GASTRIC ANTRAL VASCULAR ECTASIA (WATERMELON STOMACH)
Left: This endoscopic image shows erythematous stripes in the antral mucosa, an appearance reminiscent of the pattern
of stripes on a watermelon rind.
Right: The appearance is similar to th_at of chemical or portal gastropathy but differs by featuring fibrin thrombi, as seen
in the lower right part of the field.

tend to be older women with collagen vascular Inflamed Mucosa. Are both oxyntic and
diseases. Some of them require mucosal ablation antral mucosa present in the sample and do
or even resection to control their anemia. The the site agree with the sample label provided
mucosa of the antrum features fibrin thrombi. by the endoscopist?
Curiously, hemosiderin is absent. Although there are many causes of gastritis,
Sometimes a chemical gastropathy pattern those commonly encountered in a United States
can be accompanied by acute inflammation or population are Helicobacter pylori gastritis and
erosions or ulcers but there is minimal chron- autoimmune gastritis. Helicobacterpylori gastritis
ic inflammation (fig. 3-8). Reactive epithelial is more common than autoimmune gastritis but
changes can be striking but the normal archi- both are regularly encountered in daily practice.
tecture of the glands is preserved. An erosion As above, in the population at ]ohns Hopkins,
is an injury that results in damage to some of about 6 percent of gastric biopsies show infec-
the mucosal layer whereas an ulcer penetrated tion with H. pylori gastritis and 2 to 3 percent
into the submucosa. A cause of normal appear- show autoimmune gastritis.
ing body mucosa that belies a true condition
is gastric siderosis, which can result from iron H. PYLORI GASTRITIS
overload due to hemochromatosis or from mul- H. pylori gastritis results in a superficial pattern
tiple transfusions. The appearance is quite subtle of lamina propria chronic inflammation, some
but it is a good practice habit to check for it in acute inflammation in the pits, and tends to af-
deep pits (fig. 3-11) (8). The pattern contrasts fect the antrum more than the body (figs. 3-13).
sharply with that of iron pill gastritis, in which Some patients have infection with an alternate
the findings are mostly in the surface gastric species, H. heilmannii (figs. 3-14), which produces
epithelium with some deeper extension, and a less dramatic gastritis than H. pyloli (10), and
reactive epithelial changes tend to be prominent seems to be associated with exposure to house
(fig. 3-12) (9). cats. After treatment, organisms tend to "hide"

so
Non-Neoplastic Stomach

- ,. .. ,.
• -
.,.,
.. >· ,·

• ,' '·· ...


.:.
~
I
.:
> •
'

..
1

c
Figure 3-11
GASTRIC SIDEROSIS
A: The appearance is unremarkable at low power. This condition is seen in oxyntic mucosa.
B: This is high magnification of the deep portion of the biopsy seen in "A." Note the brownish hemosiderin granules.
This sample was from a patient who had received multiple blood transfusions but these findings can also accompany
hemochromatosis.
C: This is an iron stain. Note the bottom heavy staining qualities.

51
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-12
·''
IRON PILL GASTRITIS
,·.
.,..;l"
A: The iron is m ostly deposited at th e surface and is ))J
go lden brown. ! '\
•'
B: Note the striking associated reactive epithelial changes.
C: This is a periodic acid-Schiff (PAS)/AB stai n, which
hi gh lights the gastric foveolar cells.
D: Since th e epithelia l cells sh owed strik ing reacti ve
changes, the conce rn fo r carcin o m a was rai sed. This
reticulin sta in shows investm ent of individual glands in
keeping with a reactive lesion rath er th an a carcinom a.
E: This is an iron stain. It con t rasts rem ark ably with th e
iro n stain from gastric siderosis (fig. 3-11 , right), showing
predo min ant ly extracellular iron.

.,.,.E-.

52
Non-Neoplastic Stomach

Figure 3-13
HELICOBACTER PYLORI GASTRITIS
A: There is striking lamina propria chronic inflammation in this antral sample.
B: Organisms are adherent to th e surface foveolar cells in an area with abundant mucus.
C: Numerous organisms coa t the surface.
D: This is the gastric body from th e sa me patient as the one whose gastric antral body is seen in "A." It is far less inflamed.

53
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-14
HELICOBACTER HEILMANN/1 GASTRITIS
A: The organisms are longer and more
tightly coiled than those of H. pylori. They are
treated the same and result in similar 1isks.
B: This is a Diff Quik stain, which high-
lights the tightly coiled configuration of the
organisms.
C: This is an immunostain for H. pylori,
which cross reacts with H. heilmannii.

in the body, and immunolabeling may be required of H. pylori (1) : 1) chronic inflammation: increased
to detect them. In general, it is not necessary to inflammation, primarily plasma cells, within the
perform "up front" testing for H. pylori since it is lamina propria in a patchy loose distribution
readily seen on routine stains (11), but staining without epithelial destruction. Review at 10x
biopsies that have inflammation in which no magnification is required to identify clusters; 2)
organisms are detected can increase the yield (1). inactive chronic gastJitis: dense lymphoplasmacyt-
Organisms are sought in areas of abundant mucus ic lamina propria infiltrate readily identifiable
associated with foveolar cells. It is a waste of time to at 4x that infiltrates and destroys glands; and 3)
search for them in normal gastric mucosa. Organ- active chronic gastritis: neutrophilic infiltration of
isms are only detected if there is inactive chronic the epithelium in addition to lymphoplasmacyt-
gastritis or active chronic gastritis, rather than ic infiltration of the lamina propria.
simply lamina propria chronic inflammation. Over time, persons with H. pylori gastritis can
Here are some definitions derived from a study develop intestinal metaplasia, primarily in the

54
Non-Neoplastic Stomach

Figure 3-15
ANTRAL MUCOSA WITH INACTIVE CHRONIC GASTRITIS AND INTESTINAL METAPLASIA
Left: Generally H. pylori organisms are absent in zones of intestinal metaplasia and when intestinal metaplasia is prominent
in the antrum, organisms tend to be found instead in the body. This case is interesting because incomplete and complete
intestinal metaplasia are present in adjacent glands . The one at the top perfectly recapitulates small intestine and there is
even a brush border. The gland on the bottom with intestinal metaplasia shows both goblet cells and gastric foveolar type
cells such that is has incompletely transformed to intestinal epithelium . Epidemiologically, the incomplete type is more
likely to progress to adenoca rcinoma .
Right: Complete intestinal metaplasia is present in the gland at the top and incomplete intestinal metaplasia is found in
the gland at the bottom, with goblet cells interspersed with gastric foveolar cells.

antrum, but sometimes extending proximally. It Over time, patients become anemic, but their
tends to be of the "complete" type, with changes lack of vitamin B12 leads to central nervous
that recapitulate small intestine, including the system complications that can include paresthe-
presence of a brush border (fig. 3-15). sias, memory loss, and loss of balance (12,13).
They can be treated with large doses of vitamin
AUTOIMMUNE GASTRITIS B12 (sometimes by intramuscular injection) to
Autoimmune gastritis (or autoimmune met- prevent these complications.
aplastic atrophic gastritis; AMAG) results in Unfortunately, autoimmune gastritis is easy
autoimmune damage to oxyntic glands with re- to overlook (figs. 3-16, 3-17). The damage to
sultant loss of the parietal cell mass and gradual the parietal cells can result in biopsies that, at
inability to absorb both iron (since the resultant a glance, resemble antrum, but to the trained
high gastric pH impedes iron absorption) and eye, the appearance is quite different from that
vitamin B12. The condition is most common in of antrum. The glands are disorganized and the
middle aged to elderly females, but men are not oxyntic glands become replaced by metaplastic
spared. Many data suggest that it is triggered by glands, namely intestinal metaplasia, pancreatic
early H. pylori gastritis in the susceptible host. metaplasia, and pyloric metaplasia. Some refer

55
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-16
AUTOIMMUNE GASTRITIS
A: This is the antral biopsy from a patient with autoimmune gastritis. It is normal. Note the surface foveolar cells and
deep antral glands that contain mucin.
B: This is a gastrin stain for the biopsy shown in" A." Note the orderly distribution of the G cells in the zone between the
antral deep glands and the surface foveolar type cells.
C: This is a biopsy of the gastric body. The parietal cells are gone and the glands that have replaced them are arranged in
a disorderly fashion. The inflammation is in the deep part of the sample where the oxyntic glands previously resided such
that th e antral type glands are metaplastic (pyloric metaplasia). Those features are all clues that this sample is not from the
true antrum should a sample be labeled in an unclear fashion.
D: This example shows prominent intestinal metaplasia and bottom heavy inflammation. This sample was from the
gastric body.

56
Non-Neoplastic Stomach

,. '',J,>

Figure 3-17
AUTOIMMUNE GASTRITIS
A: The parietal cells are gone and replaced by disorganized metaplastic antral type glands (pyloric metaplasia) and
intestinal metaplasia.
B: This example shows pyloric and pancreatic acinar metaplasia . This biopsy from the gastric body lacks parietal cells.
C: This is a gastrin stain from the gastric body. It is wholly negative, which can be helpful in confirming that the sample
is from the gastric body. Note the disorganized metaplastic pyloric type glands.
D: This is a chromogranin stain from the sample that was stained for gastrin in "C." There is linear (>6 cells in a line) and
nodular enterochromaffinlike (ECL) cell hyperplasia. These are the cells that are stimulated by gastrin to secrete histamine,
which, in turn, stimulates parietal cells to secrete acid. Since parietal cells are absent, this feedback loop is interrupted and
the affected patients have hypergastrinemia.

57
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-1 8 .
GASTRIC TRANSITIONAL MUCOSA
IN A PATIENT WITH
EVOLVING AUTOIMMUNE GASTRITIS
A: There is inactive chronic gastritis and
this patient in fact had H. pylori gastritis
in addition to evolving autoimmune
gastritis. The H. pylori was detected on
an immunostain. Sometimes eradication
of H. pylori can forestall development of
autoimmune gastritis.
B: Parietal cells are present but the
inflammation is bottom heavy.
C: The deep glands are undergoing
destruction predominantly by lymphoid
cells, but focal acute gastritis is also present.

\··" ...' ' .' { ;:t.~; ..~: =:.,'

~~.~· -~.. ~..:-1 """ ·ti< • ::.:rc{v} · \, ..~.; · "" ·.... ·. . •· · ~~ ·· :~


r;.'>i-..~jt,'fr/;;'{f'~:\1~~~: ·-.· . "'.\'l~g
'";:-·-"··.;·.~!:{~J?-'-''/
~··· • he ~P' ikf"Y <'t f
v:~~~:.~E"tft :'
~~\ 11D.':o.,.
;"!~;·L .·~--.
~_!.:.' ~

to the last type as pseudopyloric metaplasia is full loss of parietal cells by noticing foci of
because there is no gastrin production. When damage to parietal cells (figs. 3-18, 3-19) .
learning to recognize this pattern, it can be The loss of parietal and chief cells in patients
helpful to perform gastrin labeling (a negative with autoimmune gastritis results in a loss of
gastrin stain confirms that the sample if from acid production. This in turn cases the antral
oxyntic mucosa that has lost its parietal cells G cells to secrete abundant gastrin in order to
rather than from the antrum) to recognize better stimulate acid production so the patient
the affected fragments when endoscopists put becomes hypergastrinemic. Since there are no
samples from both the body and antrum in the parietal cells, the hypergastrinemia does not
same container. Frequently the patient's antrum result in acid secretion. Gastrin exerts its effect
appears normal and the gastric body appears by stimulating the ECL cells of the gastric body
damaged. The inflammation is "bottom heavy". to secrete histamine, so hypergastrinemia in
It is possible to detect the findings before there these patients results in hyperplasia of ECL

58
Non-Neoplastic Stomach

A-
...·' ·-
.:; • : ••· .. ;.+
., ,~· :~:- ...

:.
...,
....
•:
. .., .,,,
... ·.
·~

"'.~:1:
.· . :·.. <
• : .~ ...... -:./ • :! '!
J . '•,

.. -~-.... -i'

Figure 3-19
GASTRIC BODY MUCOSA IN A PATIENT WITH EVOLVING AUTOIMMUNE GASTRITIS
A: Lymphoid cells are attacking parietal cells at the lower right part of the image.
B: High magnification of the area in figure "A."
C: Note the damaged appearance of the parietal cells. Some are apoptotic.
D: ECL cell hyperplasia can be detected in such cases on chromogranin staining.

cells, which can ultimately lead to very indo- However, if a report simply states "autoim-
lent well-differentiated neuroendocrine tumors. mune gastritis," there is a good chance that the
Patients can have other types of polyps too (2) . colleague receiving the report will not be aware
These changes happen over a period of many of a need for vitamin B12 replacement or the
years so there is usually ample chance to pre- patient may only have iron deficiency. Because
vent the neurodegenerative complications if the of this, reports should probably indicate that the
findings are recognized . patient may only have iron deficiency anemia

59
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-20
LYMPHOCYTIC GASTRITIS PATTERN
A: The lamina propria shows both
superficial and deep inflammation in this
antral biopsy. Celiac disease is a common
association with a lymphocytic gastritis
pattern.
B: There is mucin loss (a reactive
change) in the foveolar cells and striking
intraepitheliallymphocytosis.
C: Note the intraepitheliallymphocytosis
in both the surface epithelium and the
epithelium in the glands.

and that the patient is at risk for both pernicious sartan (Benicar), and all of which are used as
anemia arid various polyps and neoplasms (3). antihypertension drugs (15).
The associated neoplasms and polyps are fea-
tured in chapter 4. IS THERE PROMINENT PIT APOPTOSIS?
Finding prominent pit apoptosis in gastric
LYMPHOCYTIC GASTRITIS samples always requires correlation with the
This is a pattern of inflammation rather than clinical history. The key causes: graft versus
a real diagnosis. It simply indicates that there are host disease (usually unaccompanied by inflam-
abundant intraepitheliallymphocytes in gastric mation), any of a large number of medications
epithelium (fig. 3-20). It is strongly associated (mycophenolate, idelalisib, many monoclonal
with celiac disease (14), as well as with H. pylori antibodies, angiotensin receptor II antagonists,
gastritis, and several medications, including and many others) (15) . If the apoptosis is ac-
proton pump inhibitors and drugs in the class companied by abundant inflammation and loss
of medications termed angiotensin receptor II of glands, autoimmune pangastritis can also be
antagonists, the prototype of which is olme- considered (it responds to steroids) (16) but it is

60
Non-Neoplastic Stomach

Figure 3-21 Figure 3-22


MYCOPHENOLATE-ASSOCIATED GASTRIC INJURY CYTOMEGALOVIRUS GASTRITIS
There is striking pit apoptosis. This pattern can also There is an apoptotic body in the pit at the lower right
accompany graft versus host disease, viral infections, and a but there are no viral inclusions in this field . When this
host of other medications. Mycophenolate is administered pattern is seen in a patient in whom a diagnosis of graft
to persons who have had solid organ transplantation. versus host disease (GVHD) or medication-associated
gastropathy is being considered, it is important to also
consider cytomegalovirus infection. Apoptotic pit damage
a diagnosis of exclusion. Prominent pit apopto- is the hallmark of the findings associated with several
sis is also likely in the setting of cytomegalovirus medications and is often also a prominent feature in
cytomegalovirus-associated injury.
infection (figs. 3-21-3-23) .
Collagenous gastritis is a poorly understood
condition that can manifest in children and 30 eosinophils per high power field in at least
adults and is an immune disorder with many 5 fields is abnormal (17).
associations, including medications (fig. 3-24). Patients with renal failure often have disor-
The sample features prominent intraepithelial ders of calcium metabolism, which can result in
lymphocytosis and a thick, irregular subepithe- deposition of calcium in their gastric mucosa,
lial collagen table. which should not be mistaken for parasitic or-
ganisms (fig. 3-26). This differs from the injury
OTHER CONDITIONS associated with doxycycline, which features
Finding prominent eosinophils in the epithe- vessels with fibrinoid change (fig. 3-27) (18,19).
lium or muscularis mucosae is usually an allergic Lanthanum carbonate, administered to reduce
condition (fig. 3-25) and may accompany eo- serum phosphate in renal failure patients, can
sinophilic esophagitis. The normal number of accumulate in histiocytes in the gastric lamina
eosinophils in the lamina propria is not clear propria and is illustrated in the small intestinal
and it probably varies geographically, but some chapter (20). Recall that patients with renal fail-
authors have suggested that finding more than ure also develop hyperkalemia and are treated

61
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-23
CYTOMEGALOVIRUS GASTRITIS
Left: In most organs, cytomegalovirus preferentially infects endothelial cells but, in the stomach, epithelial cells are
frequently the target, as in this example. There is a background of gastritis and several enlarged nuclei are apparent.
· Right: There is an infected cell in the center with nucleomegaly and an intranuclear inclusion.

Figure 3-24
COLLAGENOUS GASTRITIS
Left: Note the striking thickness and irregularity of the subepithelial collagen.
Right: In addition to prominent subepithelial collagen, there is intraepitheliallymphocytosis.

62
Non-Neoplastic Stomach

Figure 3-25
EOSINOPHILIC
GASTRITIS PATTERN
There are eosinophils present in
the muscularis mucosae, where they
are not a normal finding, and they are
numerous in the lamina propria.

Figure 3-26
MUCOSAL CALCINOSIS
Left: There is a background of antral mucosa with chemical gastropathy and numerous deposits of dystrophic calcium.
The variable size is against an interpretation as parasites.
Right: High magnification of the calcium deposits. These are found in patients with disorders of calcium metabolism so
most have renal failure, but occasionally, this pattern is seen in persons with parathyroid adenomas.

63
Survival Guide to Gastrointestinal Mucosal Biopsies

!·.· ·
.\ }

,. ·~
'I '•
i .. ~

Figure 3-27
DOXYCYCLINE-ASSOCIATED INJURY
Left: Note the mineralized appearing surface of this oxyntic mucosa.
Right: The superficial capillaries have an appearance of fibrinoid necrosis.

with sodium polystyrene sultanate (kayexalate), Lastly, unusual infections can be encountered
which can be associated with mucosal necrosis in the stomach. In patients with gastric outlet
throughout the gastrointestinal tract. It was il- obstruction or other causes of gastric stasis
lustrated in chapter 1 (fig. 1-19) but can also be (such as diabetes), Sarcina ventriculi is an asso-
encountered in gastric biopsies (21). ciated organism. Treating it seems to alleviate
Gastric Crohn's disease can be virtually im- symptoms, but Koch's postulates have not been
possible to diagnose in the absence of pertinent fulfilled (fig. 3-31) (22). In men who have sex
history but tends to be characterized by patchy with men (MSM) and are infected with human
inflammation, as it is in the intestines (fig. 3-28). immunodeficiency virus, syphilis gastritis is a
Occasionally numerous Russell bodies are en- possibility (fig. 3-32) although syphilis is far
countered in plasma cells in the gastric lamina more likely to be encountered as an agent in
propria but most examples are incidental findings proctitis in MSM patients (23).
unassociated with plasma cell disorders (fig. 3-29).
Amyloidosis can also affect the stomach (fig. 30).

64
Non-Neoplastic Stomach

--
Figure 3-28
GASTRIC CROHN'S DISEASE
Other than the granuloma,
the features are those of severe
act ive chronic gastritis and the
diagnosis can only be made as a
clinicopathologic correlation.

Figure 3-29
RUSSELL BODY GASTRITIS
Left: There are numerous lamina propria plasma cells with prominent cytoplasmic immunoglobulin (Russell bodies).
This is usually an incidental finding.
Right: This is a PAS/ AB stain. Immunoglobulin is PAS positive, which can result in a concern for carcinoma.

65
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 3-30
GASTRIC AMYLOIDOSIS
A: Note the abundant waxy deposition of amyloid in this gastric resection. The amyloid had formed a pseudotumor.
B: There is a foreign body response to the amyloid, with giant cell formation.
C: This is a kappa stain from the lesion depicted in "A" and "B."
D: This is a lambda stain from the lesion seen in "A" and "B.". The process appears to be lambda restricted and thus
reflects a plasma cell disorder.

66
Non-Neoplastic Stomach

Figure 3-31
SARCINA VENTRICULI-ASSOCIATED GASTRITIS
Left: The bacteria are quite large and organized in tetrads that are reminiscent of the chambers of the heart. The organism
is believed to cause disease in ruminants (cattle especially) but may cause disease in persons with delayed gastric emptying
and gastric stasis.
Right: High magnification of the organisms.

Figure 3-32
SYPHILIS GASTRITIS
Left: The inflammation is bottom heavy and has produced a destructive gastropathy.
Right: The lamina propria is expanded with plasma cells and there is apoptotic damage to the pits.

67
Survival Guide to Gastrointestinal Mucosal Biopsies

REFERENCES

1. PittrnanME, KhaiarjianA, WoodLD, Montgomery 12. Stabler SP. Vitamin B12 deficiency. N Engl] Med
EA, Voltaggio L. Prospective identification of Heli- 2013;368:2041-2.
cobacter pylori in routine gastric biopsies without 13. Stabler SP. Clinical practice. Vitamin B12 defi-
reflex ancillary stains is cost-efficient for our health ciency. N Engl] Med 2013;368:149-60.
care system. Hum Pathol2016;58:90-96. 14. Wu TT, Hamilton SR. Lymphocytic gastritis: as-
2. Park ]Y, Cornish TC, Lam-Himlin D, Shi C, sociation with etiology and topology. Am] Surg
Montgomery E. Gastric lesions in patients with Pathol 1999;23:153-8.
autoimmune metaplastic atrophic gastritis 15. Vieth M, Montgomery E. Medication-associ-
(AMAG) in a tertiary care setting. Am] Surg ated gastrointestinal tract injury. Virchows Arch
Pathol 2010;34: 1591-8. 2017;470:245-266.
3. Pittman ME, Voltaggio L, Bhaijee F, Robertson 16. ]evremovic D, Torbenson M, MmTay JA, Burgart
SA, Montgomery EA. Autoimmune Metaplastic LJ, Abraham SC. Atrophic autoimmune pangas-
atrophic gastritis: recognizing precursor lesions tritis: a distinctive form of antral and fundic
for appropriate patient evaluation. Am ] Surg gastritis associated with systemic autoimmune
Pathol 2015;39:1611-20. disease. Am] Surg Pathol2006;30:1412-9.
4. Nguyen T, Park ]Y, Scudiere ]R, Montgomery 17. Lwin T, Melton SD, Genta RM. Eosinophilic
E. Mycophenolic acid (cellcept and myofortic) gastritis: histopathological characterization and
induced injury of the upper GI tract. Am] Surg quantification of the normal gastric eosinophil
Pathol 2009;33: 1355-63. content. Mod Pathol 2011;24:556-63.
5. Rubio CA, Kato Y. Classification of vacu- 18. Xiao SY, Zhao L, Hart], Semrad CE. Doxycycline-
olated cells in the gastric mucosa. J Surg Oncol induced gastric and esophageal mucosal injuries
1987;34:128-32. with vascular degeneration. Am ] Surg Pathol
6. Haber MM, Lopez I. Gastric hist-ologic findings 2013;37:1115-6.
in patients with nonsteroidal anti-inflamma- 19. Xiao SY, Zhao L, Ha.J.t], Semrad CE. Gastric mucosal
tory drug-associated gastric ulcer. Mod Pathol necrosis with vascular degeneration induced by
1999;12:592-8. doxycycline. Am] Surg Pathol2013;37:259-63.
7. Jabbari M, Cherry R, Lough]O, Daly DS, Kinnear 20. Haratake ], Yasunaga C, Ootani A, Shimajiri S,
DG, Goresky CA. Gastric antral vascular ectasia: Matsuyama A, Hisaoka M. Peculiar histiocytic
the watermelon stomach. Gastroenterology lesions with massive lanthanum deposition in
1984;87:1165-70. dialysis patients treated with lanthanum carbon-
8. Marginean EC, Bennick M, CyczkJ, Robert ME, ate. Am J Surg Pathol 2015;39:767-71.
Jain D. Gastric siderosis: patterns and signifi- 21. Abraham SC, Bhagavan BS, Lee LA, Rashid A,
cance. Am J Surg Pathol 2006;30:5 14-20. Wu TT. Upper gastrointestinal tract injury in
9. Abraham SC, Yardley JH, Wu TT. Erosive injury patients receiving kayexalate (sodium polysty-
to the upper gastrointestinal tract in patients rene sulfonate) in sorbitol: clinical, endoscopic,
receiving iron medication: an underrecognized and histopathologic findings. Am J Surg Pathol
entity. Am] Surg Pathol1999;23:1241-7. 2001;25:637-44.
10. Jhala D, Jhala N, Lechago J, Haber M. Helicobacter 22. Lam-Himlin D, Tsiatis AC, Montgomery E, et al.
heilmannii gastritis: association with acid peptic Sarcina organisms in the gastrointestinal tract:
diseases and comparison with Helicobacter pylori a clinicopathologic and molecular study. Am ]
gastritis. Mod Pathol1999;12:534-8. Surg Pathol2011;35:1700-5.
11. Batts KP, Ketover S, Kakar S, et al. Appropriate 23. Arnold CA, Limketkai BN, Illei PB, Montgomery
use of special stains for identifying Helicobacter E, Voltaggio L. Syphilitic and lymphogranu-
pylori: Recommendations from the Rodger C. loma venereum (LGV) proctocolitis: clues to a
Haggitt Gastrointestinal Pathology Society. Am frequently missed diagnosis. Am ] Surg Pathol
] Surg Pathol 2013;37:e12-22. 2013;3 7:38-46.

68
4 GASTRIC POLYPS AND NEOPLASMS

Gastric polyps and neoplasms are very dif- upper endoscopic examinations, usually in
ferent from neoplasms in the small bowel and middle-aged females. Most are small (a few
colon and are somewhat akin to esophageal neo- millimeters), sessile, and dome-shaped (fig. 4-1).
plasms since they usually arise in a background Sporadic FGPs are commonly multiple (usually
of damaged mucosa. A key point is that many a few polyps). There are rare families who have
gastric neoplasms arise in the setting of atrophic a variant of FAP termeci gastric adenocarcinoma
gastritis. As such, if attention is paid to the back- and proximal polyposis of the stomach (GAPPS)
ground flat mucosa, it can be easier to diagnose the (3). In contrast to hyperplastic polyps, FGPs are
polyps and neoplasms that arise. For example, the not associated with any type of inflammatory
following types of polyps and neoplasms, which or atrophic background mucosal pathology and
account for the majority of entities that can be are inversely correlated with H. pylori infection
encountered, are associated with autoimmune (2). They are asymptomatic but some studies
gastritis and H. pylori gastritis (1): 1) Hyperplas- suggest a temporal association between use of
tic polyp: autoimmune and H. pylori; 2) Gastric proton pump inhibitors and the development
adenoma, pyloric type (pyloric gland adenoma): of FGPs (4). Some authors have questioned it
autoimmune; 3) Gastric adenoma, intestinal (5), but most believe that proton pump inhib-
type: autoimmune and H. pylori; 4) Gastric ad- itors prompt the development of fundic gland
enocarcinoma: autoimmune and H. pylori; 5) polyps (6). Histologically, they consist of dilated
Well-differentiated neuroendocrine (carcinoid) oxyntic glands that contain parietal cells with
tumor: autoimmune; and 6) Mucosa-associated apocrine-like snouts (fig. 4-2). Because it is
lymphoid tissue lymphoma: H. pylori. sometimes difficult to distinguish between these
Discussion of every possible neoplasm and two processes, correlation relies chiefly on the
polyp is beyond the range of this slim volume, presence of an endoscopic lesion.
but the commonest can be addressed.
POLYPS
Gastric polyps are common in gastrointes-
tinal biopsies. The term polyp simply means a
projection from the surface, and some are neo-
plasms, as opposed to hamartomas or reactive
nodules. Using data from a large commercial
laboratory, about 6 percent of patients undergo-
ing upper endoscopy have gastric polyps; most
(77 percent) are fundic gland polyps followed by
hyperplastic polyps/polypoid foveolar hyperpla-
sia (17 percent), adenomas (0.69 percent), and
inflammatory fibroid polyps (0.1 percent) (2).
Fundic Gland Polyps
There are two forms of fundic gland polyps Figure 4-1
(FGPs): sporadic, and those associated with
FUNDIC GLAND POLYP
familial adenomatous polyposis (FAP). Sporadic
FGPs are found in about 2 percent of routine This endoscopic image features a pair of dome shaped
polyps.

69
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-2
FUNDIC GLAND POLYP
Left: The polyp arises in uninflamed oxyntic (fundic) mucosa, and is characterized by cystic dilation of the glands.
Right: The cysts are lined by parietal cells.

matory or neoplastic processes. Regardless, in


Hyperplastic Polyps
other studies, they are most strongly associated
Hype1plastic polyps are the second most com- with atrophic gastritis of either autoimmune or
mon gastric epithelial polyps (2,7). They can environmental (e.g., H. pylori-associated) types
range from a few millimeters to large masses that (1). As discussed in more detail in chapter 2, the
mimic carcinoma. They are composed of hy- diagnosis of autoimmune gastritis is suggested
perplastic, elongated, disorganized, and dilated histologicallywhen biopsies show corpus-predom-
foveolae within an edematous, inflamed stroma inant gastritis, glandular atrophy, and intestinal,
(fig. 4-3). Some have intestinal metaplasia, and pyloric, or pancreatic metaplasia (11).
rare cancers arise in them (8). As for when to Knowing these associations with background
diagnose foveolar hyperplasia versus hyperplas- mucosal pathology can be important since stom-
tic polyps, a simple approach can be taken: if a ach polyps encountered in patients with juvenile
polyp is seen at endoscopy and biopsy shows a polyposis or Peutz-Jeghers polyposis can appear
proliferation with prominent gastric foveolar essentially identical to gastric hyperplastic polyps
cells, hyperplastic polyp can be diagnosed. How- (figs. 4-4, 4-5). Sadly, the best clue is knowledge
ever, some such polyps simply reflect mucosal that the patient has one of those syndromes
prolapse, especially in the antrum (9). diagnosed by small bowel or colon samples
Hyperplastic polyps can arise anywhere in the (12,13). However, if there are available biopsies
stomach, with a slight preference for the antrum, of flat mucosa, the absence of atrophic gastritis
and appear in multiples in approximately 20 is a clue to a syndromic polyp.
percent of cases. Though often considered to
Adenomas
arise as a result of inflammation due to reflux
of duodenal contents (and so called chemical Gastric adenomas are essentially polypoid
gastropathy) or gastritis, a study by Melton and dysplasia and most are a result of inflammation,
Genta (10) that included 330 of such polyps which leads to metaplasia and neoplasia (14).
and over 120,000 controls found no association Most are associated with background atrophic
between gastric hyperplastic polyps and inflam- gastritis (either H. pylori-driven or autoimmune

70
Gastric Polyps and Neoplasms

Figure 4-3
GASTRIC HYPERPLASTIC POLYP
Left: There is a disorganized proliferation of glands lined by foveolar cells.
Right: This unusual example harbors a gastric adenocarcinoma (left side of image) but the polyp itself on the right features
disorganized glands in edematous stroma lined by foveolar cells. ·

gastritis/pernicious anemia) and they have in-


testinal differentiation such that they resemble
colon adenomas. However, in contrast to colon
adenomas, they arise in abnormal background
mucosa that may also have flat dysplasia. In this
respect, they are very much like the previously
termed DALM (dysplasia-associated lesion or
mass) of ulcerative colitis or Crohn's colitis. Of
course most polyps in the colon are sporadic in
otherwise normal colons, which is very different
from the situation with stomach adenomas.
As above, gastric adenomas with intestinal
differentiation have goblet cells and appear
very similar to colorectal tubular adenomas (fig.
4-6). Some have admixed intestinal and gastric
type differentiation, and these seem more likely
to progress to adenocarcinoma that those that
have pure intestinal differentiation (15).
In patients with autoimmune gastritis, since
there is pyloric metaplasia, occasionally ade-
nomas with pyloric type differentiation are en-
countered (figs. 4-7, 4-8) (16-18). Like adenomas
with intestinal differentiation, pyloric gland Figure 4-4
adenomas can progress to adenocarcinoma.
GASTRIC JUVENILE POLYP
This type consists of closely packed tubules
This lesion is difficult to distinguish from a gastric
composed of cells with ground glass cytoplasm hyperplastic polyp but it arose in otherwise normal gastric
that lack either apical mucin caps like those seen mucosa in a patient known to have juven ile polyposis.

71
Survival Guide to Gastroirztestinal Mucosal Biopsies

Figure 4-5
GASTRIC PEUTZ-JEGHERS POLYP
A: This example shows arborizing cords of smooth muscle separating groups of gastric glands but were the endoscopist to
sample only the superficial parts of the polyp, the appearance would be indistinguishable from that of a hyperplastic polyp.
B: This is a periodic acid-Schiff (PAS)/ Alcian blue stain from the lesion shown in figure "A." It highlights the foveolar cells.
C: The cords of smooth muscle can be seen separating zones of gastric mucosa.
D: This is a high magnification of the base of a gastric Peutz-Jeghers polyp. It shows the smooth muscle component well.

72
Gastric Polyps and Neoplasms

\ .•.

Figure 4-6
GASTRIC ADENOMA, INTESTINAL TYPE
Left: This is from a resection. The adenoma has an appearance similar to that of a colorectal tubular adenoma. This
particular example shows herniation of the adenoma glands into the submucosa (gastritis cystica profunda pattern). The
muscularis propria can be seen at the bottom of the image.
Right: This image is from a clinical polyp. The adenoma has high grade dysplasia with loss of nuclear polarity and goblet
cells. Were this sample taken from flat mucosa, the lesion would be classified as high grade dysplasia. Both polypoid and flat
gastric dysplasia nearly always arise in damaged gastric mucosa with metaplasia.

Figure 4-7
PYLORIC GLAND ADENOMA
When these polyps are encountered
in the stomach, there is frequently a
background of autoimmune gastritis
with pyloric metaplasia . These neoplasms
harbor GNAS mutations and consist of
closely packed tubules composed of cells
with basally placed nuclei in a monolayer
(rather than stratified as in intestinal
adenomas) with ground glass cytoplasm
that Jacks conspicuous mucin.

73
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-8
PYLORIC GLAND ADENOMA
Left: This image shows the nuclear and cytoplasmic characteristics.
Right: This example shows low grade dysplasia on the right and high grade dysplasia on the left. The nuclei at the left
have small nucleoli and some have lost t_heir polarity.

on foveolar cells or brush borders or goblet cell


type mucin. The nuclei are round and arranged ADENOCARCINOMA AND MIMICS
in a basal monolayer. Loss of polarity of these The estimate for 2016 was about 26,000 new
cells indicates high-grade dysplasia. cases of gastric carcinoma (63 percent in men)
Management of both intestinal type and with about 11,000 deaths in the United States
pyloric gland type adenomas requires complete (20). A wide geographic variation in incidence
polypectomy and mapping biopsies of the flat of gastric cancer is known; high-risk areas in-
mucosa to assess for flat dysplasia. clude China,] apan, Eastern Europe, and parts of
On the other hand, there are gastric adenomas South and Central America (21). Gastric cancer
that are quite rare, quite indolent and unlikely to is the fifth most common cancer worldwide
progress to gastric carcinoma. This is the gastric after cancers of the lung, breast, colorectum,
foveolar type, which is essentially analogous to and prostate. This is major change since 1975,
sporadic colorectal adenomas. This rare type when the stomach was the commonest site of
arises in normal (usually oxyntic type) mucosa cancers worldwide.
and has pure gastric foveolar differentiation (fig. Usually, diagnosing gastric cancer on mucosal
4-9). It is most common in patients with familial biopsies is not difficult, but there a few pitfalls.
adenomatous polyposis. In fact, the fundic gland In chapter 3, reactive changes that can mimic
polyps in patients with familial adenomatous dysplasia and early carcinomas are highlighted.
polyposis often have surface foveolar cells with Additionally, xanthomas (fig. 4-10) and so-
low grade dysplasia that appears identical to called signet ring cell change (fig. 4-11) (22-25),
gastric foveolar adenomas and these types can seen usually in mucosa with ulcers or ischemic
· be impossible to separate. However, this is of no changes, are pitfalls to be aware of. Signet cell
consequence because gastric foveolar adenomas change differs dramatically from the findings in
and fundic gland polyps with surface dysplasia hereditary diffuse gastric cancer in patients with
never progress to cancer in the sporadic setting CDHl (the gene that encodes for E-cadherin)
(19) and only rarely do in syndromic cases. germline mutations (fig. 4-1 2).

74
Gastric Polyps and Neoplasms

Figure 4-9
GASTRIC ADENOMA, GASTRIC FOVEOLAR TYPE
Left: This type of adenoma is rare in general but seen with some freq u ency in patients with familial adenomatous polyposis.
The surface foveo lar cells have dysplastic nuclei. The flat mucosa is uninflamed.
Righ t: This is the surface of the lesion seen on the left. Note that each cell has the usual apical mucin cap of a foveolar
cell but t h e nuclei are enlarged and hyperchromatic.

Figure 4-1 0

GASTRIC XANTHOMA
Left: These are incidental lesions.
Right: The lesion consists of lipid-laden macrophages.

75
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-11
SIGNET RING CELL CHANGE
A: In damaged columnar mucosa such as
this (with hemorrhage and ischemic injury),
when epithelial cells slough from the basement
membrane into the lumina, they round up and
become edematous, thereby mimicking the
appearance of the malignant cells in signet ring
cell carcinoma. However, they are not in the
tissue itself, but sloughed into the lumina and
the basement membranes are intact.
B: The sloughed damaged cells have an alanning
appearance but they are in the lumina rather than
in the lamina propria.
C: This e-cadherin stain highlights the cell
membranes of the damaged sloughed cells. 'J Q

There is massive lamina propria edema but the •·•it


concerning cells are not in the lamina propria.
~

Figure 4-12
IN SITU SIGNET RING CELL
CARCINOMA IN A PATIENT WITH
A CDH1 GERMLINE MUTATION
The lesion consists of tumor cells that
are situated between the normal epithelium
and the basement membrane in undamaged
mucosa.

76
Gastric Polyps and Neoplasms

Figure 4-13
GASTRIC CARCINOMA
Left: This would conform to the classic intestinal type gastric adenocarcinoma, presenting as a bulky mass in gastric
mucosa. The spleen is at the top.
Right: This thickened stomach lacks a discrete mass and would be expected to have a diffuse type carcinoma that infiltrates
randomly through the tissue.

Adenocarcinomas of the stomach can have exhausted. Starting with a pankeratin, S-100
intestinal differentiation as well as mixtures of in- protein, and CD20 and additional unstained
testinal and foveolar differentiation and pyloric sections is a good approach. Starting with specific
type differentiation but they are usually easy to keratins (such as CK7 /CK20) will miss poorly
recognize as carcinomas. Poorly cohesive carci- differentiated carcinomas. Expression of S-100
noma, some with signet ring morphology, can be protein captures melanomas and CD20 captures
easy to overlook. The key is to study any area of a the most likely type of lymphoma (diffuse large
gastric biopsy that shows architectural alterations B cell lymphoma). Never order a vimentin. It
at low magnification (figs. 4-13-4-17). wastes the tissue and yields no information.
Some neoplasms can be poorly differentiated
and are difficult to diagnose as carcinoma, lym- METASTATIC CARCINOMAS
phoma, melanoma, or even something else on The commonest metastases to the stomach
small biopsies. When this is the case, it is very are from breast carcinoma, renal carcinoma,
important to avoid performing a "shotgun" im- and hepatobiliary carcinomas, but any type of
munolabeling panel since the tissue may become carcinoma can spread to the stomach (figs. 4-18,

77
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-14
GASTRIC CARCINOMA
A: There is a poorly differentiated carcinoma at the upper right. This cancer arose in the gastric body but note that there
are no parietal cells although there is some pancreatic acinar cell metaplasia in the lower center and lower right. Those
features indicate autoimmune gastritis. In general the adenocarcinomas arising in patients are of the intestinal type (and
thus appear similar to colorectal carcinomas), that is not invariably the case .
B: This is a subtle lesion, seen in scarred mucosa.
C: This is a high magnification of the area seen in the bottom right of figure B. The carcinoma cells are singly distributed
in the lamina propria. A residual damaged gland appears at the lower left and allows comparison with the nuclei of the
carcinoma cells, which contain cytoplasm with mucin vacuoles.

78
Gastric Polyps and Neoplasms

-~
/t, t
'1 .
, /
...•
I'
•• ,
Figure 4 -15
GASTRIC CARCINOMA
A: At first glance, there appears to be chronic inflammation in the lamina propria but the cells are too large to be plasma cells.
B: This is high magnification of the lamina propria seen in A. The nuclei of the lamina propria smooth muscle cells at
the left center (from mucosal prolapse) offer a good internal size control, as do the epithelial cell nuclei.
C: This is an AEl/3 keratin stain from the lesion seen in A & B. It shows the raggedly infiltrating appearance of the
carcinoma.

79
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-16
GASTRIC CARCINOMA
A: This carcinoma has infiltrated into the omentum.
B: High magnification of the carcinoma seen in A.
C: This is a keratin stain of the carcinoma seen in A. The highlighted spindle cells (top left) are submesothelial cells that are
found beneath the mesothelial cells that coat the omentum. This staining can be a pitfall in nonneoplastic omental samples.

80
Gastric Polyps and Neoplasms

Figure 4-17
GASTRIC CARCINOMA
Left: This is a DOG 1 stain. About a third of gastric adenocarcinomas and more esophageal adenocarcinomas express
DOGl. Beware on small samples!
Right: This is a KIT stain of the adenocarcinoma seen on the left.

4-19). The most important one to consider in atrophic gastritis with hypergastrinemia (this is
any patient in which a new diagnosis of gastric the most common); 2) type 2: tumors associated
carcinoma is contemplated is breast carcinoma, with Zollinger-Ellison syndrome or multiple
particularly lobular carcinomas. Have a very low endocrine neoplasia (and thus with hypergastrin-
threshold to perform GATA3 staining in a woman emia from a gastrinoma); and 3) type 3: sporadic
in whom gastric carcinoma is considered. lesions (without hypergastrinemia).
Gastric WDNETs associated with hypergas-
NEUROENDOCRINE TUMORS trinemia (types 1 and 2) are relatively benign,
Though historically known as carcinoid whereas sporadic lesions require aggressive sur-
tumors, these are classified as well-differentiat- gical management. Gastric WDNETs, however,
ed neuroendocrine tumors (WDNET or NET G1, can be managed initially by endoscopic excision
neuroendocrine tumor, grade 1) by the World of accessible tumors, evaluation of the setting in
Health Organization (WHO) (26). Using pre- which they have arisen, and follow-up by regu-
cise terminology, a "carcinoid" tumor is sero- lar endoscopic surveillance. A striking increase
tonin-producing and is associated with clinical in gastric WO NETs over 50 years was reported in
evidence of carcinoid syndrome and hence the 2004, including an 800 percent increase in gas-
well-differentiated neuroendocrine tumors ter- tric carcinoids in white women (27). However,
minology has been adopted officially, but the patients did not routinely undergo endoscopies
term "carcinoid" remains in widespread use. 65 years ago and tiny WDNETs are commonly
Gastric WDNETs can be divided into three cat- found in women as a result of autoimmune
egories: 1) type 1: tumors associated with chronic gastritis. They were not sought until recently.

81
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-18
PANCREATIC CARCINOMA THAT HAS SPREAD TO TH E GASTRIC ANTRUM
Left: This deceptively bland carcinoma at the bottom center of the biopsy consists of large aberrantly shaped glands.
Right: This is a SMAD4/DPC4 stain of the lesion seen on the left. There is loss of nuclear labeling in the malignant glands.

Figure 4-19
LUNG ADENOCARCINOMA METASTATIC TO THE STOMACH
Left: A few entrapped gastric glands are at the upper left. This metastasis superficially mimics a pyloric gland adenoma.
Right: This is a TTFl stain. The entrapped gastric mucosa lacks th e nuclear labeling.

82
Gastric Polyps and Neoplasms

Figure 4-20
TYPE 1 WELL-DIFFERENTIATED
NEUROENDOCRINE (CARCINOID) TUMOR
A: This neoplasm arose in the gastric body and is at the
bottom of the field. It displays a pseudoglandular pattern.
Note that the overlying gastric mucosa lacks parietal cells
since the patient has autoimmune gastritis.
B: This is a high magnification of the tumor seen in A.
C: This is a chromogranin stain from the tumor seen in
A. There is enterochromaffin-like (ECL) cell hyperplasia in
the overlying gastric mucosa.

As for the "white" women demographic, that liferations because it is not clinically important.
may reflect patterns of patient referral or the It is more important to determine the source of
demographics of the studied population; no the excess gastrin. This is accomplished by at-
racial predilection was found among patients tention to background gastric changes: atrophy
with autoimmune gastritis in a large East coast (fig. 4-20) versus hyperplasia of the acid-pro-
urban study, although females were much more ducing cells. Hyperplasia of intact parietal cells
commonly affected than males (1). is the hallmark of Zollinger-Ellison syndrome,
Type 1 and type 2 carcinoids are essentially in which there is a gastrin-secreting neoplasm
a nuisance with little metastatic risk. Concern (often in the pancreas or duodenum). In such
with the size of a very early lesion is of no value. a case the type 2 WDNET is gastrin negative on
There is no need to be concerned with neuro- immunolabeling since it is composed of ECL
endocrine dysplasia versus neoplasia. The term cells whereas the gastrinoma of the pancreas or
"tumor" can be used when the endoscopist sees duodenum expresses gastrin on immunolabel-
a nodule; there is no need to measure small pro- ing (figs. 4-21, 4-22).

83
Survival Guide to Gastrointestinal Mucosal Biopsies

{
\

t
J

Figure 4-21
TYPE 2 WELL-DIFFERENTIATED NEUROENDOCRINE (CARCINOID) TUMOR
A: This is not a gastric lesion. This is a duodenal gastrinoma in a patient with multiple endocrine neoplasia (MEN) I.
B: This is higher magnification of the neoplasm seen in A.
C: This is a gastrin stain of a duodenal gastrinoma.
D: This is a gastric lesion from a patient with the gastrinoma seen in A and B.

84
Gastric Polyps and Neoplasms

In contrast, type 3 WDNETs (those that are the GI tract is the most common site for MALT
not associated with hypergastrinemia; fig. 4-23) lymphomas. MALT lymphoma accounts for
are likely to be metastatic at presentation (about about half of gastric lymphomas.
70 percent) and associated with patient deaths MALT lymphomas show lamina propria
in about 25 percent of patients. expansion with small uniform cells, some of
Mitotic activity or Ki-67 immunolabeling which have a "halo" around them (fig. 4-24). A
should be used for grading Type 3 tumors clue is that the muscularis mucosae is infiltrated,
(26,28): 1) G1: less than 2 mitoses/10 high
power fields or up to 2 percent Ki-67 index; and
2). G2: mitotic count 2 to 20 per 10 high power
fields or 3 to 20 percent Ki-67 index.
However, up to a third of type 1 WDNETs
show a Ki-67 index that reaches grade 2 and
yet they essentially never cause patient deaths
and only rarely metastasize as above (29). For
this reason, performing Ki-67labeling on type 1
WDNETs can result in misleading information.
Type 1 carcinoids (WDNETs) can express both
hormone receptors and GATA3 such that they
can masquerade as metastatic breast carcino-
mas. Since autoimmune gastritis affects the
same demographic group as metastatic breast
carcinoma, it is important to bear this in mind.
LYMPHOMAS

Mucosa-Associated Lymphoid
Tissue Lymphomas
Low-grade B-cell lymphomas of mucosa-asso-
ciated lymphoid tissue (MALT) are believed to
arise within organized lymphoid tissue in the
gastric mucosa that is most frequently acquired
in response to H. pylori infection. They are also Figure 4-22
termed "extranodal marginal zone lymphomas. " TYPE 2 WELL-DIFFERENTIATED
Overall, MALT lymphomas account for about 7 NEUROENDOCRINE (CARCINOID) TUMOR
to 8 percent of lymphomas throughout the body This negative gastrin stain is from the gastric type 2
and are less common than diffuse large B cell well differentiated neuroendocrine (ca rcinoid) tumor that
has resulted from the hypergastrinemia produced by the
lymphoma or follicular lymphoma. However, gastrinoma. This tumor is composed of ECL type cells.

Figure 4-23
TYPE 3 WELL-
DIFFERENTIATED
NEUROENDOCRINE
(CARCINOID) TUMOR
This lesion has arisen in
normal gastric body mucosa
in the absence of hypergas-
trinemia. This type tends to
be aggressive.

85
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-24
GASTRIC MUCOSA-ASSOCIATED LYMPHOID TISSUE (MALT) LYMPHOMA
A: The lesion is deep in the mucosa and has destroyed the muscularis mucosae (at left) such that only a few eosinophilic
wisps remain.
B: The infiltrate has destroyed the pits (lymphoepitheliallesions) .
C: Note the monotonous appearance of the lymphoid cells. Many have a halo around them.
D: This cytokeratin stain highlights residual epithelial cells in so called lymphoepithelial lesions.

something that can be assessed at low magni- gastritis). On immunophenotyping, there can
fication. The classic "lymphoepithelial lesion," be CD20-reactive B cells that aberrantly express
in which lymphocytes invade residual gland CD43. A CD3 stain highlights normal admixed
epithelium, is not present in every case. Finding T cells. Also, if one has mistaken the intraepi-
an expansion of B cells in the deep mucosa ac- thelial T cells that can accompany celiac disease
companied by lymphoepithelial lesions allows (lymphocytic gastritis pattern), learning that the
a confident interpretation. Numerous plasma intraepithelial cells are T cells rather than B al-
cells often indicate a reactive lesion (Helicobacter lows for consideration of alternate interpretations.

86
Gastric Polyps and Neoplasms

Figure 4-25
LARGE B-CELL LYMPHOMA IN GASTRIC MUCOSA
A: The infiltrate is monotonous and highly cellular.
B: Such tumors often feature prominent apoptosis.
C: This is a CD20 immunostain.

MALT lymphomas have the following profile: lesions aberrantly eo-express CDS. CD10 can be
CD20(+), CD79a (+), BCL2(+), CDS(-), CDlO(-), added since follicular lymphoma expresses CD10
CD23(-), CD43(+/-). The cells often express IgM; and MALT lymphoma does not. Cyclin D1 (BCLl)
immunoglobulin light chain restriction may be can be added, which labels the nuclei in mantle
encountered in cells having plasmacytoid differ- cell lymphoma but not MALT lymphoma.
entiation (26,30). The pitfall with plasmacytoid Treatment involves eradication of H. pylori ini-
MALT is that it can lack CD20 expression. tially and a few patients require other treatments.
Only half of MALT lymphomas coexpress For gastric lesions, radiation therapy is the most
CD20 and CD43, but the diagnosis can still common used. Many chemotherapy regimens
be made on a B-cell rich infiltrate with typical have been used but rarely as first line treatments.
morphology. It is important to document the These are summarized by Raderer et al. (31).
presence (or absence) of Helicobacter. Most high grade gastric lymphomas are diffuse
It is sometimes worthwhile to add CDS testing large B-celllymphomas (DLBCL) (fig. 4-25), some
to exclude chronic lymphocytic leukemia/small presumably tr·ansforming from MALT lymphomas.
lymphocytic lymphoma (CLL/SLL) since the B cell The 2016 classification has separated DLBCL into

87
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-26
INFLAMMATORY FIBROID POLYP
Left: The tumor is centered in the antral submucosa and features scattered lymphoid aggregates.
Right: This is a CD34 stain.

germinal center B (GCB) cell type and activated gastric gastrointestinal stromal tumors (GISTs).
B cell (ABC) type as follows using immuno- In contrast to GISTs, inflammatory fibroid pol-
labeling although many oncologists prefer to yps are always benign.
see results of gene expression profiling in order Inflammatory fibroid polyps arise in the sub-
to best manage patients (32). The reasoning is mucosa and are well-marginated, but nonencap-
that so-called GCB DLBCL is associated with a sulated. They are composed of uniform spindled
better outcome than ABC type DLBCL. The ABC cells, mixed inflammatory cells, and prominent
type requires more aggressive treatment: GCB = vasculature (figs. 4-26, 4-27). The spindle cells
CD10+ OR CDlO-, BCL6+, and MUMl-; and ABC have amphophilic elongate cytoplasm and pale
= CDlO-, BCL6-, OR CDlO-, BCL6+ and MUMl +. nuclei, ranging from ovoid to spindle shape,
that display variable collagen deposition. Most
INFLAMMATORY FIBROID POLYP examples display a whorled, "onion skin" pro-
Most of these tumors occur in the stomach, liferation around vessels, and all examples have
where they account for up to 3 percent to 4 variable numbers of background eosinophils,
percent of all gastric polyps (7,33), but they lymphocytes, and plasma cells. Mitoses are in-
can arise throughout the gastrointestinal tract. frequent. These polyps express CD34 but lack
Patients with gastric examples were typically CD117 and DOGl expression.
60 to 80 years old, These polyps are quite rare
in children (34) . Most are antral. Presentation GASTROINTESTINAL STROMAL
is somewhat site-specific-small intestinal ex- TUMORS (GISTS) OF THE STOMACH
amples often lead to intussusception (35) or The stomach is the most common site for
obstruction, and gastric examples are found in gastrointestinal stromal tumors (GISTs) and they
patients with pain, nausea, and vomiting. These are occasionally diagnosed on mucosal biopsies
polyps were considered reactive in the past but (fig. 4-28).
they harbor mutations in the platelet-derived Overall, GISTs are the most common mesen-
growth factor receptor alpha (PDGFRA) gene chymal lesions throughout the entire GI tract
(36), a feature that they share with a subset of with two exceptions: leiomyomas outnumber

88
Gastric Polyps and Neoplasms

Figure 4-27
INFLAMMATORY FIBROID POLYP
Left: This example happens to be associated with gastric intestinal type low grade dysplasia, which resembles a colorectal
adenoma, a very unusual finding in Western patients. However, note that the inflammatory fibroid polyp whorls around
the two superficial submucosal vessels in an onion skin fashion.
Right: The tumor cells are quite bland appearing and associated with eosinophils.

GISTs in both the esophagus and the colon (37). predominance for malignant GISTs. GISTs are
The majority harbor either a KIT (CD117, a encountered throughout the gastrointestinal
receptor tyrosine kinase) or a PDGFRA (platelet tract: about 60 percent in the stomach, 35
derived growth factor alpha) mutation. PDG- percent in the small intestine, and less than 5
FRA mutated GISTs are nearly always gastric percent in the rectum, esophagus, omentum,
(37). Some GISTs lack both of these mutations, and mesentery; most GISTs found in the last
especially those found in children and asso- site are metastatic rather than primary. About
ciated with various genetic syndromes and five percent of GISTs are found in patients
harbor different mutations that affect down- with neurofibromatosis type 1 (multiple small
stream signaling. With the exception of those intestinal tumors (38) and Carney triad (para-
found in patients with neurofibromatosis type ganglioma, GIST, and pulmonary chondroma,
1 (NF1), GISTs that lack KIT and PDGFRA mu- usually in young females (39). Familial GISTs
tations nearly always affect the stomach. The arise in patients with inherited germline KIT or
histologic appearance of the cells comprising PDGFRA mutations.
GISTs varies from spindled to epithelioid. GISTs Most recently, succinate dehydrogenase defi-
(including all sites) usually arise in adults over cient GISTs have been described (40-54). These
age 50 (median age, 55 to 60 years) and are rare tumors are restricted to the stomach and do
in children (less than 1 percent). There is no not conform to the typical risk stratification of
gender predilection overall, but there is a male non-SDH deficient GISTs.

89
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 4-28

GASTROINTESTINAL STROMAL TUMOR (GIST)


A: These tumors nearly invariably arise in the muscularis propria so are seldom detected on mucosal biopsies, but there
are exceptions, as in this case.
B: This example extended into the submucosa, entrapping ganglion cells in the center right of the field.
C: This is a KIT stain from the neoplasm depicted in A and B.
D: This example features stunning paranuclear vacuoles. Note that the tumor cells have very uniform nuclei. When gastrointestinal
lTact spindle cell tumors have nuclear pleomorphism, it is important to consider alternative diagnoses to GIST.

90
Gastric Polyps and Neoplasms

About 20 percent of gastric GISTs are malig- Epithelioid GISTs are most likely to cause
nant. The consensus is that 5 mitoses per 50 diagnostic problems on mucosal biopsies,
high-power fields (hpf) and >5 cm are adverse because they are readily mistaken for a host
prognostic factors (55,56). With current target- of epithelioid and epithelial neoplasms. It is
ed therapy using imatinib as a first line drug advisable to perform an immunohistochemi-
and sunitinib or regorafinib as second and third cal panel in assessing them to include CD 117 I
line drugs, outcome has improved (57). In the KIT, S-100 protein (to address melanoma) and
past, the 5 year survival for malignant GISTs a cytokeratin stain to address signet cell carci-
was on the order of 50 percent, whereas now it noma. Remember that up to a third of gastric
is <;loser to 75 percent. Unfortunately about 80 carcinomas express DOG 1 (58) and melanomas
percent of these tumors develop resistance to are likely to express CD 117.
the available drugs over time.

REFERENCES

1. Park ]Y, Cornish TC, Lam-Himlin D, Shi C, clinicopathologic group including a distinct sub-
Montgomery E. Gastric lesions in patients with set best categorized as mucosal prolapse polyp.
autoimmune metaplastic atrophic gastritis Am] Surg Pathol 2011;35:670-7.
(AMAG) in a tertiary care setting. Am ] Surg 10. Melton SD, Genta RM. Gastric cardiac polyps: a
Pathol 2010;34:1591-8. clinicopathologic study of 330 cases. Am] Surg
2. Carmack SW, Genta RM, Schuler CM, Saboorian Pathol2010;34:1792-7.
MH. The current spectrum of gastric polyps: a 11. Pittman ME, Voltaggio L, Bhaijee F, Robertson
1-year national study of over 120,000 patients. SA, Montgomery EA. Autoimmune metaplastic
Am] Gastroenterol 2009;104:1524-32. atrophic gastritis: recognizing precursor lesions
3. Li J, Woods SL, Healey S, et al. Point mutations for appropriate patient evaluation. Am ] Surg
in Exon 1B of APC reveal gastric adenocarcinoma Pathol2015;39:1611 -20.
and proximal polyposis of the stomach as a 12. Lam-Himlin D, Park ]Y, Cornish TC, Shi C,
familial adenomatous polyposis variant. Am ] Montgomery E. Morphologic characterization
Hum Genet 2016;98:830-42. of syndromic gastric polyps. Am ] Surg Pathol
4. Choudhry U, Boyce HW Jr, Coppola D. Proton 2010;34:1656-62.
pump inhibitor-associated gastric polyps: a 13. Ma C, Giardiello FM, Montgomery EA. Upper
retrospective analysis of their frequency, and tract juvenile polyps in juvenile polyposis pa-
endoscopic, histologic, and ultrastructural char- tients: dysplasia and malignancy are associated
acteristics. Am] Clin Pathol1998;110:615-21. with foveolar, intestinal, and pyloric differentia-
5. Vieth M, Stolte M. Fundic gland polyps are not tion. Am] Surg Pathol 2014;38:1618-26.
induced by proton pump inhibitor therapy. Am 14. Abraham SC, Montgomery EA, Singh VI<, Yardley
] Clin Pathol 2001;116:716-20. ]H, Wu TT. Gastric adenomas: intestinal-type and
6. Zelter A, Fernandez JL, Bilder C, et al. Fundic gastric-type adenomas differ in the risk of adeno-
gland polyps and association with proton pump carcinoma and presence of background mucosal
inhibitor intake: a prospective study in 1,780 pathology. Am] Surg Pathol2002;26:1276-85 .
endoscopies. Dig Dis Sci 2011;56:1743-8. 15. Park DY, Srivastava A, Kim GH, et al. Adenoma to us
7. Stolte M, Sticht T, Eidt S, Ebert D, Finkenzeller and foveolar gastric dysplasia: distinct patterns
G. Frequency, location, and age and sex distribu- of mucin expression and background intestinal
tion of various types of gastric polyp. Endoscopy metaplasia. Am] Surg Pathol2008;32:524-33.
1994;26:659-65. 16. Chen ZM, Scudiere]R, Abraham SC, Montgom-
8. Abraham SC, Singh VK, Yardley ]H, Wu TT. Hy- ery E. Pyloric gland adenoma: an entity distinct
perplastic polyps of the stomach: associations from gastric foveolar type adenoma. Am ] Surg
with histologic patterns of gastritis and gastric Pathol 2009;33: 186-93.
atrophy. Am] Surg Pathol2001;25:500-7. 17. Vieth M, Kushima R, Borchard F, Stolte M. Pyloric
9. Gonzalez-Obeso E, Fujita H, Deshpande V, et gland adenoma: a clinico-pathological analysis
al. Gastric hyperplastic polyps: a heterogeneous of 90 cases. Virchows Arch 2003;442:317-21.

91
Survival Guide to Gastrointestinal Mucosal Biopsies

18. Vieth M, Montgomery EA. Some observations on 33. Stolte M, Finkenzeller G. Inflammatory fibroid
pyloric gland adenoma: an uncommon and long polyp of the stomach. Endoscopy 1990;22:203-7.
ignored entity!] Clin Pathol 2014;67:883-90. · 34. Righetti L, Parolini F, Cengia P, et al. Inflam-
19. Genta RM, SchulerCM, Robiou Cl, Lash RH. No matory fibroid polyps in children: A new case
association between gastric fundic gland polyps report and a systematic review of the pediatric
and gastrointestinal neoplasia in a study of over literature. World] Clin Pediatr 2015;4: 160-6.
100,000 patients. Clin Gastroenterol Hepatol 35 . Liu TC, Lin MT, Montgomery EA, Singhi AD.
2009;7:849-54. Inflammatory fibroid polyps of the gastrointes-
20. Siegel RL, Miller KD, ]emal A. Cancer statistics, tinal tract: spectrum of clinical, morphologic,
2016. CA Cancer] Clin 2016;66:7-30. and immunohistochemistry features. Am] Surg
21. Parkin DM, Bray F, Ferlay ], Pisani P. Global cancer Pathol 2013;3 7:586-92.
statistics, 2002. CA Cancer] Clin 2005;55:74-108. 36. Schildhaus HU, Cavlar T, Binot E, Buttner R,
22. Boncher ], Bronner M, Goldblum ]R, Liu X. Re- Wardelmann E, Merkelbach-Bruse S. Inflamma-
ticulin staining clarifies florid benign signet ring tory fibroid polyps harbour mutations in the
cell change with mitotic activity in a penetrating platelet-derived growth factor receptor alpha
gastric ulcer. Am] Surg Pathol 2011;35:762-6. (PDGFRA) gene.] Pathol2008;216:176-82.
23. Cameselle-Teijeiro ], Abdulkader I, Forteza ]. 37. Miettinen M, Lasota ]. Gastrointestinal stro-
Signet-ring cell change in pseudomembranous mal tumors. Gastroenterol Clin North Am
colitis versus signet-ring cell carcinoma. Am ] 2013;42:399-415.
Surg Pathol 2004;28:1111. 38. Miettinen M, Lasota]. Gastrointestinal stromal
24. Dimet S, Lazure T, Bedossa P. Signet-ring cell tumors: review on morphology, molecular pa-
change in acute erosive gastropathy. Am ] Surg thology, prognosis, and differential diagnosis.
Pathol 2004;28: 1111-2. Arch Pathol Lab Med 2006;130:1466-78.
25. Wang K, Weinrach D, Lal A, et al. Signet-ring cell 39. Carney ]A, Sheps SG, Go VL, Gm·don H. The
change versus signet-ring cell carcinoma: a com- triad of gastric leiomyosarcoma, functioning
parative analysis. Am] Surg Pathol2003;27:1429-33. extra-adrenal paraganglioma and pulmonary
26. Bosman F, Carneiro F, Hruban· R, et al. WHO chondroma. N Engl] Med 1977;296:1517-8.
Classification of Tumours of the Digestive Sys- 40. Belinsky MG, Rink L, van Mehren M. Succinate
tem. In: Bosman F, Jaffee E, Lakhani S, Ohgaki dehydrogenase deficiency in pediatric and adult
H, eds. World Health Organization Classification gastrointestinal stromal tumors . Front Oncol
of Tumours. Lyon: IARC Press; 2010. 2013;3:117.
27. Modlin IM, Lye KD, Kidd M. A 50-year analysis 41. Bolland M, Benn D, Croxson M, et al. Gastroin-
of 562 gastric carcinoids: small tumor or larger testinal stromal tumour in succinate dehydro-
problem? Am] Gastroenterol 2004;99:23-32. genase subunit B mutation-associated familial
28. Washington MK, Tang LH, Berlin ], et al. Pro- phaeochromocytoma/paraganglioma. ANZJ Surg
tocol for the examination of specimens from 2006;76:763-4.
patients with neuroendocrine tumors (carcinoid 42. Celestino R, Lima J, Faustino A, et al. Molecular
tumors) of the stomach. Arch Pathol Lab Med alterations and expression of succinate dehydro-
2010;134:187-91. genase complex in wild-type KIT/PDGFRA/BRAF
29. Chen WC, Warner RR, Ward SC, et al. Manage- gastrointestinal stromal tumors. Eur ] Hum
ment and disease outcome of type I gastric neu- Genet 2013;21:503-10.
roendocrine tumors: the Mount Sinai experience. 43 . Doyle LA, Hornick JL. Gastrointestinal stromal
Dig Dis Sci 2015;60:996-1003. tumours: from KIT to succinate dehydrogenase.
30. Swerdlow S, Campo E, Harris NL, et al. WHO Histopathology 2014;64:53-67.
Classification of Tumours of Haematopoietic and 44. Doyle LA, Nelson D, Heinrich MC, Carless CL,
lymphoid tissues. In: Bosman F,] affee E, Lakhani HornickJL. Loss of succinate dehydrogenase sub-
S, Ohgaki H, eds. World Health Organization Clas- unit B (SDHB) expression is limited to a distinc-
sification of Tumours. Lyon: IACR Press; 2008. tive subset of gastric wild-type gastrointestinal
31. Raderer M, Kiesewetter B, Ferreri A] . Clinico- stromal tumours: a comprehensive genotype-
pathologic characteristics and treatment of phenotype correlation study. Histopathology
marginal zone lymphoma of mucosa-associated 2012;61:801-9.
lymphoid tissue (MALT lymphoma). CA Cancer 45 . Dwight T, Benn DE, Clarkson A, et al. Loss of
] Clin 2016;66:153-71. SDHA expression identifies SDHA mutations
32. Swerdlow SH, Campo E, Pileri SA, et al. The in succinate dehydrogenase-deficient gastro-
2016 revision of the World Health Organization intestinal stromal tumors. Am ] Surg Pathol
classification of lymphoid neoplasms. Blood 2013;37:226-33.
2016;127:2375-90.

92
Gastric Polyps and Neoplasms

46. Janeway KA, Kim SY, Lodish M, et al. Defects in succinate dehydrogenase complex dysfunction.
succinate dehydrogenase in gastrointestinal stro- Future Oncol 2013;9:121-6.
mal tumors lacking KIT and PDGFRA mutations. 53. Pantaleo MA, Nannini M, Astolfi A, Biasco G;
Proc Natl Acad Sci US A 2011;108:314-8. GIST Study Group Bologna . A distinct pediatric-
47. Jove M, Mora], Sanjuan X, et al. Simultaneous type gastrointestinal stromal tumor in adults: po-
KIT mutation and succinate dehydrogenase tential role of succinate dehydrogenase subunit
(SDH) deficiency in a patient with a gastrointes- A mutations. Am J Surg Pathol 2011 ;35:1750-2.
tinal stromal tumour and Carney-Stratakis syn- 54. Wang JH, Lasota ], Miettinen M. Succinate De-
drome: a case report. Histopathology 2014;65: hydrogenase subunit B (SDHB) is expressed in
712-7. Neurofibromatosis 1-associated gastrointestinal
48. Killian JK, Kim SY, Miettinen M, et al. Succinate stromal tumors (GISTs): implications for the
dehydrogenase mutation underlies global epig- SDHB expression based classification of GISTs. J
enomic divergence in gastrointestinal stromal Cancer 2011;2:90-3 .
tumor. Cancer Discov 2013;3:648-57. 55. Miettinen M, Lasota]. Gastrointestinal stromal
49. Mason EF, Hornick JL. Succinate dehydroge- tumors-definition, clinical, histological, im-
nase deficiency is associated with decreased munohistochemical, and molecular genetic
5-hydroxymethylcytosine production in gas- features and differential diagnosis . Virchows
trointestinal stromal tumors: implications for Arch 2001;438:1-12.
mechanisms of tumorigenesis. Mod Pathol 56. Miettinen M, Sobin LH, Lasota]. Gastrointestinal
2013;26:1492-7. stromal tumors of the stomach: a clinicopatho-
50. Miettinen M, Killian JK, Wang ZF, et al. Immu- logic, immunohistochemical, and molecular
nohistochemicalloss of succinate dehydrogenase genetic study of 17 65 cases with long-term fol-
subunit A (SDHA) in gastrointestinal stromal low-up. Am J Surg Pathol 2005;29:52-68.
tumors (GISTs) signals SDHA germline mutation. 57. Jakhetiya A, Garg PK, Prakash G, Sharma], Pan-
Am J Surg Pathol 2013;37:234-40. dey R, Pandey D. Targeted therapy of gastroin-
51. Miettinen M, Lasota J. Succinate dehydrogenase testinal stromal tumours. World J Gastrointest
deficient gastrointestinal stromal tumors (GISTs) Surg 2016;8:345-52.
-a review. IntJ Biochem Cell Biol2014;53:514-9. 58. Miettinen M, Wang ZF, LasotaJ. DOGl antibody
52. Nannini M, Astolfi A, Paterini P, et al. Expression in the differential diagnosis of gastrointestinal
of IGF-1 receptor in KIT/PDGF receptor-alpha stromal tumors: a study of 1840 cases. Am J Surg
wild-type gastrointestinal stromal tumors with Pathol 2009;33 :1401-8.

93
NON-NEOPlASTIC
5 SMAll INTESTINE

NORMAl AND REACTIVE CONDITIONS record, particularly reconciliation with the pa-
·When reviewing small bowel samples without tient's medication list (Table 5-1).
neoplasms, the main questions revolve around Normal small intestinal mucosa should have
celiac disease and other entities that result in long slender villi that protrude from a base in
damage to the mucosa in patients who present which there are crypts. The crypts at the bottom
with diarrhea. The key possibilities to consider of the sample are nearly identical to the ones
are celiac disease, medication-associated injury, in the colon. If they are embedded perfectly,
infections and inflammatory bowel disease. they are aligned like test tubes in a rack (fig.
However, many biopsies come from patients 5-1) but in daily practice they seldom are and
who do not have malabsorption symptoms and often instead appear as lots of glands that are
have undergone upper endoscopic biopsies for organized in an orderly fashion akin to the test
another indication; the small intestinal samples tubes viewed from above (fig. 5-2). Frequently
are "along for the ride." Most such samples are samples are imperfectly embedded, impa1ting an
normal but they have artifacts that can cause appearance of villous blunting. In general, if any
concerns. Lastly, the small bowel only has a well-formed villi are present, in all likelihood,
limited set of responses to a host of injuries so villi that appear attenuated are probably simply
interpretation of small bowel pathology often embedded sub-optimally. Figures 5-3-5-5 show
requires attention to the patient's medical normal duodenum. Important feature to note:

Table 5-l
SMALL INTESTINE PATTERNS OF INJURY
Feature Associations
Intraepithelial lymphocytosis Celiac disease
Infections (small bowel bacterial overgrowth, giardiasis, "tropical sprue,"
cytomegalovirus)
Medications ("sartans" such as olmesartan and losartan (angiotensen Il recep-
tor antagonists), anti-CTLA4 drugs such as ipilimumab, PDL-1 blockade
drugs such as pembrolizumab and nivolumab, proton pump inhibitors)
Crohn disease
Prominent crypt apoptosis; normal is Medications ("sartans" such as olmesartan and losartan, mycophenolate, anti-
1-2 apoptotic bodies per pinch biopsy CTLA4 drugs such as ipilimumab, PDL-1 blockade drugs such as pembro-
lizumab and nivolumab)
Infe ctions (consider cytomegalovirus, cryptosporidium)
Graft versus host disease
Autoimmune enteropathy
Absent or markedly reduced lamin a Common variable immunodeficiency
propria plasma cells
Absent or markedly reduced goblet cells Autoimmtme enteropathy
and/or Paneth cells
Erosions and Ulcers Nonsteroidal anti-inflammatory drugs
Infections (consider cytomegalovirus)
Crohn disease

95
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure S-1 Figure S-2


TEST TUBES IN A RACK TEST TUBES IN A RACK
While this applies most to colon rather than small This is as seen from above, much like mucosa embedded
intestinal samples, the crypts of the intestine can be en face. Note the round structures (the test tubes or crypts)
considered as analogous to text tubes with the muscularis are evenly spaced.
mucosae akin to the bottom of the rack upon which the
tubes rest. The crypts are separated by uniform amounts of
space {lamina propria) and are aligned in an orderly fashion.

Figure S-3
NORMAL SMALL INTESTINAL MUCOSA
Left: Long slender villi protrude into the lumen at the right of the image. The lamina propria contains abundant plasma
cells. Also, note that there are a few lymphocytes in the epithelium but not many.
Right: This is a high magnification of the bases of the crypts. Each crypt contains several Paneth cells, which contain
plump eosinophilic granules that are aimed towards the lumen of the crypt. The lamina propria (the loose connective tissue
between the glands/crypts) contains lymphocytes, plasma cells, and a few eosinophils.

96
Non-Neoplastic Small Intestine

Figure 5-4
NORMAL SMALL INTESTINAL MUCOSA
Left: This is the surface. There are a few lymphocytes in the surface epithelium. The lamina propria beneath the surface
contains many plasma cells, a few lymphocytes, and a few eosinophils.
Right: In this tangentially embedded sample the villi are sliced both longitudinally and in cross section. Each contains
delicate capillaries, scattered lymphocytes and plasma cells and strands of smooth muscle.

1) Nearly every crypt has lots of Paneth cells. 5) The ileum often appears damaged at scan-
This differs from the colon and finding lots of ning magnification because there are prominent
Paneth cells in crypt bases can be a clue that lymphoid aggregates (Peyer patches). Histiocytes
a damaged sample is from the small intestine in the lymphoid tissue in the ileum often contain
rather than the colon (fig. 5-3, right) . black material that corresponds to titanium in
2) There are delicate strands of smooth toothpaste and various foods (figs. 5-8, 5-9).
muscle in the core of each villus. This smooth There is an important pitfall in interpreting
muscle helps the villi beat to increase nutrient duodenal biopsies. Well differentiated neuro-
absorption. It can be mistaken for other things endocrine (carcinoid) tumors of the duodenum
(Whipple disease, Gaucher's disease) in tangen- are easily crushed and are very vascular. They
tially embedded villi (fig. 5-5). tend to have an appearance similar to that of
3) Some duodenal samples feature crushed lymphoid aggregates on crushed samples. Such
Brunner glands that can be mistaken for mesen- a case is seen in figure 5-10.
chymal neoplasms and Whipple disease (fig. 5-6). When the stomach or esophagus is damaged,
Brunner glands are strongly periodic acid-Schiff the mucosa tends to be replaced by intestinal
(PAS)-reactive, which can add to the confusion. type mucosa as seen in chapters 2, 3 and 4. In
4) Crushed samples also lose their epitheli- a similar vein, when the small intestine is dam-
um, akin to squeezing sebum from the pores of aged, it is replaced by mucosa with gastric fea-
nasal skin (fig. 5-7). This should not be mistaken tures, either in the form of glands that resemble
for ischemia, which will be illustrated later. gastric foveolar cells with surface neutral mucin

97
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure S-5
NORMAL SMALL INTESTINAL MUCOSA

Left: Note the delicate strands of smooth muscle in the lamina propria. The surface shows the brush border with microvilli
and a few goblet cells. The cells between the goblet cells are absorptive cells.
Right: This is a villus cut in cross section. Note the eosinophilic smooth muscle cells cut in cross section.

Figure 5-6
NORMAL SMALL INTESTINAL MUCOSA
Left: A few well preserved Brunner glands are present in the lower part of the center of the image. Their mucin globules
are multivacuolated. Some have become crushed and extruded into and amongst the crypts in the field. This is a common
artifact in duodenal samples.
Right: Note the crushed Brunner gland in the center of the image.

98
Non-Neoplastic Small Intestine

Figure S-7
NORMAl SMAll
INTESTINAl MUCOSA
This sample was crushed and
the crypts have been squeezed
out. There is no response to the
missing epithelium, supporting
an interpretation of an artifactual
loss of glands/crypts.

Figure S-8
NORMAl SMAll INTESTINAl MUCOSA
Left: This is an ileal sample. Abundant lymphoid tissue is a normal finding in the ileum. The black pigment is not strictly
normal but commonly detected. It is titanium from toothpaste and food.
Right: This is a high magnification of the sample seen on the left. The titanium is found in macrophages.

99
Survival Guide to Gastroin.testinal Mucosal Biopsies

Cholesterol Less than 300mg 300mg


Sodium Less than 2,400mg 2,400mg
Total Carbohydrate 300g 375g
Dietary Fiber 25g 30g
INGREDIENTS:
CORN SYRUP; SWEETENED CONDENSED MILK (MILK;
Figure S-9 SUGAR); HIGH FRUCTOSE CORN SYRUP; PALM
LABEL FROM A CANDY BAR KERNEL OIL; SUGAR; GLYCERIN; DAIRYBUTIER(MILK);
CONTAINS 2% OR LESS OF: GLYCERYL MONOSTEA-
Note that titanium dioxide is listed as an ingredient.
RATE; SALT; TITANIUM DIOXIDE (COLOR); SOY
LECITHIN; NATURAL FLAVORS; DISODIUM
PHOSPHATE; TOCOPHEROLS, TO MAINTAIN
FRESHNESS; SODIUM CARBONATE; MINERAL OIL. @o

Dist. by The Hershey Company


Hershey, PA 17033-08 15, U. S.A.
Mfd. in Canada

Figure 5-10
WELL-DIFFERENTIATED NEUROENDOCRINE
(CARCINOID) TUMOR, DUODENAL BIOPSY
It is important to consider this possibility in samples
that appear cmshed and inflamed at low magnification.
B: This is a high magnification of the lesion depicted in
A. There are crushed lymphocytes at the lower right but the
lesion has a nested appearance in the periphery of the field.
C: This is a chromogranin stain of the lesion seen in A.

~ c

100
Non-Neoplastic Small Intestine

Figure 5-11
DUODENITIS WITH GASTRIC
ML!CIN CELL METAPLASIA
A: This .pattern is often seen in the
presence of damage from gastric acid and
was initially recognized in the setting of
H. pylori gastritis. Some have used the term
"peptic duodenitis." However, any source of
chronic damage can produce gastric mucin
cell metaplasia, including Crohn disease
and nonsteroidal anti-inflammatory drug
use. Note the reactive appearance of the
surface at the right. This appearance can
be concerning for duodenal adenoma.
B: Some ·of the surface glands have an
apical cap of mucin that is of the gastric
foveolar type.
C: This is a high magnification showing
metaplastic gastric foveolar type mucin.

or glands that resemble cardiac or pyloric type history of hemodyalisis, hypertension, diabetes
glands. Knowing about this is helpful because mellitus, coronary artery disease, and/or iron
reactive changes in the duodenum can resemble deficiency anemia (1). The endoscopist may
adenomas. If the surface of such areas is coated note gray-black pigmentation. The pigment is
with gastric foveolar cells (fig. 5-11), this usually ingested in lysosomes and composed of varying
indicates a reactive process. Sometimes these amounts of iron, sulfur, and calcium. Histologic
reactive changes result in a nodular appearance. examination shows macrophages containing
The term "nodular duodenitis" can be applied granular black or brown pigment, which may
in this instance. A less common finding is resemble melanin but is not. There is variable
melanosis duodeni (fig. 5-12). It is also termed staining with trichrome and iron stains. The
"pseudomelanosis" and consists of accumula- material is darker, larger, and denser than hemo-
tion of pigmented macrophages in the lamina siderin. Melanosis duodeni is usually regarded
propria of the small bowel. Patients may have a as an incidental, benign finding.

101
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 5-12
MELANOSIS DUODENI
Left: The pigment is not melanin so the term "pseudomelanosis duodeni" is probably more accurate. Note that the
sample is otherwise normal with slender villi and nicely aligned crypts with an arrangement similar to that of the test tubes
seen figure 5-1.
Right: High magnification from the case seen on the left.

INFECTIOUS AGENTS present with loose stools, flatulence, malab-


Various organisms prone to infect the small sorption, weight loss, and in children, impaired
intestine include cytomegalovirus (fig. 5-13), growth and cognitive development resulting
Giardia lamblia (fig. 5-14), Strongyloides stercoralis from iron and micronutrient deficiencies (2,3).
(figs. 5-15, 5-16), Mycobacterium avium (figs. 5-17, On hematoxylin and eosin (H&E), trophozoites
5-18), and Trophyrema whipplei (fig. 5-19). appear as pear-shaped structures. They have
Giardiasis is the most commonly encountered two ovoid nuclei and four pairs of flagella, but
parasitic infection in the United States. Giardia these structures are often difficult to identify on
lamblia cysts are transmitted by the fecal-oral H&E stain. The background duodenal mucosa
route, sometimes in contaminated food or wa- may be normal but more often shows acute
ter. Cattle and sheep excrete abundant Giardia inflammation, villous attenuation, increased in-
cysts, which can contaminate water sources traepitheliallymphocytes, lymphoid aggregates,
and can survive for months. Once the cysts are and/or gastric mucin cell metaplasia. Some
swallowed, gastric acid and intestinal proteases biopsies may lack organisms but still manifest
allow excystation into the trophozoite. the inflammatory changes.
Giardia infection may be asymptomatic or Strongyloides stercoralis is a nematodal infec-
result in acute symptoms including diarrhea, tion endemic to Africa, Southeast Asia, South
steatorrhea, flatulence, nausea and vomiting, America, and parts of the United States. In its
and fever. Chronic infection occurs in approx- lifecycle, the female worm lays her eggs without
imately half of infected individuals and may fertilization (parthenogenesis) in small intestinal

102
Non-Neoplastic Small Intestine

Figure 5-13

t • ~~-·~ ~. •-:- I
..
I~•
CYTOMEGALOVIRUS ENTERITIS
A: This case has injury due to more than one thing.

I
'· .' .
..... .
;,

This patient had undergone a bone marrow transplant and
probably has graft versus host disease since most of the crypts
are gone and there was ample apoptotic injury. However,
. . ... . tl , apoptotic injury can also reflect cytomegalovirus enteritis.
·•
'· '
·~ ,;•
. '.. J.
'7' .
•·• . . . •
I It
, • o
e•
• .· w• B: The residuum of a few glands is apparent but a small
·' ~ . : .... \.r· , · ·.''· capillary at the lower right contains an endothelial cell with
a cytoplasmic inclusion. This is not diagnostic in isolation
' .~
but prompted obtaining cytomegalovirus immunolabeling.
C: This is an immunostain from the lesion depicted
in A and B. The patient probably also has graft versus
host disease, but the cytomegalovirus infection must be
addressed first .
•.
. ...
.
..' r ,. ' , . ·.. · ··,~:

...
\ b . "' ~· ·~ . f , ,
\
: .. i ' - . ~~; ;t, \ '
•• • ~ ,. a
I
•·
,'
.
crypts . Eggs hatch into rhabditifonn larvae, through blood vessels to the lungs and into the
which pass into the lumen and are expelled with upper airways. This results in coughing and the
feces. The passed rhabditiform larvae develop larvae are swallowed and reach the small intes-
into filariform larvae, which penetrate human tine to mature into adults. Males are rapidly
skin (often through bare feet) and penetrate expelled after fertilizing the females whereas

103
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 5-14
GIARDIASIS
Left: In this case, there is minimal tissue response, making it easy to overlook the organisms, which are present as debris
separate from the tissue itself.
Right: This is a high magnification from the sample seen on the left.

Figure 5-15
STRONGYLOIDIASIS
Left: The villi are shortened and the lamina propria is expanded with increased inflammation. The organisms are seen
in the crypts.
Right: In addition to organisms, there is abundant acute inflammation in both the lamina propria and the crypts.

104
Non-Neoplastic Small Intestine

Figure 5-16
STRONGYLOIDIASIS
Note the inflammatory backdrop.

Figure 5-17
MYCOBACTERIUM AVIUM
COMPLEX ENTERITIS
A: The lamina propria is stuffed with pale
foamy macrophages in solid sh eets.
B: Each lamina propria macrop h age
contains ample fluffy pink cytoplasm.
C: This is a periodic acid-Schiff (PAS)
stain. It highlights the goblet cells, the brush
borders of the enterocyte surface and the waxy
mycobacterial organisms, which appear as
delicate slender rods.

105
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 5-18
MYCOBACTERIUM AVIUM COMPLEX ENTERITIS
Left: This PAS stain shows the delicate rods within the histiocytes to advantage. Note the brush border and goblet cells
on the luminal surface.
Right: This is an acid fast stain.

the female worms can remain in the intestine to Mycobacterium tuberculosis, well-defined gran-
for decades. If the patient becomes immuno- ulomas are not seen.
suppressed (such as during chemotherapy for Whipple disease is the differential diagnosis
a malignancy later in life), rhabditiform larvae for Mycobacterium avium infection. It is a rare
undergo a transformation into the penetrating systemic bacterial infection caused by Trophe1Y'na
filariform larvae, penetrate intestinal mucosa, whippleii (an actinomycete). The small intestine is
and disseminate throughout the body, which the organ most commonly affected but infection
can sometimes prove lethal. Biopsies appear can also involve the brain, endocardium, and
damaged with ulceration and granulation tissue. other sites where it can mimic a neoplasm (5).
The organisms are found in mucosal crypts or White males between the fourth and fifth decade
can invoke an eosinophilic response. are most commonly affected. Patients present
Mycobacterium avium complex infection is with protean symptoms that include diarrhea,
a significant cause of opportunistic infection low-grade fever, weight loss, malabsorption, ab-
in immunocompromised patients, notable in dominal pain, chronic intermittent seronegative
patients with acquired immunodeficiency syn- polyarthritis (6), anemia,lymphadenopathy (50
drome (AIDS). Disseminated GI tract infection percent), cardiac conditions, nausea, vomiting,
with Mycobacterium avium is common in AIDS GI bleeding, and central nervous system symp-
patients. Mycobacterium avium usually affects toms (10 percent). Because of the unusual pre-
both the small and large bowels. Endoscopically, sentations, WD is challenging to recognize (7).
the small bowel mucosa either appears normal Small bowel samples show blunted and round-
or shows white nodules/plaques or a coarse ed villi, with distention of lamina propria by
granularity (4) without significant exudate foamy, pink macrophages. The villi show cystic
(implying an attenuated local immunologic spaces that presumably correspond to dilated ob-
response). The granular mucosa corresponds to structed lacteals, a feature that allows distinction
infiltration of small intestinal villi by histiocytes from Mycobacterium avium infection on the H&E
loaded with acid-fast bacilli. Unlike the response alone. The macrophages contain PAS-positive,

106
Non-Neoplastic Small Intestine

Figure 5-19
WHIPPLE DISEASE
A: On h ematoxylin and eosin (H&E),
the appearance is sim ilar to that of
Mycobacterium avium complex enteritis .
However, there are cystic dilations that are
presumably dilated lacteals.
B: This is a PAS stain . The chunky
disorganized deposits differ dramatically
from the delicate orga nism s of Myco-
bacterium avium seen in figure 5-18, left.
C: This is an immunohistochemical
stain for Troplmyma whipplei.

diastase-resistant rod-, sickle-, or globular-shaped spectrum of findings and only display prom-
bacterial inclusions. Whipple antigen can be also inent intraepithelial lymphocytes (fig. 5-21).
detected by immunohistochemical staining (8). Unfortunately intraepitheliallymphocytosis is
a nonspecific findings and must be evaluated in
CELIAC DISEASE the setting of the clinical history and together
Celiac disease is often a consideration and with assessment of the presence of crypt apop-
has characteristic but not specific features (fig. tosis and evaluation of the presence of absence
5-20) . However, some examples lack the full of plasma cells, goblet cells, and Paneth cells.

107
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 5-20
CELIAC DISEASE
A: There are no villi in this well-developed
example. The lamina propria is expanded with
plasma cells and lymphocytes and there are
many intraepitheliallymphocytes in the surface
epithelium. There is no need to count them to see
that they are too numerous. Unfortunately this
pattern is not specific and must be interpreted in
the context of laboratory findings.
B: This is the surface epithelium of the lesion
depicted in A. It is easy to see that there are too
many lymphocytes. Note all the plasma cells in
the lamina propria.
C: The crypts have intraepithelial lympho-
cytosis but the surface is the important area
to assess since intraepithelial lymphocytes are
normal in crypts. There is no increase in crypt
apoptosis.

108
Non-Neoplastic Small Intestine

Figure 5-21
CELIAC DISEASE
Left: Some examples show a "latent" pattern with villi that are only slightly attenuated. However, even at low magnification ,
there are too many intraepitheliallymphocytes in the tips of the villi. This pattern is not specific for celiac disease. In this case,
the patient had appropriate serologic confirmation. Requests to "score" th e findings are useless unless a diagnosis of celiac
is already established. When this pattern is seen de novo, about 10 to 20 percent of the cases will prove to be celiac disease.
Right: High magnification of the lesion on the left. Note that the intraepitheliallymphocytosis is "top heavy."

Celiac disease is a relatively common sys- suggest celiac disease serology as patients with
temic autoimmune disorder induced by gluten mild enteropathy and positive serology benefit
proteins found in wheat, barley, and rye. Injury from early dietary treatment (15) . As a rapid and
to the small intestine is the prototypic finding, simple method to assess whether there are too
but manifestations are systemic. Lymphocytic many lymphocytes in the tips of the villi, count
gastritis is a well-known association. The human 20 enterocyte nuclei in the very tip of a villus
leukocyte antigens (HLAs) that confer the high- (10 on one side of the tip, and 10 on the other)
est risk are HLA-DQ2 and HLA-DQS. Prevalence and (on H&E) count the lymphocytes in this
of celiac disease has been reported as high as area. If there are six or more, this is too many.
1.5 percent in Finnish school children (9) and It is not necessary to immunolabel T cells to as-
only slightly less in the general population of sess these biopsies. Scattered neutrophils can be
the United States (10). The incidence very low present. Crypt apoptosis is not a typical feature.
in Southeast Asians, while there are endemic
populations in the Sahara/Middle East with a AUTOIMMUNE ENTEROPATHY AND
high incidence of celiac disease (11,12). COMMON VARIABLE IMMUNODEFICIENCY
The biopsy findings in celiac disease are An important concept in assessing samples
variable (13,14). Early or partly treated disease from patients with dianhea is to assess for com-
shows only intraepitheliallymphocytosis that ponent that are normally present! It is important
is prominent in the tips of the villi. In the to always check for lamina propria plasma cells,
normal small bowel biopsy, the number of in- Paneth cells, and goblet cells. An absence of
traepitheliallymphocytes decreases in the tips lamina propria plasma cells indicates common
of the villi when compared to the bases. The variable immunodeficiency (CVID) and an ab-
finding of prominent "tip heavy" intraepithelial sence of goblet cells and/or Paneth cells suggests
lymphocytosis can prompt the pathologist to autoimmune enteropathy, although the findings

109
Survival Guide to Gastrointestinal Mucosal Biopsies

:~'t . . • f: :.

Figure 5-22
AUTOIMMUNE ENTEROPATHY PATTERN IN A PATIENT WITH COMMON VARIABLE IMMUNODEFICIENCY
A: Something is missing! The sample appears normal at first glance but there are no goblet cells or Paneth cells! Notice
also that the lamina propria has fewer constituents than usual. Always have a mental checklist when reviewing small bowel
samples-be sure that plasma cells, Paneth cells, and goblet cells are present and , lastly, study the debris that is detached
from the mucosa to assess for Ginrdia.
B: High power shows that the lamina propria is devoid of plasma cells. There are also several apoptotic bodies in the crypts.
C: No plasma cells are present. In this case, the patient had minimal IgG formation, but what little that was produced
consisted of auto antibodies.
0: There are no Paneth cells in the bases of the crypts.

in these two entities overlap (fig. 5-22). Unfortu- without villous blunting, and granulomas. Inter-
nately plasma cells are absent in only two thirds estingly, patients with CVID make few antibodies
of patients with CVID (16) but patients' samples but often the ones that they create are autoanti-
may show prominent lymphoid aggregates, bodies so they can present with syndromes that
increased apoptosis, increased IELs with or overlap with autoimmune enteropathy.

110
Non-Neoplastic Small Intestine

Autoimmune enteropathy is rare and char- colchicine (fig. 5-25), kayexalate (fig. 5-26),
acterized by: 1) small intestinal villous atrophy lanthanum carbonate (fig. 5-27), mycopheno-
unresponsive to dietmy resnictions; 2) unrelenting late (fig. 5-28), and ipilimumab. Yttrium can be
diarrhea; and 3) predisposition to autoimmune associated with ischemic enteritis and radiation
disease 17. It was initially believed to affect change (fig. 5-29). The differential diagnosis in
newborns and young children; adult onset this case can include amyloidosis (fig. 5-30).
is well-documented in the literature (18-21). A severe sprue-like enteropathy is associated
Pathophysiology is poorly understood but a pro- with olmesartan and other anti-hypertensives
posed mechanism involves loss of self-tolerance in this class (angiotensin II receptor antagonists)
and a hyperreactive immune system with inap- (26). Patients present with chronic diarrhea and
propriate T-cell activation and cytotoxicity Auto- weight loss. Biopsies show villous blunting with
antibodies associated with this condition include crypt hyperplasia and increased intraepithelial
antigoblet cell and antienterocyte antibodies. lymphocytes with our without prominent sub-
One variant of the syndrome is IPEX (immune epithelial collagen (collagenous sprue pattern).
dysregulation, polyendocrinopathy, enterop- An accompanying collagenous, lymphocytic, or
athy, X-linked) syndrome. It is fatal, X-linked, autoimmune gastroenteropathy-like gastritis or
and characterized by polyendocrinopathy and colitis may be seen in a subset of cases. Crypt
various autoimmune conditions in association apoptosis may be a prominent feature such that
with severe, prolonged diarrhea (22). Another, autoimmune enteropathy can be considered.
termed APECED (autoimmune polyendocr- Lanthanum (Fosrenol), is used in dialysis
inopathy-candidiasis-ectodermal dystrophy) patients to reduce their hyperphosphatemia.
syndrome or APS-1 (autoimmune polyglandular Biopsies show lamina propria CD68-positive
syndrome 1), is autosomal recessive and affected macrophages (some multinucleated) containing
patients suffer from autoimmune enteropathy in cytoplasmic non-birefringent, brown, finely
association with endocrine abnormalities (hy- granular material that may be focally nee-
poparathyroidism, adrenocortical insufficiency, dle-shaped. These are rarely highlighted with
diabetes mellitus, thyroid disease), mucocutane- Prussian blue stain (27).
ous candidiasis, and skin manifestations such as Many drugs are now used to harness the
alopecia and vitiligo, and nail deformities (23). body's immune system to kill cancer cells. The
Intestinal biopsies show total or partial vil- key medications include CTLA4 antibodies and
lous atrophy, crypt hyperplasia, and expansion antibodies to the PD-1 system. Both of these im-
of the lamina propria by a lymphoplasmacytic munologic systems have to do with our bodies'
infiltrate. Some cases display intact villous immune recognition of "self" and "non-self"
architecture. Increased IELs may be seen but cells. Tumor cells sometimes evolve to display
often to a lesser degree than in celiac disease antigens that are recognized as "self" and thus
(18,20). Crypt apoptosis is striking in some cases the cancer cells can evade the immune system.
but may be obscured in cases with significant Programmed death-1 (PD-1) is an inhibitory
cryptitis (24). Goblet and/or Paneth cells may T-cell receptor that is involved in suppressing
be absent. The diagnosis should be considered antitumor immunity. PD-1 inhibitors have
in biopsies from adult patients labeled as having been studied with promising results, especially
"refractory sprue" and from babies and children in patients with mismatch repair deficient col-
with intractable diarrhea. m·ectal and noncolorectal cancers but initially in
melanoma (28). Ipilimumab and related drugs
MEDICATION-ASSOCIATED INJURY target the cytotoxic T-lymphocyte-associated
Medication-associated injury is always an antigen 4 (CTLA4) to enhance T-cell mediat-
issue when assessing small bowel samples (25). ed tumor destruction. The main side effect is
Medications that can be associated with small diarrhea. Small intestine biopsies can show
intestinal findings include angiotensin II re- active inflammation, increased apoptosis and
ceptor antagonists. The prototype of these is villous blunting with prominent intraepithe-
olmesartan but drugs in this class typically in- lial lymphocytosis, mimicking celiac disease
clude" sartan" in their names (figs. 5-23, 5-24), (29). Ischemic enteritis usually results from

111
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 5-23
MUCOSAL INJURY ASSOCIATED WITH LOSARTAN
A: There is intra-epithelial lymphocytosis in the small intestine.
B: This is the colon from the same patient as that depicted in A. There is striking intraepitheliallymphocytosis.
C: Note the intraepitheliallymphocytes and eosinophils. These findings are not specific but should prompt a review of
pertinent serology findings and reconciliation with the patient's medication list.
D: Note the prominent crypt apoptosis.

112
Non-Neoplastic Small Intestine

Figure 5-24
MUCOSAL INJURY
ASSOCIATED W ITH
OLMESARTAN
The findings are essentially
indistinguishable from those
associated with celiac disease
in this example where as some
examples show striking crypt
apoptosis and thus features that
overlap with those of autoimmune
enteritis and other medication
associated forms of injury.

Figure 5-25
COLCHICINE TOXICITY
Left: This sample is not particularly abnormal at low magnification but note the crypts in the center of the field .
Right: Ring mitoses are a characteristic feature.

113
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 5-26
ISCHEMIC ENTERITIS ASSOCIATED WITH KAYEXALATE
A: The medication crystals can be seen as purple structures
in the luminal exudate. The mucosa is hemorrhagic and
damaged.
B: The lamina propria is collapsed and the lamina propria
vessels are engorged. Note the luminal exudate at the top
of the field.
C: This is a kayexalate crystal. Note the rectangular
cracks in the crystal.

c
mechanical alterations (leading to resections) mars (typically hepatic lesions) to radiate them
rather than chronic vascular insufficiency. It is locally. SIRSs are Yttrium-90-incorporated mi-
seldom seen on endoscopic biopsies. A possible crospheres that are embolized into the arterial
scenario in which one might encounter an isch- supply of the liver. The spheres may become
emic picture in duodenal biopsies involves the lodged in the duodenal or gastric circulation
utilization of selective internal radiation spheres resulting in ulceration and ischemic changes.
(SIRSs)-these are injected into metastatic tu- It should be remembered that whenever there

114
Non-Neoplastic Small Intestine

Figure 5-27
LANTHANUM CARBONATE
A: The lamina propria contains macrophages filled with foreign material.
B: The material in the macrophages is purple to brown.
C: High magnification.
D: This particular example also had a component of calcium, as shown in this calcium stain.

115
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 5-28
MYCOPHENOLATE-
ASSOCIATED INJURY.
A: At low magnification this
example shows villous blunting.
B: This medication is admin-
istered to patients who have had
solid organ transplantation to
reduce the activity of cytotoxic T
cells and thereby protect the graft.
The finding associated with injury
is crypt apoptosis, which is seen
here. Of course, apoptosis is also
associated with cytomegalovirus
infection, so this must always also
be considered.
C: High magnification shows
several apoptotic bodies. The nuclei
show a popcorn type appearance.

is any doubt about injury to the GI tract, non- metaplasia. Lymphoplasmacytic expansion of
steroidal inflammatory drugs (NSAIDs) can the lamina propria is not a feature ofNSAID asso-
be blamed! They are strongly associated with ciated injury (figs. 5-31, 5-32). NSAIDs may even
mucosal damage in the small intestine. The lead to a form of strictures called "diaphragm
typical endoscopic findings consist of ulcers, disease," based on their macroscopic appearance
which may lead to strictures. The histologic (30- 32). NSAID associated lesions tend to lack
features are not specific and biopsies can show chronic inflammation, which differs from Crohn
mild to moderate active inflammation, ulcers or disease, which is discussed in chapter 7.
erosions, reactive epithelial changes, and pyloric

116
Non-Neoplastic Small Intestine

Figure 5-29
ISCHEMIC/RADIATION
TYPE INJURY ASSOCIATED
WITH YTTRIUM SPHERES
The radiation spheres have
a characteristic appearance but
note the hyalinized lamina
propria and reactive epithelial
changes.

' .
I .
.. .;,.
_-j't
I . ,); '
'1

' •I
...
I .
,,
•'
'r
~ ..
'~


' ·{
., ,~

. - ~.,·
·-

I
- I

..·.

(' !, •·
· '·.. -- .J ,...

Figure 5-30
AMYLOID DEPOSITION IN THE SMALL BOWEL MUSCULARIS MUCOSAE AND SUBMUCOSAL VESSELS
Left: Note the thick muscularis mucosae and submucosal vessels.
Right: The amyloid has an eosinophilic waxy appearance.

117
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 5-31
ULCER ASSOCIATED WITH
NONSTEROIDAL
ANTI-I NFLAMMATORY DRUGS
The lesion is "punched out"
and essentially devoid of chronic
inflammation.

Figure 5-32
DIAPHRAGM DISEASE
Left: The endoscopist has encountered a circumferential ridge that constricts the lumen.
Right: The obstructing ridge in diaphragm disease is formed by a proliferated submucosa with scarring and reactive
hyperplasia of nerves, fibrous tissue and smooth muscle that results in obstruction requiring resection. Note that chronic
inflammation is a minimal component of the process.

118
Non-Neoplastic Small Intestine

REFERENCES

1. Kim SY, Choung RS, Kwon BS, et al. Small bowel of antiendomysium and antigliadin antibodies in
pseudomelanosis associated with oral iron ther- untreated celiac disease: disappointing in clinical
apy.] Korean Med Sci 2013;28:1103 -1106. practice. Am] Gastroenterol 1999;94:888-894.
2. Van Lint P, Rossen ]W, Vermeiren S, et al. De- 15. Kurppa K, Collin P, Viljamaa M, et al. Diagnosing
tection of Giardia lamblia, Cryptosporidium mild enteropathy celiac disease: a randomized,
spp. and Entamoeba histolytica in clinical controlled clinical study. Gastroenterology
stool samples by using multiplex real-time PCR 2009;136:816-823.
. after automated DNA isolation. Acta Clin Belg 16. Daniels ]A, Lederman HM, Maitra A, Montgom-
2013;68:188-192. ery EA. Gastrointestinal tract pathology in pa-
3. Cantey PT, RayS, Lee B, et al. Study of non outbreak tients with common variable immunodeficiency
giardiasis: novel findings and implications for re- (CVID): a clinicopathologic study and review.
search. Am] Med 2011;124:1175 ell?l-1178. The Am] Surg Pathol2007;31:1800-1812.
4. Yamada E, Endo H, Nakajima A, Yamanaka S. 17. Unsworth DJ, Walker-Smith ]A. Autoimmunity
Disseminated Mycobacterium avium disease in diarrhoea! disease.] Pediatr Gastroenterol Nutr
with nodular infiltration in the small intestine, 1985;4:375-380.
detected by capsule endoscopy. Endoscopy 18. Akram S, Murray ]A, Pardi DS, et al. Adult au-
2011;43(Suppl 2);UCTN:E332-333. toimmune enteropathy: Mayo Clinic Rochester
5. Costa JH, Maciel AB, Godinho Mde N, Paula experience. Clin Gastroenterol Hepatol 2007;5:
RB, Gotardelo DR. Tumoural form of Whipple's 1282-1290; quiz 1245.
disease simulating carcinomatosis. Braz J Infect 19. Carroccio A, Volta U, Di PrimaL, et al. Autoim-
Dis 2014;18:346-349. mune enteropathy and colitis in an adult patient.
6. Meunier M, Puechal X, Hoppe E, et al. Rheumatic Dig Dis Sci 2003;48:1600-1606.
and musculoskeletal features of Whipple disease: a 20. Casis B, Fernandez-Vazquez I, Barnardos E, et
repmt of 29 cases.] Rheumatol2013 ;40:2061-2066. al. Autoimmune enteropathy in an adult with
7. Arnold CA, Moreil·a RK, Lam-Himlin D, De Petris autoimmune multisystemic involvement. Scand
G, Montgomery E. Whipple disease a century J Gastroenterol2002;37:1012-1016.
after the initial description: increased recognition 21. Volta U, De Angelis GL, Granito A, et al. Auto-
of unusual presentations, autoimmune comor- immune enteropathy and rheumatoid arthritis:
bidities, and therapy effects. Am ] Surg Pathol a new association in the field of autoimmunity.
2012;36:1066-1073. Dig Liver Dis 2006;38:926-929.
8. Dumler ]S, Baisden BL, Yardley JH, Raoult D. 22. Powell BR, Buist NR, Stenzel P. An X-linked syn-
Immunodetection ofTropheryma whipplei in in- drome of diarrhea, polyendocrinopathy, and fatal
testinal tissues from Dr. Whipple's 1907 patient. infection in infancy.] Pediatr 1982;100:731-737.
N Engl] Med 2003;348:1411-1412. 23. Perheentupa]. APS-I/ APECED: the clinical disease
9. Maki M, Mustalahti K, Kokkonen ], et al. Prev- and therapy. Endocrinol Metab Clin North Am
alence of Celiac disease among children in Fin- 2002;31:295-320, vi.
land. N Engl J Med 2003;348:2517-2524. 24. Singhi AD, Goyal A, Davison JM, Regueiro MD,
10. Fasano A. European and North American pop- Roche RL, Ranganathan S. Pediatric autoimmune
ulations should be screened for coeliac disease. enteropathy: an entity frequently associated
Gut 2003;52:168-169. with immunodeficiency disorders. Mod Pathol
11. Catassi C, Ratsch IM, Gandolfi L, et al. Why is 2014;27:543-553.
coeliac disease endemic in the people of the 25. Vieth M, Montgomery E. Medication-associat-
Sahara? Lancet 1999;354:647-648. ed gastrointestinal tract injury. Virchows Arch
12. Rostami K, Malekzadeh R, Shahbazkhani B, 2017;470:245-266.
Akbari MR, Catassi C. Coeliac disease in Middle 26. Rubio-Tapia A, Herman ML, Ludvigsson JF, et
Eastern countries: a challenge for the evolution- al. Severe spruelike enteropathy associated with
ary history of this complex disorder? Dig Liver olmesartan. Mayo Clin Proc 2012;87:732-738.
Dis 2004;36:694-697. 27. Haratake ], Yasunaga C, Ootani A, Shimajiri S,
13. Marsh MN. Gluten, major histocompatibility Matsuyama A, Hisaoka M. Peculiar histiocytic
complex, and the small intestine. A molecular lesions with massive lanthanum deposition in
and immunobiologic approach to the spectrum dialysis patients treated with lanthanum carbon-
of gluten sensitivity ('celiac sprue'). Gastroenter- ate. The Am] Surg Pathol2015;39:767-771.
ology 1992;102:330-354.
14. Rostami K, Kerckhaert ], Tiemessen R, von
Blomberg BM, Meijer JW, Mulder CJ. Sensitivity

119
Survival Guide to Gastrointestinal Mucosal Biopsies

28. Le DT, Uram JN, Wang H, et al. PD-1 blockade vascular hamartoma of the small bowel. Am ]
in tumors with mismatch-repair deficiency. New Surg Pathol1999;23:1414-1417.
Engl] Med 2015;372:2509-2520. 31. Lang ], Price AB, Levi A], Burke M, Gumpel JM,
29. Oble DA, Mino-Kenudson M, Goldsmith], et al. Bjarnason I. Diaphtagm disease: pathology of
Alpha-CTLA-4 mAb-associated panenteritis: a disease of the small intestine induced by non-ste-
histologic and immunohistochemical analysis. roidal anti-inflammatory drugs. ] Clin Pathol
Am] Surg Pathol2008;32:1130-1137 . 1988;41:516-526.
30. Cortina G, WrenS, Armstrong B, Lewin K, Fajardo 32. Smith ]A, Pineau BC. Endoscopic therapy of
L. Clinical and pathologic overlap in nonsteroi- NSAID-induced colonic diaphragm disease: two
dal anti-inflammatory drug-related small bowel cases and a review of published reports. Gastro-
diaphragm disease and the neuromuscular and intest Endosc 2000;52:120-125.

120
6 SMAll BOWEl NEOPlASMS

.Nodules and masses of the duodenum and POLYPS


ileum are often biopsied whereas few biopsies
Tubular/Tubulovillous Adenoma
are received from the jejunum or proximal
ileum (since these areas cannot be reached en- Small intestinal adenomas can be classified
doscopically). Many of the nodules sampled are as tubular, tubulovillmis, and villous (figs. 6-3,
benign reactive lesions and they can often be 6-4). So-called traditional serrated adenoma
distinguished from adenomas by noting gastric (fig. 6-5) of the duodenum is rare and reported
mucin cell metaplasia as discussed in chapter cases have had inconsistent molecular profiles
5. The most important concept to recall is that (1). Adenomas usually arise in the colon and
neoplasms from other sites that spread to the are further presented in chapter 8.
small bowel tend to colonize the surface and Adenomas should be removed via endoscopic
mimic an in situ component (figs. 6-1, 6-2). polypectomy to prevent progression to carcino-
However, there are a few types of adenomas that ma but large sessile lesions often require surgery.
can be encountered in the small bowel. Many surgical colleagues manage high grade

Figure 6-1
PANCREATIC CARCINOMA COLONIZING DUODENUM
Left: The tumor extends nearly to the surface thereby mimicking an in situ component.
Right: This is a CDX2 stain. It labels the nuclei of the duodenal mucosa but not of the cancer cells. Of course some
pancreatic carcinomas have intestinal diffe rentiation but this one does not.

121
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 6-2
METASTATIC COLORECTAL
CARCINOMA COLONIZING DUODENUM
A: The cancer is on the left and has spread onto the
surface.
B: This is a CK20 stain, which labels the colorectal
carcinoma. This image is from the same case as figure
A. Primary small intestinal adenocarcinomas are often
CK7+/CK20- or CK7 /20 double positive whereas colorectal
carcinomas are generally CK20+/CK7-.
C: This is a negative CK7 stain. This image is from
the same case as figure 6-1, left. Primaiy small intestinal
adenocarcinomas are often CK7 +/CK20- or CK7 /20 double
positive whereas colorectal carcinomas are generally CK20+/
CK7- .

dysplasia in small bowel adenomas with radical that have spread from other sites to mimic them
surgery so the pathologist should have a high by "colonizing" the surface epithelium. The
threshold for diagnosing high grade dysplasia. presence of gastric type surface cell metaplasia
Most small intestinal adenomas arises in the is a clue to recognizing reactive lesions, as seen
duodenum where they are quite uncommon in chapter 5. Remember that adenomas have
and rare in the jejunum and ileum. Duodenal their initiation point just beneath the surface
adenomas are detected in 0.3 to 0.4 percent (2) epithelium (4), whereas reparative lesions that
of persons undergoing upper endoscopy at a occur with peptic duodenitis have theirs at the
median age of 65 years with no gender preva- base of the mucosa. The surface cells of duode-
lence (3). nal adenomas tend to have lipid accumulation
The difficulty in diagnosing adenomas in the in the neoplastic enterocytes.
small bowel is the proclivity of reparative lesions Some lesions are reasonably interpreted "in-
to mimic them and the capacity of carcinomas definite for dysplasia/adenoma" with a request

122
Small Bowel Neoplasms

Figure 6-3
DUODENAL TUBULAR ADENOMA
Left: This polyp is from a patient with familial adenomatous polyposis. There are numerous Paneth cells, a characteristic
feature of small intestinal adenomas.
Right: Note the Paneth cells. Note also the nuclear features . The nuclei are slim and elongated and more hyperchromatic
that those in normal colorectal epithelial cells. Their long axes are perpendicular to the basement membranes. Compare
them to the nuclei in the normal entrapped normal colorectal at the lower part of the field just left of center that contains
two plump goblet cells.

Figure 6-4
SMALL INTESTINAL TUBULOVILLOUS ADENOMA
Left: The villi are rather edematous in this example.
Right: This is th e surface of part of the lesion seen on the left. Note that the surface nuclei appear similar to the nuclei
below but that the cytoplasm is stuffed with lipid. Presumably adenomatous surface epithelium has the capacity to take in
lipid but not to process it for digestion in the manner of normal enterocytes. Cytoplasmic lipid is commonly seen at the
surface of small bowel adenomas.

123
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 6-5
TRADITIONAL SERRATED ADENOMA
Left: These polyps are further discussed in chapter 8. They are quite rare outside the distal colon. Even at this magnification
it is apparent that the nuclei are smaller than those in typical adenomas.
Right: This image shows nuclear detail, which can be compared to that of the tubulovillous adenoma seen in figure 6-3, right.

for additional samples. There is a precedent for aggregates and have abundant grey, PAS-positive
treating large ampullary adenomas with pancre- cytoplasm and small, basally located nuclei.
atoduodenectomy, so only diagnose adenoma Small Brunner gland nodules are commonly
if there is certainty. encountered in daily practice, mostly as part
of reactive changes.
Pyloric Gland Adenoma
Brunnergland lesions are usually hyperplastic or
Like their gastric counterparts, small bowel hamartomatous (6), but some are neoplastic (ade-
pyloric gland adenomas harbor GNAS mutations nomas and carcinomas) (figs. 6-7, 6-8). However,
(5). It is not clear if they arise from a metaplasia it can be difficult to ascertain whether a Brunner
dysplasia sequence as in the stomach but small gland lesion is hyperplastic, hamartomatous, or
bowel pyloric gland adenomas can also progress neoplastic, and hence, the noncommittal term
to adenocarcinoma. As per gastric examples, "Brunner gland polyp" can be used.
small bowel pyloric adenomas consist of closely Tumors believed to be adenomas have larger
packed tubules of neoplastic glands lined by nuclei than those associated with hyperplasias
cells with a monolayer of round nuclei and and hamartomas. Lesions reported as carcino-
amphophilic cytoplasm (fig. 6-6). mas and adenomas adjoining Brunner glands
have had overlapping morphologic, immuno-
Brunner Gland Lesions
histochemical, and genetic features with pyloric
and Pancreatic Heterotopia
gland adenomas (5, 7).
Submucosal Brunner glands are unique to the The presence of GNAS mutations in such
duodenum. They usually form tightly packed duodenal lesions is of interest, since GNAS

124
Small Bowel Neoplasms

Figure 6-6
PYLORIC GLAND ADENOMAS
A: The cytoplasm has a ground glass appearance and the nuclei are round. There are no goblet cells but nor is there
gastric foveolar type mucin.
B: This high magnification image shows the nuclear detail. Nuclei are round and arranged in a monolayer.
C: This example shows high grade dysplasia. There is normal small intestinal mucosa in the fragment at the top and the
lesion is in the fragment at the bottom. The closely packed tubules appear eosinophilic.
D: There is high grade dysplasia with enlarged nuclei that have lost their polarity and no longer form a basal monolayer at
the left. Those at the right invade the lamina propria (intramucosal carcinoma). In the small bowel, stomach, and esophagus,
this is staged as Tla.

125
Survival Guide to Gastrointestinal Mucosal Biopsies

'l

Figure 6-7
BRUNNER GLAND POLYP
A: This impressive lesion, wholly benign,
resulted in obstruction . It is lobulated and
composed of closely packed Brunner glands. In
such cases, it is often not clear whether the lesion
is hamartomatous, hyperplastic, or adenomatous,
such that the noncommittal term "polyp", which
simply indicated a projection, can be useful. Some
observers believe that an admixture of adipose
tissue confirms an interpretation of hamartoma,
which is lacking in this lesion.
B: This is a MUC6 stain from the lesion seen
in A. This staining was performed for illustrative
purposes only and not for diagnosis.
C: The admixture of adipose tissue in this
example suggests that it is a hamartoma.

mutations are characteristic of pyloric gland in such patients has been estimated at 93 percent
adenomas, and several other types of lesions, in one study (9) but others have recorded lower
including McCune Albright syndrome. figures. Extraintestinal neoplasms include those
Sometimes pancreatic heterotopia can pro- of the endometrium, lung, ovary (sex cord tumor
duce a small intestinal polyp and masquerade with annular tubules), cervix (adenoma malig-
as a neuroendocrine tumor (figs. 6-9). num), testis (sertoli cell tumors), pancreas (car-
cinomas) and breast (carcinomas). The average
Peutz-Jeghers Polyps
age of malignancy in patients with PJ syndrome
Peutz-[eghers (P]) syndrome is an inherited is about 40 years (10). This syndrome is about
cancer syndrome characterized by mucocutane- one tenth as common as FAP (the incidence of
ous melanin pigmentation and hamartomatous FAP is between 1:7,000 and 1:30,000).
intestinal polyposis, preferentially affecting the The diagnostic criteria include: 1) three or
small intestine (8). Patients with PJ syndrome more histologically confirmed PJ polyps; and/or
also develop colorectal cancer followed by 2) any number of P] polyps in an individual with
breast, small bowel, stomach, and pancreas family history of this syndrome; and/or 3) prom-
cancer. The cumulative lifetime risk of cancer inent mucocutaneous melanin pigmentation in

126
Small Bowel Neoplasms

Figure 6-8
BRUNNER GLAND ADENOMA
Left: At least some of the glands in this lesion appear neoplastic.
Right: High magnification of the lesion seen on the left.

Figure 6-9
PANCREATIC ACINAR CELL HETEROTOPIA
Left: Such lesions present as polyps. When the tissue is crushed, as in this example, these lesions are easily confused wi th
well differentiated neuroendocrine (carcinoid) tumors.
Right: This is a chromogranin stain from the lesion seen on the left.

127
Survival Guide to Gastrointestinal Mucosal Biopsies

an individual with affected relatives; or 4) any


number of polyps in an individual with promi-
nent mucocutaneous pigmentation.
Mutations or deletions in an involved gene,
LKB/STKll are found in most (80 to 94 percent)
of affected patients (11, 12). · ·
Peutz-Jeghers polyps are often easy to recognize
on large endoscopic biopsies that show the
characteristic branching smooth muscle (figs.
6-10-6-12). When biopsies are superficial or
when ulceration distorts them, the features are
unrecognizable. The polyps display arborizing,
smooth muscle cores from which the mucosa
emanates. These polyps occasionally have asso-
ciated dysplasia or (rarely) invasive carcinoma.
P] polyps may also produce a striking transmural
polyposis cystica profunda ("pseudoinvasion":
in which the glands herniate rather than in-
vade into the small bowel wall [fig. 6-12]) that
is unassociated with pseudomyxoma peritonei.
ADENOCARC INOMA
Small bowel adenocarcinomas account for
about 2 percent of GI tract turners, resulting in
Figure 6-10
less than 1 percent of GI tract cancer deaths (13) .
PEUTZ-JEGHERS POLYPS Our ability to detect them has improved over
This small bowel resection shows some large polyps as well time (14,15). Small bowel adenocarcinomas arise
as some more subtle early ones in the center of the sample.

Figure 6-11
PEUTZ-JEGHERS POLYPS
Left: Note the eosinophilic cords of smooth muscle that partition groups of site specific mucosa. The appearance is that
of disorganized site specific mucosa that has produced a polyp.
Right: This is a higher magnification image of the polyp seen on the left.

128
Small Bowel Neoplasms

in older adults (median age, 65 to 70 years) with


a male predominance and are over-represented in
African-Americans compared to Caucasians.
They arise in association with adenomatous
polyps. Alcohol consumption, smoking, and di-
ets rich in refined sugar, refined carbohydrates,
red meat, and smoked food are risk factors for
small intestinal adenocarcinoma (14). Small bow-
el adenocarcinoma can also arise in patients with
polyposis syndromes (primarily FAP, MUTYH
polyposis, hereditary nonpolyposis colon car-
cinoma syndrome [HNPCC]/Lynch syndrome,
biallelic mismatch repair deficiency syndrome,
Peutz-Jeghers syndrome, and juvenile polyposis
syndrome) (15, 16). Some are also associated with
Crohn's disease; gluten-sensitive enteropathy
(GSE); ileostomy; and ileal conduits. The relative
risk of duodenal adenocarcinoma is highest in
persons with familial adenomatous polyposis
(relative risk, 330.82) (17); whereas the relative
risk of small intestinal cancer in Crohn's disease
and celiac disease are each about 50- to lOO-fold;
but these are still rare.
Most small intestinal adenocarcinomas arise
in the duodenum, where most (65 percent) are
periampullary, and their prevalence decreases Figure 6-12
distally. The exception to this proximal loca- PEUTZ-JEGHERS POLYPS
tion occurs in patients with Crohn's disease, in This example has h erniated through the muscularis
whom 70 percent of adenocarcinomas are ileal. propria in an enteritis cystica profunda configuration . This
Small bowel adenocarcinomas are similar carries no risk for pseudomyxoma peritonei.
histologically to colorectal adenocarcinomas
and most are moderately differentiated, and
one third is poorly differentiated. Often an the ampulla of Vater but the term ampullary
adenomatous precursor can be detected (fig. 6- carcinoma indicates carcinomas centered at the
13). Unfortunately, adenomatous epithelium is ampulla (figs. 6-14-6-16).
often mimicked by tumors metastatic to the GI Ampullary carcinomas are rare, accounting
mucosa and thus the pathologist should be cau- for less than 1 percent of gastrointestinal ma-
tious about reporting small bowel adenocarci- lignant neoplasms presumably because the area
nomas as primary (18). Immunohistochemistry in which they can arise is small. Most patients
can be helpful to exclude metastatic disease. are in the seventh to ninth decade, and there
In persons with Lynch syndrome or its vari- is a male predominance. Syndromic cases (FAP,
ants, immunolabeling for mismatch repair Lynch syndrome) arise in younger patients.
proteins (MLHl, PMS2, MSH2, MSH6) can be Patients present with jaundice, abdominal pain,
informative. pancreatitis, and weight loss.
Most ampullary carcinomas have an intesti"
AMPULLARY CARCINOMAS nal phenotype versus a pancreatobiliary phe-
Ampullary adenocarcinoma has either in- notype (8). The most critical rule to remember
testinal or pancreatobiliary differentiation and is not to diagnose ampullary adenomas or car-
originates in the ampulla of Vater (8). Carcino- cinomas if there is any degree of uncertainty.
mas from the distal common bile duct, head Pancreatic carcinoma often spreads directly to
of the pancreas, or duodenum can all involve the ampulla (fig. 6-17).

129
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 6-13
SMALL BOWEL ADENOCARCINOMA
Left: This small bowel adenocarcinoma (left) arose in association with an adenoma in the duodenum in a patient with
celiac disease.
Right: This is high magnification of the lesion seen on the left.

Figure 6-14
AMPULLARY ADENOMA
Left: The lesion is mostly in the upper right part of the field. There are many ampullary glands in the center. They tend
to be admixed with smooth muscle in this site and are biliary type glands that lack gastric or intestinal type mucin.
Right: This is a higher magnification of the area seen on the left.

130
Small Bowel Neoplasms

Figure 6-15 Figure 6-16


AMPULLARY ADENOCARCINOMA AMPULLARY ADENOCARCINOMA,
COLONIZING DUODENAL MUCOSA GROSS SPECIMEN
Note the Brunner glands at the lower left. The carcinoma An adenoma is seen to the left of the carcinoma. Small
at the top has colonized the surface at the top thereby intestine and a massively dilated biliary tree appears at
mimicking an in situ component. the top.

Figure 6-17
SUBTLE ADENOCARCINOMA IN AN AMPULLARY BIOPSY
Left: This well differentiated adenocarcinoma is deceptively bland. There is a large malignant gland at 10:00 to 12:00 and
small ampullary glands in the center of the field.
Right: The malignant gland at left center does not match the bland biliary glands at the lower right. A small intestinal
gland at the upper left is partly colonized by carcinoma and a malignant gland is at the upper right.

131
Survival Guide to Gastrointestinal Mucosal Biopsies

. ,(, .
.~f\
~ .

Figure 6-18
SARCOMATOID LUNG CANCER
Top: The lesion from a lung has a "bottom up" growth pattern.
Bottom: This is a CAM5 .2 keratin stain from the lesion seen on the left. The mesothelial cells on the small bowel serosa,
the small bowel epithelium, and the carcinoma are all reactive.

Metastases must always be considered (figs.


6-18, 6-19). Features supporting a metastatic
tumor include the presence of multiple lesions,
the absence of a precursor adenoma, a histologic
appearance of tumor being "bottom heavy,"
and lack of ulceration. Common metastatic
lesions include melanoma, colonic adenocar-
cinoma, renal cell carcinoma, testicular/germ
cell neoplasms, and breast and gynecologic
malignancies.
NEUROENDOCRINE TUMORS
As noted in chapter 5, well differentiated
neuroendocrine (carcinoid) tumors are easy to
overlook in mucosal biopsies but are always
worth looking for in duodenal biopsies from
nodules (fig. 6-20).
Small bowel well-differentiated neuroen-
docrine tumors (WDNETs, carcinoid tumors)
comprise about one third of small intestine
neoplasms. They appear to be over-represented
in African-Americans (19).
Duodenal WDNETs account for approximate-
ly 20 percent of GI tract WDNETs (20) . Overall,
duodenal WDNETs are more common in men
Figure 6-19 (the male-to-female ratio is 1.5:1), with a me-
SARCOMATOID LUNG CANCER FROM THE LUNG
dian age of about 60 years. About two thirds of
Pleomorphic sarcomas are rare as primary small bowel
duodenal WDNETs show G cell differentiation
lesions such that this appearance should suggest sarcomatoid (by immunolabeling with gastrin antibodies).
carcinoma or dedifferentiated liposarcoma. They can only be termed gastrinomas (G-cell

132
Small Bowel Neoplasms

., .

t I ": •t i'

Figure 6-20
WELL-DIFFERENTIATED NEUROENDOCRINE (CARCINOID) TUMOR
Left: The lesion is present throughout the bottom part of the image but is subtle.
Right: This is a chromogranin stain from the lesion depicted on the left.

tumors) if they are functional and produce Grade 3 tumors are high-grade neuroendonine
gastrin; only one third of gastrin-labeled tu- carcinomas (large cell or small cell types) and are
mars is functional and causes Zollinger-Ellison characterized by >20 mitoses /10 high power fields
syndrome (ZES) (see the images in chapter 4). or >20 percent Ki-67 index (fig. 6-23).
About a third of somatostatin expressing tumors WDNETs consist of a monotonous prolifera-
is associated with von Recklinghausen disease tion of small, bland, polygonal cells with mod-
(neurofibromatosis, type l/NF1) (fig. 6-21). erate amounts of cytoplasm and round, regular
Gastrinomas tend to be smaller than so- nuclei with "salt and pepper" chromatin.
matostatinomas, and a minority (15 percent) are Duodenal WDNETs are generally indolent
multicentric. They can be associated with MEN1 (overall about 5 percent mortality); however,
(multiple endocrine neoplasia, type 1, with two thirds of somatostatinomas and one half
parathyroid lesions, pancreatic endocrine/islet of functional gastrinomas behave aggressively.
cell tumors, and pituitary lesions) . They can be Poor outcome in duodenal tumors is best pre-
very diminutive and yet produce massive bulky dicted by invasion beyond the submucosa and
lymph node metastases (21, with (inaccurate) lymph node or distant metastases. Jejunoileal
reports of "primary lymph node gastrinomas." WDNETs have a worse prognosis than those
The grading system is currently in use for of the duodenum, with a 20 percent mortality
gastrointestinal neuroendocrine tumors are G 1: rate. Survival has been negatively correlated
<2 mitoses/10 high power fields or ~2 percent with distant metastases (liver), lymph node
Ki-67 index (fig. 6-22); and G2: mitotic count metastases, tumor multiplicity, mitotic rate/ki-
2-20 per 10 high power fields or 3-20 percent 67labeling index, invasion beyond submucosa,
Ki-6 7 index. and female gender.

133
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 6-21
WELL-DIFFERENTIATED NEUROENDOCRINE (CARCINOID) TUMOR
Left: This is a somatostatinoma and it arose in a patient with neurofibromatosis, type 1 (NFl).
Right: Psammoma bodies (lower right) are characteristic of duodenal somatostatinomas.

Figure 6-22
WELL-DIFFERENTIATED NEUROENDOCRINE (CARCINOID) TUMOR
Left: This tumor is centered in the subm ucosa.
Right: This ileal carcinoid shows a very characteristic feature of ileal carcinoids, namely th e presence of reddish cytoplasmic
granules that point towards the outside of the tumor nests, with their contents thus "aimed" at blood vessels. Note the "salt
and pepper" chromatin.

134
Small Bowel Neoplasms

Figure 6-23
HIGH-GRADE NEUROENDOCRINE CARCINOMA
Left: In this small cell type arising in association with adenoma, the malignant cells at the top show abundant nuclear
molding.
Right: This is a high magnification of the lesion seen on the left.

An occasional pitfall is paraganglia or para- On immunohistochemistry, the tumors


gangliomas, which is more an issue in resection are reactive with S-100 protein in the spindle
specimens than in mucosal biopsies (fig. 6-24). cells and synaptophysin in ganglion-like cells.
In doubtful cases, keratin staining can be added, About half of the cases display keratin in the
which labels WDNETs but not paraganglia and epithelioid cells.
paragangliomas. Gangliocytic paragangliomas are usually
benign, but there are rare reports of regional
GANGLIOCYTIC PARAGANGLIOMA metastases (28) and a single report of a tumor-
Gangliocytic paragangliomas are essentially al- associated death (29).
ways found in the duodenum in adult patients
with an average age of about 54 years (22- 28) . LYMPHOMAS
Rare examples are found in the jejunum or even Diffuse large B-celllymphomas can arise in
the pylmus. They are usually centered in the sub- the duodenum, as can mucosa-associated lym-
mucosa. Most tumors are 3 to 4 cm, with a soft, phoid tissue (MALT) lymphomas, but the ones
yellowish, cut surface, and infiltrative borders. that are interesting in the small bowel include
These tumors have varying proportions of follicular lymphomas, mantle cell lymphomas,
three cell types (fig. 6-25): 1) spindled nerve and T-celllymphomas. Large B-celllymphomas
sheath cells; 2) ganglion-like cells; and 3) epi- can be evaluated as per those in the stomach.
thelioid cells arranged in nests ("endocrine" pat- Lymphoid follicles themselves are common
tern), and trabeculae or papillary structures. in the small intestine and ileum and most are

135
Survival Guide to Gastrointestinal Mucosal Biopsies

·,.,.'
.,1.
Figure 6-24
PARAGANGLION
A: This is a normal structure. There is a
nerve coursing through adipose tissue along the
bottom of the image with an adjoining structure
that contains both ganglion cells (center of ..·
image) and paraganglia! cells (top left).
B: This is the paraganglia! component from \~ .
the paraganglion seen in A. The nuclei are \
similar to those of carcinoid tumors but the
cytoplasm is grey. If there is any doubt, keratin
stains are negative in paraganglia.
C: This is the ganglion cell component from
the paraganglion seen in figure A. There are
admixed Schwann cells with spindled nuclei.
The ganglion cells have prominent nucleoli
(upper left and lower right) and abundant
cytoplasm that contains purple Nissl substance
(see best in the cells at the top).

reactive and require no immunolabeling. A lymphoma must be distinguished from mantle


pitfall is that sometimes nonneoplastic B cells cell lymphoma, which can be aggressive and
in the terminal ileum eo-label with CD43 (30). relentless. The key immunostain to use for
The special lymphoma to recognize in the mantle cell lymphoma is BCLl/cyclin Dl. A
duodenum is follicular lymphomas, which is panel for lymphomas containing small B cells
frequently an indolent process detected in the as detected on CD20 can include BCL2, cyclin
evaluation of an unrelated condition such as Dl, and CDS (to capture small lymphocytic
gastroesophageal reflux disease. This type of lymphoma/leukemia).

136
Small Bowel Neoplasms

Figure 6-25
GANGLIOCYTIC PARAGANGLIOMA
A: The tumor is centered in the submucosa.
B: There are three components. Epithelioid
nests of endocrine type cells (lower half of the
image, Schwann cells (top) and ganglion cells
(throughout) comprise the lesion.
C: This image shows Schwann cells and
ganglion cells. In small biopsies that lack all
components, this lesion can be interpreted as a
nerve sheath tumor whereas if only the epithelioid
component is sampled, an interpretation of
carcinoid is likely. .Gangliocytic paragangliomas
have keratin expression in the epithelioid
component in about half of cases in contrast
to paraganglia, which lack keratin expression.

follicular neoplasia (31). Rare cases progress to


Follicular Lymphoma
diffuse large B cell lymphoma.
In the absence of concomitant disease else- At low magnification, prominent lymphoid
where, small intestinal follicular lymphoma can follicles are seen but the apparent germinal
usually be managed by observation alone. In the centers lack typical tingible body macrophages
2016 World Health Organization (WHO) clas- (figs. 6-26, 6-27). On immunolabeling, the
sification, duodenal type follicular lymphoma is tumor cells (the ones inside the prominent fol-
a separate category that is akin so-called in situ licles) eo-express CD20, CD10, BCL2, and BCL6,

137
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 6-26
FOLLICULAR LYMPHOMA
Left: There is a large follicle at the bottom that lacks tingible body macrophages in its center.
Right: The center of the follicle contains monotonous cells with spaces ("halos" /fried egg appearance) around. The
architecture of the lesion (this is a high magnification of the large follicle on the left) is that of follicular lymphoma, but the
cytologic features overlap with those of mantle cell lymphoma and chronic lymphocytic leukemia/lymphoma, so adding
immunolabeling is sometimes important.

but not CD5 or CD43. Normal cells inside the The lymphoma cells are small- to medium-
follicles are BCL2 negative so the co-labeling sized with minimal cytoplasm and there is often a
of the cells inside the follicles with BCL2 and "fried egg" appearance (fig. 6-28); the nuclei have
BCL6 is a clue to the diagnosis. CD21 or CD23 irregular outlines and indistinct nucleoli. MCL
can highlight follicular dendritic cells if they are has a much more monotonous appearance at low
difficult to discern on routine stains. magnification than other low-grade lymphomas.
Mitotic figures are easily identified. While displace-
Mantle Cell Lymphoma
ment and/or obliteration of the glands may also
Mantle cell lymphoma (MCL) is incurable, aris- be evident, lymphoepitheliallesions characteristic
ing in older adults (mean age, 55 years) with a of MALT-type lymphomas are absent.
male predominance (of at least 2:1). Extranodal The tumor cells are B-cells that express both
disease is common, with GI tract involvement CD19, CD20, and BCL2, and aberrantly express
in about one third of MCL patients. CD5 and CD43. They lack CDlO and CD23. Cy-
When MCL involves the GI tract, both distal clin D-1 (Bel-l) expression (nuclear) is virtually
small bowel and colon involvement are typical always present.
(32). A "lymphomatous polyposis" pattern is
T-Cell Lymphomas
commonly associated with MCL but not specific
for it. Mesenteric node involvement often ac- The majority of T-cell primary GI lympho-
companies GI tract disease. mas occur in the setting of gluten-sensitive

138
Small Bowel Neoplasms

Figure 6-27
FOLLICULAR LYMPHOMA
Left: This BCL6 stain highlights the follicular center areas where aberrant expression of BCL2 can be sought.
Right: This BCL2 stain highlights cells in abnormal follicles.

Figure 6-28
MANTLE CELL LYMPHOMA
Left: Note the similarity of the cytologic features to those of the follicular lymphoma in figures 6-26 and 6-27. This lesion,
however, did not form follicles.
Right: This is a cyclin Dl (BCLl) stain. It labels the nuclei of the lymphoma cells but also the proliferating nuclei of the
reactive enterocytes.

139
Survival Guide to Gastrointestinal Mucosal Biopsies

MESENCHYMAL TUMORS

Gastrointestinal Stromal Tumors


(GISTs) of the Small Bowel
Small intestinal GISTs are more aggressive
than gastric GISTs (34,35).
Gastrointestinal Tumors of Duodenum
Duodenal GISTs are most common in the
second part of the duodenum and the most
common presenting sign is GI bleeding. Most
have spindled morphology with common focal
nuclear atypia and over 50 percent have skenoid
fibers. Duodenal GISTs are consistently immu-
noreactive with CD117 but S-100 positivity and
focal nuclear palisading in these tumors (20
percent) may raise the possibility of schwan-
noma (fig. 6-30).
Gastrointestinal Tumors of Jejunum and Ileum
Patients most commonly present with GI
bleeding (mostly insidious), but acute abdomen
with obstruction, tumor rupture with intraab-
dominal hemorrhage, and abdominal pain
mimicking appendicitis are all recorded (34).
Figure 6-29
Most tumors have spindled morphology, but
T-CELL LYMPHOMA, ENTEROPATHY-ASSOCIATED some may be epithelioid, or mixed (spindled
This example is subtle but features enlarged atypical cells and epithelioid).
in the lamina propria at the top center of the image. The presence of multiple GISTs, especially
in the small bowel (including the duodenum),
enteropathy (GSE) and are specifically referred should raise the possibility of neurofibromatosis
to as "enteropathy-type T-celllymphoma" (ETL) 1 (NF1) (34,36,37). While NF1-associated GISTs
but these are rarer than B cell tumors (fig. 6-29). are immunoreactive with CD 117, they lack KIT
ETL is rare (about 5 percent of all GI tract lym- or PDGFRA mutations, in keeping with different
phomas). Refractory GSE and ulcerative enteritis pathogenesis from that of sporadic GISTs (37).
(jejunitis) are probably represent the first step Jejuna-ileal GISTs essentially all express KIT,
toward ETL (33). sometimes in a dot-like Golgi zone distribution
The WHO 2016 classification divides intesti- whereas CD34 is found in only about 40 percent
nal T cell lymphomas into two types (31): 1) En- of cases. Smooth muscle actin expression oc-
teropathy-associated T cell lymphoma (EATL). curs in about a third and desmin expression is
This type is associated with celiac disease and exceptional and focal. S-1 00 protein expression
tends have pleomorphic cytologic features and is found in about 15 percent.
is likely to be found in persons with Northern DOG1 (discovered on GISTs1, also termed
European ancestry. Immunophenotype: CD3+, anoctamin 1 or AN01 and ORAOV2 or- over-
CDS-, CD7+, CD8 +/-, CD4-, CD56-; and 2) expressed in oral carcinoma) is another marker
Monomorphic epitheliotropic intestinal T cell (38). DOG1 is a chloride channel protein that
lymphoma (termed EATL, type 2 in the past) is was identified in GISTs by gene profiling studies
NOT associated with celiac disease and is found (3 9-41). DOG 1 is expressed in 3 6 to 9 2 perc:ent
in Asians and Hispanics and has monomorphic of KIT-negative GISTs (38). KIT is more sensitive
cytologic features. Immunophenotype: CD3+, for intestinal GISTs than DOG 1 but the two
CD4-, CD8+, CD56+. markers together (42) mark nearly all GISTs.

140
Small Bowel Neoplasms

Figure 6-30
GASTROINTESTINAL STROMAL TUMOR (GIST)
Left: The nuclei are highly monotonous and arranged in palisades that suggest a Schwannoma. Nuclear pleomorphism
is unusual in GISTs.
Right: This is a CD117 /KIT stain, showing strong membranous labeling.

The most common mutations in GISTs are Vascular malformations are often best diag-
those of exon 11 of the KIT gene. About 20 per- nosed on imaging studies but finding thick-
cent of GISTs have mutations in exon 9 instead, walled small caliber vessels in the lamina pro-
and these usually arise in the intestines (small pria suggests the diagnosis (45) .
and large). Tumors with mutations in exons 13, Angiosarcoma may involve the GI tract and
17, and 8 also tend to affect the small bowel has a tendency to display epithelioid morphol-
although there are few data on the latter since ogy. Small bowel angiosarcomas are grossly red,
they are rare (38,42). Occasional small intestinal polypoid mucosal- or serosal-based hemorrhag-
GISTs have BRAF V600E mutations. ic, friable lesions. The tumors are infiltrative
and grow as diffuse sheets of epithelioid cells
VASCULAR LESIONS with areas of clefting. Cytologically the tumors
Vascular tumors of the small bowel are uncom- show uniform, epithelioid cells with eosinophil-
mon. Lymphangiomas usually arise in children ic cytoplasm and hyperchromatic nuclei with
and can involve any portion of small intestine very prominent nucleoli (figs. 6-32, 6-33). Im-
including the Ampulla of Vater (43,44). The munohistochemical staining can be misleading
tumor is composed of numerous thin-walled since the epithelioid variant of angiosarcoma
lymphatic spaces filled with proteinaceous ma- is immunoreactive to AE1/ AE3 and may also
terial (fig. 6-31). The vessel walls may contain show immunoreactivity to other keratins but
lymphoid aggregates. "Lymphangiomatosis" not CK20.
has been applied to extensive, diffuse lesions.

141
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 6-31
LYMPHANGIOMA
Left: Dilated lymphatic spaces contain lymphocytes. Unfortunately it is impossible to know whether a mucosal lesion
found in a small biopsy such as this is a small isolated lesion or the "tip of the iceberg" of lymphangiomatosis/lymphatic
malformation, especially in biopsies from children.
Right: This is a high magnification of the lesion seen on the left.

Figure 6-32
ANGIOSARCOMA
Note the epithelioid and spindle cell
appearance at the left.

142
Small Bowel Neoplasms

Figure 6-33
ANGIOSARCOMA
Left: The malignant cells feature prominent nucleoli. Often immunolabeling is needed to confirm the interpretation;
many express keratins such that carcinoma is considered.
Right: This is a CD31 stain from the tumor seen in figure 6-32 and on the left.

REFERENCES

1. Rosty C, Campbell C, Clendenning M, Betting- 9. Giardiello FM, Brensinger]D, Tersmette AC, et al.
ton M, Buchanan DD, Brown IS. Do serrated Very high risk of cancer in familial Peutz-Jeghers
neoplasms of the small intestine represent a dis- syndrome. Gastroenterology 2000;119:1447-
tinct entity? Pathological findings and molecular 1453.
alterations in a series of 13 cases. Histopathology 10. van Lier MG, Wagner A, Mathus-Vliegen EM,
2015;66:333-342. Kuipers EJ, Steyerberg EW, van Leerdam ME.
2. Genta RM, Feagins LA. Advanced precancerous High cancer risk in Peutz-Jeghers syndrome: a
lesions in the small bowel mucosa. Best Pract Res systematic review and surveillance recommenda-
Clin Gastroenterol 2013;27:225-233. tions. Am] Gastroenterol 2010;105:1258-1264;
3. Genta RM, Hurrell]M, Sonnenberg A. Duodenal author reply 1265.
adenomas coincide with colorectal neoplasia. 11. Aretz S, Stienen D, Uhlhaas S, et al. High proportion
Dig Dis Sci 2014;59:2249-2254. of large genomic STK11 deletions in Peutz-]eghers
4. Shih IM, Wang TL, Traverso G, et al. Top-down syndrome. Hum Mutat 2005;26:513-519.
morphogenesis of colorectal tumors. Proc Natl 12. Volikos E, Robinson ], Aittomaki I<, et al. LKB1
Acad Sci US A 2001;98:2640-2645. exonic and whole gene deletions are a common
5. Vieth M, Montgomery EA. Some observations on cause of Peutz-Jeghers syndrome. ] Med Genet
pyloric gland adenoma: an uncommon and long 2006;43:e18.
ignored entity!] Clin Pathol2014;67:883-890. 13. Siegel RL, Miller KD, ]emal A. Cancer statistics,
6. Kim I<, ]ang S], Song H], Yu E. Clinicopatho- 2016. CA Cancer] Clin. 2016;66:7-30.
logic characteristics and mucin expression in 14. Aparicio T, Zaanan A, Svrcek M, et al. Small
Brunner's gland proliferating lesions. Dig Dis Sci bowel adenocarcinoma: epidemiology, risk
2013;58:194-201. factors, diagnosis and treatment. Dig Liver Dis
7. Vieth M, Kushima R, Bm·chard F, Stolte M. Pyloric 2014;46:97-104.
gland adenoma: a clinico-pathological analysis 15. Pounnand I<, Itzkowitz SH. Small bowel neoplasms
of 90 cases. Virchows Arch 2003;442:317-321. and polyps. CurT Gastroenterol Rep 2016; 18:23.
8. Bosman F, Carneiro F, Hruban R, Theise N, eds. 16. Durno CA, Sherman PM, Aronson M, et al.
WHO Classification of tumours of the digestive Phenotypic and genotypic characterisation of
system. In: Bosman F, ]affee E, Lakhani S, Ohgaki biallelic mismatch repair deficiency (BMMR-D)
H, eds. World Health Organization Classification syndrome. Eur] Cancer 2015;51:977-983.
of tumours. Lyon: IARC; 2010.

143
Survival Guide to Gastrointestinal Mucosal Biopsies

17. Offerhaus G], Giardiello FM, Krush A], et al. The its clinical significance in mantle cell lymphoma.
risk of upper gastrointestinal cancer in famil- Cancer 2003;97:586-591.
ial adenomatous polyposis. Gastroenterology 33. Daum S, Weiss D, Hummel M, et al. Frequency
1992;102:1980-1982 .. of clonal intraepithelial T lymphocyte prolif-
18. Estrella]S, Wu TT, Rashid A, Abraham SC. Mucosal erations in enteropathy-type intestinal T cell
colonization by metastatic carcinoma in the gas- lymphoma, coeliac disease, and refractory sprue.
trointestinal tract: a potential mimic of primary Gut 2001;49:804-812.
neoplasia. Am] Surg Pathol 2011;35 :563-5 72. 34. Miettinen M, Makhlouf H, Sobin LH, Lasota ].
19. Modlin IM, Lye KD, Kidd M. A 5-decade analysis of Gastrointestinal stromal tumors of the jejunum
13,715 carcinoid tumors. Cancer 2003;97:934-959. and ileum: a clinicopathologic, immunohisto-
20. Burke AP, Federspiel BH, Sobin LH, Shekitka chemical, and molecular genetic study of 906
KM, Helwig EB. Carcinoids of th e duodenum. A cases before imatinib with long-term follow-up.
histologic and immunohistochemical study of Am] Surg Pathol2006;30:477-489 .
65 tumors . Am] Surg Pathol 1989;13:828-837. 35. Miettinen M, Sobin LH, Lasota]. Gastrointestinal
21. Anlauf M, Enosawa T, Henopp T, et al. Primary stromal tumors of the stomach: a clinicopath-
lymph node gastrinoma or occult duodenal ologic, immunohistochemical, and molecular
microgastrinoma with lymph node metastases genetic study of 1765 cases with long-term fol-
in a MEN1 patient: the need for a systematic low-up. Am J Surg Pathol 2005;29:52-68.
search for the primary tumor. Am] Surg Pathol 36. Hirashima K, Takamori H, Hirota M, et al. Mu!-
2008;32:1101-1105. tiple gastrointestinal stromal tumors in neuro-
22. Burke AP, Helwig EB. Gangliocytic paraganglia- fibromatosis type 1: report of a case. Surg Today
ma. Am J Clin Pathol 1989;92:1-9. 2009;39 :979-983.
23. Hashimoto S, Kawasaki S, Matsuzawa K, Harada 37. Miettinen M, Fetsch ]F, So bin LH, Lasota]. Gas-
H, Makuuchi M. Gangliocytic paraganglioma of trointestinal stromal tumors in patients with
the papilla of Vater with regional lymph node me- neurofibromatosis 1: a clinicopathologic and
tastasis. Am] Gastroenterol1992;87:1216-1218. molecular genetic study of 45 cases. Am J Surg
24. Inai K, Kobuke T, Yonehara S, Tokuoka S. Duo- Pathol 2006;30:90-96.
denal gangliocytic paraganglioma with lymph 38. Doyle LA, Hornick JL. Gastrointestinal stromal
node metastasis in a 17-year-old boy. Cancer tumours: from KIT to succinate dehydrogenase .
1989;63:2540-2545. Histopathology 2014;64:53-67.
25. Perrone T, Sibley RI<, Rosai]. Duodenal gangliocyt- 39. Espinosa I, Lee CH, Kim MK, et al. A novel
ic paraganglioma. An immunohistochemical and monoclonal antibody against DOG 1 is a sensitive
ultrastructural study and a hypothesis concerning and specific marker for gastrointestinal stromal
its origin. Am] Surg Pathol1985;9:31-41. tumors. Am J Surg Pathol 2008;32:210-218.
26. Reed R], Caroca PJ Jr, HarkinJC. Gangliocytic para- 40. Gomez-Pinilla P] , Gibbons SJ, Bardsley MR, et al.
ganglioma. Am] Surg Pathol1977;1:207-216. Ano1 is a selective marker of interstitial cells of
27. Scheithauer BW, Nora FE, LeChago ], et al. Cajal in the human and mouse gastrointestinal
Duodenal gangliocytic paraganglioma. Clinico- tract. Am J Physiol Gastrointest Liver Physiol
pathologic and immunocytochemical study of 2009;296:G 13 70-1381.
11 cases. Am] Clin Pathol 1986;86:559-565 . 41. West RB, Corless CL, Chen X, et al. The novel mark-
28. Witkiewicz A, Galler A, Yeo C], Gross SD. Ganglia- er, DOG 1, is expressed ubiquitously in gastmintes-
cytic paraganglioma: case repmt and review of the tinal stromal tumors irrespective of KIT or PDGFRA
literature. J Gastrointest Surg 2007;11:1351-1354. mutation status. Am] Pathol2004;165:107-113.
29. LiB, Li Y, Tian XY, Luo BN, Li Z. Malignant gan- 42. Miettinen M, Lasota ]. Gastrointestinal stromal
gliocytic paraganglioma of the duodenum with tumors. Gastroenterol Clin North Am 2013;42:
distant metastases and a lethal course. World J 399-415.
Gastroenterol 2014;20:15454-15461. 43. Artaza T, Potenciano ]M, Legaz M, Munoz C,
30. Mojtahed A, Pai RI<, Anderson MW, Arber DA, Talavera A, Sanchez E. Lymphangioma of Vater's
Longacre TA. Reactive lymphoid hyperplasia of ampulla: a rare cause of obstructive jaundice.
the terminal ileum: a benign (lymphoma-like) Endoscopic therapy. Scand J Gastroenterol
condition that may harbor aberrant immunohis- 1995;30:804-806.
tochemical patterns or clonal immunoglobulin 44. Elliott RL, Williams RD, Bayles D, Griffin] .
heavy chain gene rearrangements. Appl immu- Lymphangioma of the duodenum: case report
nohistochem Mol Morphol 2014;22:585-592. with light and electron microscopic observation.
31. Swerdlow SH, Campo E, Pileri SA, et al. The Ann Surg 1966;163:86-92.
2016 revision of the World Health Organization 45 . Handra-Luca A, Montgomery E. Vascular mal-
classification of lymphoid neoplasms . Blood formations and hemangiolymphangiomas of
2016;127:2375-2390. the gastrointestinal tract: morphological fea -
32. RomagueraJE, Medeiros LJ, Hagemeister FB, et al. tures and clinical impact. Int] Clin Exp Pathol
Frequency of gastrointestinal involvement and 2011 ;4:430-443.

144
7 NON -NEOPlASTIC COlON

Like the small intestine, the colon has a lim- it is not difficult to develop a differential diag-
ited set of responses to injuries that can be di- nosis. In addition to recognizing the patterns
vided into a few general patterns. These include and some of the associations, there are a few
acute colitis, an ischemic pattern, active chronic infections that are important to recognize and
colitis, lymphocytic colitis, collagenous colitis, those are discussed initially, followed by discus-
and apoptotic colopathy with or without any sion of the patterns. Some infections can affect
of the other patterns! These patterns and some both the small intestine and colon and have
of the entities associated with them appear in been discussed in chapter 5 (strongyloidiasis,
Table 7-1. Once these patterns are recognized, Mycobacterium avium complex).

Table 7-1
PATTERNS OF NON-NEOPLASTIC DISEASE

Pattern Description Associations


Pure acute colitis Neutrophils in crypts and lamina Bacterial infections
propria; no crypt distortion, no Chemical injuries
basal plasmacytosis Phosphosoda bowel preparations (seldom used)
Ischemic colitis Hyalinized ("collapsed") lamina Vascular insufficiency
propria, atrophic crypts, surface Infections (C. difficile, enterohemorrhagic E. coli,
erosions, reactive changes; cytomegalovirus)
overall intact crypt architecture Drugs (sometimes drugs of abuse such as cocaine but
also decongestants, oral contraceptives)
Vasculitis
Active chronic colitis Cryptitis, crypt abscesses, crypt Ulcerative colitis
distortion, basal plasmacytosis, Crohn's disease
Paneth cell metaplasia in distal Diverticular disease
colon, pyloric metaplasia Some infections (histoplasmosis, amebic colitis)
Lymphocytic colitis Intraepitheliallymphocytosis Idiopathic
with or without prominent Celiac disease
crypt apoptosis Autoimmune conditions
Common variable immunodeficiency
Medications (proton pump inhibitors, angiotensin II
receptor antagonists such as losartan and olmesartan)
Collagenous colitis Intraepitheliallymphocytosis Idiopathic
with or without prominent Autoimmune conditions
crypt apoptosis and abnormal Common variable immunodeficiency
thickened subepithelial collagen Medications (proton pump inhibitors, angiotensin Il
table receptor antagonists such as losartan and olmesartan)
Apoptotic colopathy Prominent crypt apoptosis Graft versus host disease
(surface apoptosis is normal) Medications (mycophenolate, angiotensin II receptor
with or without inflammation antagonists such as losartan and olmesartan, idelalisib,
PDLl blockade drugs and other monoclonal antibodies)
Common variable immunodeficiency
Autoimmune enterocolopathy
Infections (cytomegalovirus)

145
Survival Guide to Gastrointestinal Mucosal Biopsies

..
'·..
(
•(
/

Figure 7-1
' ·- .
NORMAL COLONIC MUCOSA
The top etypt shows Paneth cells (one is indicated), which Figure 7-3
contain plump pink granules that contain lysozyme. The LEFT COLON
cytoplasm points towards the lumen so that the granules can
release their contents to protect the body. Ail endocrine cell There are far fewer inflammatory cells in the lamina propria
is indicated in the lower crypt and features tiny red granules of the left colon, than in that of the right colon.
pointed towards the lamina propria and thus the circulation.
Paneth cells are normally found in the right colon but not NORMAL COLONIC MUCOSA
the left colon. Their presence in the left colon is metaplastic.
As seen in chapter 5, the colonic crypts can
be compared to test tubes lined up in a test tube
rack (see figs. 5-1, 5-2). This basic architecture
is preserved in some abnormal conditions and
deranged in others. Paneth cells are normally
found in the cecum and ascending colon and
even sometimes the transverse colon but should
be absent in the left colon and also need to be
distinguished from neuroendocrine cells (fig.
7-1). There are more lamina propria inflamma-
tory cells in the right colon than the left colon
(figs. 7-2, 7-3). Lymphoid aggregates are also
normal and it is normal to encounter a break
in the muscularis mucosae in the area of the
lymphoid aggregates (fig. 7-4). This becomes an
important issue in assessing early carcinomas
and adenomas, shown in chapter 8. Remember
that patients with irritable bowel syndrome have
Figure 7-2
normal microscopic anatomy. Because of this, it
can be important to report normal to establish
RIGHT COLON
this diagnosis. Many rectal biopsies contain mu-
It is easy to spot inflammatory cells in the lamina ciphages (fig. 7-5), which have a very different
propria, a mixture of plasma cells, lymphocytes, and
eosinophils. A few intraepithelial eosinophils are normal appearance from cells infected with Whipple
in the right colon, and can be dismissed. bacillus and are an incidental finding.

146
Non-Neoplastic Colon

:...
- ·C\
Figure 7-4
LYMPHOID AGGREGATE
A: Note that there is a break in the muscularis mucosae at the site of the lymphoid aggregate. This is an entirely normal
finding. Note also that the nonneoplastic epithelium has herniated a bit into the aggregate. It is important to be aware of
this since adenomatous epithelium can do the same, and this does not indicate invasive carcinoma.
B: In this example, the epithelium is seen right in the gap in the muscularis mucosae and the lymphoid aggregate has
a germinal center.
C: This example produced a polyp.
D: Note the appearance of the surface epithelium over the lymphoid aggregate seen in "C." The presence of many
intraepithelial lymphocytes is entirely normal. However, finding neutrophils atop lymphoid aggregates is not normal and
can be the earliest sign of Crohn's disease in mucosal biopsies, especially those from children.

147
Survival Guide to Gastrointestinal Mucosal Biopsies

Cytomegalovirus is associated with immuno-


suppression, including that intrinsic to old age.
The viral cytopathic inclusions preferentially
affect endothelial cells but can be found in ep-
ithelial cells and stromal cells (fig. 7-7).
ACUTE COLITIS
Acute colitis essentially means finding neu-
trophils in colon crypts. It can be very focal (fig.
7-8), which can raise the possibility of Crohn's
disease in children whereas focal acute colitis
in adults can be essentially regarded as an inci-
dental finding (5,6) sometimes associated with
self-limiting infection and drugs (e.g., NSAIDs).
Campylobacter jejuni can sometimes be invoked
(7) . In children, about a quarter with focal acute
colitis are later shown to have Crohn's disease
(8). In Greenson's studies, no adult patient with
FAC developed Crohn's disease (5). In general,
infections such as Campylobacter jejuni pro-
duce an acute colitis picture with neutrophils
in crypts and in the lamina propria (fig. 7-9).
The important thing is that the basic mucosal
architecture remains intact.
Figure 7-5 ISCHEMIC COLITIS PATTERN
MUCIPHAGES The pattern of injury associated with isch-
These are very common in rectal biopsies. When they emic colitis includes marked surface injury, mu-
contain pigment (lipofuscin), the term melanosis coli can
be invoked. The latter is associated with overuse of laxatives.
cin loss in crypts, "atrophic microcrypts", and
Rectal muciphages are found in the deep lamina propria. the hyalinization of the lamina propria, wherein
the normal loose connective tissue (dotted with
plasma cells, lymphocytes, and eosinophils) is
INFECTIONS W ITH replaced with a dense eosinophilic matrix, and
CHARACTERISTIC FEATURES the residual glands become more closely spaced
Colonic spirochetosis is associated with ab- (lamina propria "collapse") (fig. 7-10) (9). Cyto-
dominal pain, appendicitis, chronic diarrhea, logic atypia can even include abnormal mitoses
and rectal bleeding but is often is an inciden- and pseudomembranes.
tal finding with no clear clinical correlates. Ischemic colitis can be associated with both
The mucosa may appear entirely normal to legal and illegal drugs (prescribed and "over-
the endoscopist or may show areas of ulcer- the-counter") (10), as well as hypertension, dia-
ation, erosion, edema, and/or erythema (1-3) . betes, chronic obstructive pulmonary disease,
Brachyspira aalborgi and Brachyspira pilosicoli, coronary artery disease, abdominal surgery,
anaerobic bacteria, seem to be responsible for use of opioids, antihypertensives, hormone
most cases of spirochetosis. The organism was replacement therapy, performance-enhancing
found in 0.7 percent of patients with chronic supplements, kayexalate (as seen in chapter 5),
watery diarrhea (4). On histologic examination and immunomodulators (10-12).
the organism produces a fuzzy pattern on the The infectious agents that can produce an
surface of the colon (fig. 7-6). Symptomatic pa- ischemic colitis pattern include enterohemor-
tients are typically treated with metronidazole, rhagic (verotoxin-producing) E. coli and Clos-
while antibiotics are not typically administered n·idium dif(icile. Stool studies are required to
to asymptomatic individuals. confirm the diagnosis.

148
Non-Neoplastic Colon

Figure 7-6
INTESTINAL SPIROCHETOSIS
A: At first glance, the sample appears entirely normal with preserved crypt architecture.
B: At high magnification, the surface appears "fuzzy."
C: This is an immunostain for syphilis but the organisms of intestinal spirochetosis cross react with the antibody. Finding
intestinal spirochetosis in a sample from a child does not indicate child abuse.
D: High magnification of the immunostain.

Radiation colitis has overlapping features with vacuoles and/or neutrophils, as well as eosinophil-
ischemic colitis. It can be acute or chronic. In acute rich crypt abscesses may be seen (13) . Chronic ra-
radiation colitis, crypts are dilated and lined by diation colitis features hyalinized vessel walls that
"flattened" atypical epithelial cells showing large tend to be aligned parallel to the surface epithelial
irregular nuclei with prominent nucleoli but a low basement membrane (fig. 7-11). Amyloidosis can
nuclear to cytoplasmic ratio . Intracytoplasmic have similar appearances (figs. 7-12, 7-13).

149
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 7-7
CYTOMEGALOVIRUS COLITIS
Left: The viral cytopathic effect is typically seen in the endothelial cells. Accompanying features can include hemorrhage,
acute inflammation and prominent crypt apoptosis. There is an enlarged infected cell in the center of the field.
Right: Two endothelial cells in the center of the image show cytoplasmic inclusions and the nuclei are smudged. Other
stromal cells in the field show smudged nuclei (Cowdry type B inclusions).

Figure 7-8
FOCAL ACUTE COLITIS
There .are a few neutrophils
in the crypt epithelium at center
right. In adults this is generally
an incidental findings akin to a
"pimple" of the colon whereas
in children it is a harbinger of
inflammatory bowel disease in 20
to 25 percent of patients.

150
Non-Neoplastic Colon

Figure 7-9
ACUTE BACTERIAL COLITIS
Left: In this acute bacterial colitis (such as from Campylobacter sp) the crypts are basically arranged in "their normal
configuration but they contain neutrophils, as does the lamina propria.
Right: In this case, the crypts are cut tangentially but they appear as evenly spaced tubules that contain neutrophils (acute
colitis); the crypts are stuffed with neutrophils so the term "crypt abscess" is used.

Figure 7-10
ISCHEMIC COLITIS
Left: The lamina propria is hyalinized compared to the looser appearance in figures 7-2 and 7-3. The number of goblet
cells is reduced.
Right: There is goblet cell loss and striking reactive change. The cells have prominent nucleoli, and an atypical mitosis
is present.

151
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 7-11
RADIATION COLITIS
Left: The fragmented portion at the upper right has hyalinized lamina propria but note the dilated lamina propria vessels
that are oriented parallel to the surface.
Right: The lamina propria capillaries have thicker walls than the normally essentially invisible lamina propria
capillaries.

Figure 7-12
AMYLOIDOSIS
Consider amyloidosis whenever
hemorrhagic colorectal samples are
encountered. The tissue is friable/
crumbly in amyloidosis and bleeds
readily on manipulation.

152
Non-Neoplastic Colon

Figure 7-13
AMYLOIDOSIS
Left: The amyloid is deposited in the muscularis mucosae, imparting a waxy appearance.
Right: This high magnification images shows the amyloid deposition in the muscularis mucosae.

sociated with exacerbations and remissions of


ACTIVE CHRONIC COLITIS hemorrhagic diarrhea (14). Disease presents at
Active chronic colitis is the key pattern for di- all ages but the peak age at presentation in the
agnosing inflammatory bowel disease. In other 15- to 25-year age range and a minor peak in
words, there is evidence of ongoing damage to the seventh decade. Children are often brought
the mucosa as well as acute damage (figs. 7-14, for medical attention before these features are
7-15). Some observers prefer the term chronic fully developed (whereas adults wait before seek-
active colitis. This does not matter, but the ing evaluation) (8,15-17). Biopsies show crypt
concept does. Acute injury is characterized by architectural distortion and increased mucosal
neutrophils, but chronic injury is characterized inflammatory cells. Neutrophils are located
not only by prominent chronic inflammation within crypt epithelium (cryptitis) and crypt
but also by crypt distortion and crypt atrophy lumina (crypt abscesses), with fewer within the
and various metaplasias. In crypt atrophy, the lamina propria around the crypts. Architectural
crypts do not extend all the way to the basement distortion refers to decreased numbers of crypts
membrane and that space may be stuffed with ("crypt drop-out") and shortened, branched,
plasma cells (basal plasmacytosis). malformed, regenerated crypts
Ulcerative colitis, the prototype for active Biopsy fragments from the same area often
chronic inflammatory disease, is a chronic show the same degree of inflammation and in-
inflammatory process that results from altered jury. Granulomas are absent, but occasionally,
immune function, characterized by a predomi- a foreign body response to a ruptured crypt
nantly mucosal-based disease and clinically as- may be found. Paneth cell metaplasia is much

153
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 7-14
ULCERATIVE COLITIS
A: The appendiceal orifice can be seen in the lower left side of the image and the entire colon is severely damaged. This is a
sample from the 1970s, when treatments were less effective than modern ones, and such colectomies are seldom seen more recently.
B: This is a so-called cecal patch, an acceptable skip area in patients with ulcerative colitis. The opening of the ileum is
in the center. A nodular red patch is present at the right in otherwise normal-appearing cecal mucosa. The patient also had
disease from the descending colon to the distal rectum.
C: The lamina propria is expanded and some crypts are missing at the bottom. There is band-like inflammation along
the muscularis mucosae and there is chronic inflammation between the bottoms of the crypts and the top of the muscularis
mucosae (basal plasmacytosis). Note that the entire tissue fragment is affected.
D: This image shows both basal plasmacytosis and Paneth cell metaplasia (the sample was from the rectum, where Paneth
cells are not normally found).

154
Non-Neoplastic Colon

Figure 7-15
CROHN'S DISEASE
A: Note that the tissue at the bottom of the image appears normal whereas the tissue at the top is damaged. In an untreated
patient, this is a clue to a diagnosis of Crohn's disease. However, once patients are treated, the disease distribution can be altered.
B: There is tremendous crypt distortion and basal plasmacytosis at the top part of the sample whereas the bottom part
is unaffected .
C: The pyloric gland metaplasia in the center is characteristic. Pyloric metaplasia is a common finding in ileal Crohn's
disease but is occasionally seen in the colon.
D: There is a granuloma at th e right of th e image and mild acute colitis in the crypt at the left. Granulomas are a marker
for Crohn's disease but not an indication of acute disease. This example shows minimal crypt distortion. In general the
degree of crypt distortion is less prominent than that in ulcerative colitis.

155
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 7-16
DIVERSION COLITIS
Left: The numerous red nodules are lymphoid
aggregates.
Below: Lymphoid aggregates are striking. At higher
magnification, cryptiti s was prominent but crypt
distortion is mild.

more common than pyloric gland metaplasia. degree of inflammation and some might be
Upper tract findings (duodenitis, gastritis) are completely uninflamed. Discrete foci of in-
sometimes reported in patients who otherwise flammation adjacent to histologically normal
have classic ulcerative colitis (18-20). Overall, crypts are common. Aphthous erosions or
however, most ulcerative colitis patients have ulcers, characterized by focal surface epithelial
no or minimal nonspecific upper GI inflamma- necrosis atop lymphoid aggregates, are typical
tion in biopsies (20). early lesions.
Crohn disease (regional enteritis, granulo-
matous enterocolitis, and terminal ileitis) is a ACTIVE CHRONIC COLITIS MIMICKERS
chronic, relapsing, and remitting inflammato- OF INFLAMMATORY BOWEL DISEASE
ry disease of unknown etiology that is often Diversion colitis is sometimes encountered. In
multifocal and can affect any portion of the GI some situations, a segment of bowel is resected,
tract. Its features focally distributed glandular but the surgeon is forced to create an ileostomy
destruction, aphthous erosions, and serpiginous or colostomy because there is simply too much
ulcers, as well as transmural inflammation, inflammation (or tumor) to reanastomose the
fibrosis, and sometimes granulomas. Because remaining bowel without clinical compromise.
of its transmural nature, fissures, sinuses, and The remaining bowel (often the rectum) has
fistulas may develop. It has an equal gender no passing fecal stream which can result in so-
incidence and occurs at all ages, with the major called diversion colitis (fig. 7-16) . If the rectum
peak incidence between 20 and 30 years of age. is left as a blind opening, continuous with the
On mucosal biopsies, several biopsies from anus, it is called a "Hartmann pouch," and the
the same general site often show a different pathologist should consider that there might

156
Non-Neoplastic Colon

,v ,·
' I
~:

i"'
. '

Figure 7-17 Figure 7-18


DIVERTICULITIS DIVERTICULAR DISEASE-ASSOCIATED COLITIS
Note the prominent inflammation in the diverticulum. This condition tends to mimic Crohn's disease but
Such inflammation can also be prominent at the surface and Crohn 's disease restricted to the sigmoid colon is distinctly
mimic inflammatory bowel disease on mucosal samples. unusual. This example shows basaLplasmacytosis and mild
crypt distortion.

be diversion colitis. The features can perfectly diagnosing IBD affecting only the sigmoid colon
mimic both ulcerative colitis and Crohn colitis. in patients with diverticular disease.
Regression of the colitis generally follows res- There are also a few infections that can
toration of the normal continuity of the fecal produce features that mimic Crohn's disease.
stream (21). The colitis is believed to result These include yersiniosis and histoplasmosis
from absence of luminal, short-chain fatty acids and sexually transmitted proctitides (syphilis
(the preferred metabolic substrate of colonic and lymphogranuloma venereum) proctitis. The
epithelium) from a change in the bacterial mi- latter should be considered in rectal samples in
lieu (21,22). The key feature is the presence of which the degree of architectural distortion does
striking numbers of lymphoid aggregates. not match the degree of chronic inflammation
Diverticular disease-associated colitis can also (figs. 7-19, 7-20). Invariably the patients prove
mimic inflammatory bowel disease (figs. 7-17, to be men who have sex with men who are HIV
7-18). The inflammatory process is restricted to positive, but this history is not always provided.
the segment of the colon containing diverticula,
LYMPHOCYTIC AND COLLAGENOUS COLITIS
which is usually the sigmoid colon, and the rec-
tum and proximal colon are not involved (23) . Lymphocytic and collagenous colitis (figs.
The histologic findings can closely mimic those 7-21-7-23) have been considered patterns of
of ulcerative colitis (24) or Crohn disease (25). colitis unified by being forms of "microscopic
Basal plasmacytosis and lymphoid aggregates colitis." This term has been used to describe
may be present but acute inflammation is usually biopsies taken from patients with symptomatic
mild. Some cases of diverticular disease even have colitis in whom (usually) there are no endo-
granulomas, which may also be found in resected scopic (gross) anomalies, but in which specific
lymph nodes (25). It is important to hesitate in microscopic features are found. Neither seems

157
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 7-19
SYPHILIS PROCTITIS
Left: There is massive chronic inflammation out of proportion to the modest degree of crypt distortion. The inflammation
is also prominent in the superficial submucosa.
Right: This is a syphilis immunostain, which highlights the organisms. Unfortunately, most examples of syphilis proctitis
are nonreactive using the syphilis immunostain so correlation with serology is required for diagnosis. Also, lymphogranuloma
venereum proctitis has the same appearances and must be confirmed by rectal swab studies.

Figure 7-20
LYMPHOGRANULOMA
VENEREUM PROCTITIS
Note that most of the inflammation
is submucosal and there is minimal
crypt distortion.

158
Non-Neoplastic Colon

Figure 7-21
LYMPHOCYTIC COLITIS PATTERN
Left: This pattern can be idiopathic but can be associated with celiac disease and a number of medications. The crypt
architecture is intact but there is prominent intraepitheliallymphocytosis in both the surface epithelium and crypt epithelium.
Right: There is no need to count to see that there are too many lymphocytes in the epithelium. There are also a few
subepithelial macrophages, which is an unremarkable finding.

Figure 7-22
COLLAGENOUS COLITIS PATTERN
Left: There is mild surface intraepithelial lymphocytosis but not much crypt lymphocytosis . No te the subepithelial
collagen that encircles capillaries and traps fibroblast nuclei. It is the quality of this collagen rather than the quantity that
matters for diagnosis. There are prominent eosinophils.
Right: This example features an expanded lamina propria. Collagenous colitis is usually most prominent in the right and
transverse colon and can be missed if only the left colon is biopsied.

159
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 7-23
EROSION IN COLLAGENOUS COLITIS
Left: Erosion is an area of damage that involves only the mucosa and does not involve the full mucosal thickness.
Right: Note the reactive stromal cells.

to impart an increased risk for development of (30). There is a broad age range at diagnosis, the
malignancy or chronic inflammatory bowel dis- mean is in the sixth to seventh decades. Men
ease such as Crohn disease or ulcerative colitis and women are equally affected. Some patients
but these patterns of injury have associations report abdominal pain and/or weight loss. Lym-
with several medications (26). phocytic colitis has a stronger association with
Collagenous colitis is usually associated with celiac disease than collagenous colitis. As with
a normal endoscopic appearance, although collagenous colitis, there is be an association
some cases of collagenous colitis may feature with medication use (e.g., NSAIDs, PPis).
linear mucosal tears or defects, especially those Collagenous and lymphocytic patterns both
associated with the use of the proton pump feature increased lamina propria cellularity,
inhibitor lansoprazole (27-29). Collagenous which is primarily attributable to plasma cells,
colitis has a female-to-male ratio of 6 to 8:1 but also lymphocytes and eosinophils. The
and a median age at diagnosis of SS years. As- crypts are nondistorted or minimally distorted.
sociated diseases include rheumatoid arthritis, There is surface epithelial injury and infiltration
thyroid disorders, celiac disease, and diabetes by lymphocytes with loss of mucin, nuclear
mellitus. Since the latter conditions are treated irregularity, pyknosis, and flattening of the
with medications that further alter the immune surface epithelium. Collagenous colitis, in ad-
system, some examples of collagenous colitis are dition to the lymphocytosis shows deposition
undoubtedly medication-associated. of an abnormal, irregular layer of collagen un-
Patients with lymphocytic colitis have chron- derneath the surface epithelium. It tends to be
ic watery diarrhea and normal colonoscopy more pronounced in the proximal colon and

160
Non-Neoplastic Colon

Figure 7-24
GRAFT VERSUS HOST DISEASE
A: There is crypt dropout and some of the crypts are
tiny and atrophic. There is a loss of inflammation in the
lamina propria.
B: The crypt in the center shows several apoptotic
bodies and the crypt at the left is nearly destroyed and
all that remains is a few endocrine cells with the granules
pointed to the lamina propria. These endocrine nests are
very characteristic of graft versus host disease.
C: The large center crypt shows an apoptotic body at
the lower left and there is an atrophic gland with residual
endocrine cells. The lamina propria lacks eosinophils. Some
patients who have had bone marrow transplants are also
taking mycophenolate, which can produce crypt apoptosis
but it is usually associated with eosinophils and few
endocrine cell nests. It is also always important to search for
cytomegalovirus before diagnosing graft versus host disease.

I
.
I
...
•I •
~

••
.• " •• -· ...
;
-'

APOPTOTIC COLOPATHY
less so in the rectosigmoid. Scattered neutro-
phils are often found in collagenous colitis but Prominent crypt apoptosis can accompany
are not a feature of lymphocytic colitis. When the patterns noted previously but itself refines
neutrophils are prominent, it suggests superim- the differential diagnosis (figs. 7-24-7-29) . There
posed infectious colitis. The surface epithelium are a few points to note when an apoptotic
tends to separate from the basement membrane colopathy pattern is seen: 1) If the patient is
beneath, leaving zones in which abnormal asymptomatic and crypt apoptotic bodies are
collagen is exposed on the surface. The surface prominent (such is in samples performed at the
lymphocytosis is usually more prominent in time of polypectomies in asymptomatic patients
lymphocytic colitis, eosinophils are less com- undergoing colorectal carcinoma screening),
mon, and prominent crypt-epithelial lympho- consider the possibility of bowel preparation
cytosis is often a feature . It is not necessary to artifact; 2) See if plasma cells are present! If
count lymphocytes. they are absent, consider common variable

161
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 7-25
CHRONIC MYCOPHENOLATE-ASSOCIATED INJURY
Left: There is crypt distortion but the sample lacks the prominent inflammation seen in inflammatory bowel disease.
Mycophenolate is administered to solid organ transplant patients to maintain their grafts (usually kidney) but is used for a
number of autoimmune conditions as well. Even at low magnification, eosinophils are notable in the center of the image.
Right: Eosinophils are readily apparent in contrast to in graft versus host disease as seen in figure 7-24.

Figure 7-26
CHRON IC MYCOPHENOLATE-
ASSOCIATED INJURY
Crypt apoptosis is striking in
this field.

162
Non-Neoplastic Colon

Figure 7-27
COLITIS ASSOCIATED WITH PEMBROLIZUMAB
Left: There is nothing specific about the findings in this image. Pembrolizumab interferes with the mechanism that allows cells
to be recognized as self to avoid destruction by our immune system. Some tu m or cells acquire "self" antigens to evade immune
surveillance and pembrolizumab and other such preparations block this system thereby creating an iatrogenic autoimmune
diathesis in the patient. There is intraepitheliallymphocytosis, prominent crypt apoptosis, and variable acute inflammation.
Right: Note the crypt apoptosis. The only way to diagnose this condition is by knowing the history, but the apoptosis
is a clue to seek it.

Table 7-2
DECODING THE NAMES OF SOME MONOCLONAL ANTIBODY DRUGS ( - mab)

Type of drug "Code" Examples


Human antiboclies -limu- Ada-limu-mab, lpi-limu-mab
Murine antibodies -Iima- Afe-limo-mab
Chimeric antibodies -lixi- Inf-lixi-mab
Humanized -lizu- Pembro-lizu-mab
Chimeric and humanized -lixizu- Ote-lixizu-mab
Interleukin -kinu- Cana-kinu-mab
Inflammatory lesion -leso- Su-leso-mab

immunodeficiency (31); 3) See if goblet cells nolate (CellCept, Myofortic); 2) Colchicine; 3)


are present. If they are absent, consider auto- Idelalisib; 4) PD-1 inhibitors (pembrolizumab,
immune enterocolopathy; and 4) Search for nivolumab); 5) Anti CTLA4 medications (ipili-
cytomegalovirus and cryptosporidium. mumab); 6) -"Sartans": Olmesartan and other
Learn the history. If the patient has had a angiotensin II receptor antagonists
solid organ transplant, consider mycopheno- Any of the monoclonal antibodies can re-
late-associated injury. Study the medication sult in an apoptotic colopathy pattern. Some
list and learn if the patient is taking any of of the monoclonal antibody preparations are
the following medications (32): 1) mycophe- described in Table 7-2.

163
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 7-28
COLITIS IN A PATIENT TAKING BOTH IPILIMUMAB AND NIVOLUMAB
A: Since both dmgs have the same associated alterations, there is no way to know which is responsible for the findings. Ipilimumab
is a CTLA4 inhibitor. This is the arm of the T cells that controls attack on self antigens to augment tumor attack by T cells.
B: There is striking intraepitheliallymphocytosis, and a few intraepithelial neutrophils are also seen.
C: There is prominent crypt apoptosis.

164
Non-Neoplastic Colon

Figure 7-29
COLITIS IN A PATIENT WITH A MUTATION IN THE GENE THAT ENCODES FOR CTLA4
Left: This patient's immune system is therefore continually activated in an autoimmune fashion. The crypt architecture
is intact. The features are essentially identical to those in persons taking anti-CTLA4 medications (ipilimumab).
Right: This is a high magnification image.

REFERENCES

1. Khashab M, Wilson S, Cho WK. Image of the 5. GreensonJK, Stern RA, Carpenter SL, BarnettJL.
month. Intestinal spirochetosis: an unusual The clinical significance of focal active colitis.
cause of asymptomatic colonic ulceration. Clin Hum Pathol 1997;28:729-733.
Gastroenterol Hepatol 2010;8:A22. 6. Volk EE, Shapiro BD, Easley KA, GoldblumJR. The
2. Tsuzawa K, Fujisawa N, Sekino Y, et al. Educa- clinical significance of a biopsy-based diagnosis
tion and imaging. Gastrointestinal: colonic of focal active colitis: a clinicopathologic study
spirochetosis. J Gastroenterol Hepatol2008;23(Pt of 31 cases. Mod Pathol1998;11:789-794.
1):1160. 7. Schneider EN, Havens JM, Scott MA, et al. Molec-
3. UmenoJ, Matsumoto T, Nakamura S, et al. Intes- ular diagnosis of Campylobacter jejuni infection
tinal spirochetosis due to Brachyspira pilosicoli: in cases of focal active colitis. Am J Surg Pathol
endoscopic and radiographic features. J Gastro- 2006;30: 782-785.
enterol 2007;42:253-256. 8. Xin W, Brown PI, Greenson JK. The clinical
4. Esteve M, Salas A, Fernandez-Banares F, et al. significance of focal active colitis in pediatric
Intestinal spirochetosis and chronic watery diar- patients. AmJ Surg Pathol2003;27:1134-1138.
rhea: clinical and histological response to treat- 9. Dignan CR, Greenson JK. Can ischemic colitis
ment and long-term follow up. J Gastroenterol be differentiated from C difficile colitis in biopsy
Hepatol2006;21:1326-1333. specimens? AmJ Surg Pathol1997;21:706-710.

165
Survival Guide to Gastrointestinal Mucosal Biopsies

10. Longstreth GF, Yao ]F. Diseases and drugs that logical study in children. Indian] Gastroenterol
increase risk of acute large bowel ischemia. Clin 2015;34:292-299.
Gastroenterol Hepatol 2010;8:49-54. 22. Harig]M, Soergel KH, Komorowski RA, Wood CM.
11. Magee CD, Moawad F], Moses F. NO-Xplode: a Treatment of diversion colitis with shmt-chain-fatty
case of supplement-associated ischemic colitis. acid irrigation. N Engl] Med 1989;320:23-28.
Mil Med 2010;175:202-205. 23. Tursi A, Elisei W, Brandimarte G, et al. The endo-
12. Zou X, Cao ], Yao Y, Liu W, Chen L. Endoscopic scopic spectrum of segmental colitis associated with
findings and clinicopathologic characteristics of diverticulosis. Colorectal Dis 2010; 12:464-470.
ischemic colitis: a report of 85 cases. Dig Dis Sci 24. Makapugay LM, Dean P]. Diverticular disease-
2009;54:2009-2015. associated chronic colitis. Am ] Surg Pathol
13. Leupin N, CurschmannJ, Kranzbuhler H, Maurer 1996;20:94-102.
CA, Laissue ]A, Mazzucchelli L. Acute radiation 25. Goldstein NS, Leon-Armin C, Mani A. Crohn's
colitis in patients treated with short-term preop- colitis-like changes in sigmoid diverticulitis speci-
erative radiotherapy for rectal cancer. Am] Surg mens is usually an idiosyncratic inflammatory
Pathol 2002;26:498-504. response to the diverticulosis rather than Crohn's
14. Tsang P, Rotterdam H. Biopsy diagnosis of coli- colitis. Am] Surg Pathol 2000;24:668-675.
tis: possibilities and pitfalls. Am ] Surg Pathol 26. Kao KT, Pedraza BA, McClune AC, et al. Micro-
1999;23:423-430. scopic colitis: a large retrospective analysis from
15. Glickman ]N, Bousvaros A, Farraye FA, et al. Pe- a health maintenance organization experience.
diatric patients with untreated ulcerative colitis World] Gastroenterol2009;15:3122-3127.
may present initially with unusual morphologic 27. Nomura E, Kagaya H, Uchimi K, et al. Linear
findings. Am] Surg Pathol 2004;28:190-197. mucosal defects: a characteristic endoscopic
16. Washington K, Greenson JK, Montgomery E, et finding of lansoprazole-associated collagenous
al. Histopathology of ulcerative colitis in ini- colitis. Endoscopy 2010;42(Suppl2):E9-10.
tial rectal biopsy in children. Am ] Surg Pathol 28. Umeno ], Matsumoto T, Nakamura S, et al. Linear
2002;26:1441-1449. mucosal defect may be characteristic of lansopra-
17. Robert ME, Tang L, Hao LM, Reyes-Mugica M. zole-associated collagenous colitis. Gastrointest
Patterns of inflammation in mucosal biopsies of Endosc 2008;67:1185-1191.
ulcerative colitis: perceived differences in pediatric 29. Yusuke H, Jun T, Naotaka M, Yuichi T, Yutaka E,
populations are limited to children younger than Kazuaki I. Lansoprazole-associated collagenous
10 years. Am] Surg Pathol2004;28:183-189. colitis: unique presentation, similar to ischemic
18. Rubenstein ], Sherif A, Appelman H, Chey WD. colitis. Endoscopy 2009;41(Suppl 2):E281-282.
Ulcerative colitis associated enteritis: is ulcerative 30. ]essurun], Yardley ]H, Lee EL, Vendrell DD, Schil-
colitis always confined to the colon?] Clin Gas- ler LR, Fordtran]S. Microscopic and collagenous
troenterol 2004;38(1):46-51. colitis: different names for the same condition?
19. Valdez R, Appelman HD, Bronner MP, Greenson Gastroenterology 1986;91:1583-1584.
JK. Diffuse duodenitis associated with ulcerative 31. Daniels]A, Lederman HM, Maitra A, Montgom-
colitis. Am] Surg Pathol 2000;24:1407-1413. ery EA. Gastrointestinal tract pathology in pa-
20. LinJ, McKenna B], Appelman HD. Morphologic tients with common variable immunodeficiency
findings in upper gastrointestinal biopsies of pa- (CVID): a clinicopathologic study and review. Am
tients with ulcerative colitis: a controlled study. ] Surg Pathol2007;31:1800-1812.
Am] Surg Pathol2010;34:1672-1677. 32. Vieth M, Montgomery E. Medication-associ-
21. Pal K, Tinalal S, AI Buainain H, Singh VP. Diver- ated gastrointestinal tract injury. Virchows Arch
sion proctocolitis and response to treatment 2017;470:245-266.
with short-chain fatty acids--a clinicopatho-

166
COLON POLYPS
8 AND NEOPLASMS

As in the stomach, in the colon, any process low the colonoscopist to search for lesions and
that produces a protuberance creates a clinical some of it seeps into the mucosa, often in areas
polyp, and some polyps are incidental benign of lymphoid aggregates, where it resembles adi-
lesions and some are clinically significant. Most pose tissue. Generally it is an incidental finding,
polyps are readily managed by endoscopic pol- but it can sometimes produce a nodule.
ypectomy. Of course it is well-established that Vegetable matter, especially pulses (beans,
removing colorectal adenomas prevents their chickpeas, lentils) can become lodged in the
progressing to adenocarcinomas . mucosa, especially in or around diverticula (and
thus in the sigmoid colon), where it can result
BENIGN FINDINGS THAT CAN MIMIC in granulomas, so-called pulse granulomas
NEOPLASMS OR OTHER DISEASES (fig. 8-3) that have appearances reminiscent of
Sometimes lymphoid aggregates result in parasitic ova!
a clinical polyp. Remember that epithelium Occasionally, heterotopic gastric mucosa can
can herniate through breaks in the muscularis be found in the rectum and form a polyp; such
mucosae associated with lymphoid aggregates polyps can result in localized erosions because
(fig. 8-1). Knowing this becomes especially they are formed by oxyntic mucosa, which
important when adenomatous epithelium produces acid (fig. 8-4). If the patient has had
involves the mucosa that has herniated into an appendectomy, the stump can occasionally
the lymphoid aggregates. It is also common to invert and present as a polyp. When this is
encounter air that has entered the tissue in the biopsied, the distorted muscularis propria can
area of lymphoid aggregates (fig. 8-2). The air is mimic a mesenchymal lesion. As a general rule,
introduced into the colon during colonoscopy it is prudent to inform colonoscopy colleagues
(air insufflation) to stretch the mucosa and al- when muscularis propria is present in the

Figure 8-1
NORMAL COLON
Notice that there is a small lymphoid
aggregate tucked just under the muscularis
mucosae . A few benign glands have
herniated into it, h aving extended through
a break in th e muscularis mucosae. Please
compare this image to figure 7-4A,B. The
colon glands (crypts) are obviously benign
in this image but it is important to be aware
of this phenomenon when adenomas
involve such glands.

167
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-2
PSEUDOLIPOMATOSIS (AIR INSUFFLATION ARTIFACT)
Left: Note the bubbles in the lymphoid aggregate and deep lamina propria. These are composed of air rather than adipose tissue.
Right: There is no tissue response to the air that forms the cleared spaces.

Figure 8-3
PULSE GRANULOMA
Left: This biopsy was from a nodule in the sigmoid colon in a patient with diverticula. There is partially mineralized
vegetable material embedded in the tissue. The mineralization suggests that the foreign material has been lodged in the
tissue for a prolonged period.
Right: The appearance can suggest a parasitic organism, but the material shows varying sizes and shapes.

168
Colon Polyps and Neoplasms

Figure 8-4
RECTAL GASTRIC HETEROTOPIA
Left: There is oxyntic mucosa (with parietal cells) at one end of the sample but the normal colorectal type mucosa is at
the other end. This lesion presented as a polyp.
Right: This image shows adjoining gastric and colonic type mucosa in rectal gastric heterotopia.

sample, since finding it often indicates that an A mimicker of sarcoma in polyps and ulcers
iatrogenic perforation has occurred at the time in the GI tract is pseudosarcomatous change,
of colonoscopy (fig. 8-5). which is seen in hypoxic fibroblasts at the
As in any other part of the gastrointestinal interface between viable and nonviable zones
tract mucosa (or in any organ with glands), isch- (figs . 8-9) (3,4). These lesions are diagnosed by
emic damage or ulcers can result in sloughing morphology; immunolabeling is typically a
of the epithelium into the lumen. This benign meaningless expensive exercise.
mimicker of carcinoma is termed signet ring cell
change (1,2). It mimics signet ring cell carcino- MUCOSAL PROLAPSE POLYPS
ma but there are differences. Figures 8-6 and 8-7 There are certain histologic features that are
shows signet ring cell change, whereas figure 8-8 associated with mucosal prolapse (fig. 8-10), and
shows a signet ring cell carcinoma in the lamina chronic mucosal prolapse can result in ulcers and
propria of the colon. The "imposter" is found in polyps. Solitary rectal ulcer syndrome is a term
the lumen of the glands and has degenerative used to denote a pattern of mucosal prolapse
cytologic features. The signet ring cell carcinoma changes (encompassing polyps that lack ulcer-
is found in viable lamina propria. The nuclei are ation) of the rectum. The classic history is of a
larger as well. Figures 8-7, left (imposter) and 8-8, young woman with constipation and straining.
right (carcinoma) were taken at the same magni- At endoscopy, ulcers are seen in up to 70 percent
fication. Compare the appearance of signet ring of patients, usually on the anterior or anterolateral
cell change also to the in situ signet ring cell car- rectal wall, but a mass-like lesion can also be pres-
cinoma of the stomach (chapter 4, figure 4-12). ent. These patients are believed to have difficulty

169
Survival Guide to Gastrointestinal Mucosa l Biopsies

Figure 8-5
INVERTED APPENDICEAL STUMP
PRESENTING AS A POLYP
A: The center has the appearance of a mesenchymal
neoplasm but consists of muscularis propria, which
indicated that the patient has undergone an iatrogenic
perforation from the biopsy of a "polyp." When such
cases are encountered, it is good practice to contact
the colonoscopist to suggest monitoring the patient
for bowel perforation. Note that the base of the lesion
has curled such that the latera l (mucosal) margin has
the appearance of a deep m argin .
B: This is a desmin stain, which highlights the
muscularis propria.
C: This is an S-100 protein stain, which highlights
nerves a nd Auerbach's plexus in the m uscularis
propria from the same sample as that seen in A and B.

Figure 8-6
SIGNET RING CELL CHANGE IN
COLORECTAL MUCOSA ADJOINING
THE ANAL SQUAMOUS MUCOSA
This area was from a hemorrhoidectomy
specimen and there was a nearby ulcer.
These entrapped colorectal type glands show
sloughing of the epithelium and the sloughed
cells have pooled in the lumina together with
entrapped mucin. Each distended glan d is
encircled by basement membrane and well-
demarcated from the lam ina propria.

170
Colon Polyps and Neoplasms

Figure 8-7
SIGNET RING CELL CHANGE IN COLORECTAL MUCOSA ADJOINING THE ANAL SQUAMOUS MUCOSA
Left: The sloughed cells have rounded up, imparting an alarming appearance reminiscent of that of signet ring cell
carcinoma, but the cells in question are restricted to the lumen.
Right: The cells have signet ring shapes such that the nucleus has the appearance of the jewel and the outlines of the
cytoplasm the appearance of the bad portion of the ring, but the nuclei have a degenerated appearance.

Figure 8-8
COLORECTAL SIGN ET RING CELL CARCINOMA
Left: The cells have invaded the mucosa and are an integral component of the tissue rather than restricted to the lumen
of a gland.
Right: The nuclei are far larger than those of the "imposter" benign sloughed cells in figure 8-6C. Compare their size to
that of the endothelial cell in the right center portion of the image.

171
Survival Guide to Gastrointestinal Mucosal Biopsies

..
r.·.
. \r 1

.
••

:. r .
.. .

' ; ......... ]. -·
·.; ~·

,.
.,.
'!. .... '
..
Figure 8-9
PSEUDOSARCOMATOUS CELLS IN AN ULCER
Left: The surface of the image shows ulcer exudate (neutrophils and fibrin) whereas the base shows ingrowth of capillaries and
fibroblasts attempting to repair the ulcer. Note the enlarged atypical fibroblasts at the interface. These cells are entirely benign.
Right: At high magnification, the cell il} the center of the image is enlarged but has a low nucleus to cytoplasmic ratio.

Figure 8-10
MUCOSAL PROLAPSE CHANGE
Left: This is a rectal biopsy from a male patient who reported men who have sex with men (MSM) sexual practices. In
mucosal prolapse, individual strands of smooth muscle separate the crypts, replacing the lamina propria and imparting a
pink appearance in the tissue. This muscle replacement of the lamina propria also can be seen in association with neoplasms,
especially those in the sigmoid colon.
Right: This very high magnification image shows the smooth muscle that has replaced the lamina propria. Smooth muscle
features brightly eosinophilic cytoplasm and delicate longitudinal cytoplasmic striations.

172
Colon Polyps and Neoplasms

Figure 8-ll
MUCOSAL PROLAPSE POLYP OF THE RECTUM
A: Note that the crypts have serrated contours (like the
business side of a steak knife or bread knife) and the lamina
propria between the crypts is replaced by smooth muscle.
The surface of the polyp is eroded (such lesions are also
termed "solitary rectal ulcer" when they arise in the rectum).
B: At high magnification, the lamina propria smooth
muscle is readily apparent by its eosinophilic (pink)
appearance.
C: Note the delicate longitudinal striations in the smooth
muscle cells.

coordinating smooth muscle functions during yps or sessile serrated adenomas have associated
the defecation process, such that the puborec- perineuriomas/benign fibroblastic polyps in their
talis sling fails to relax at the proper time. lamina propria (fig. 8-12) (7-12) .
The lesions can have a "polypoid phase" or A pitfall in diagnosing mucosal prolapse pol-
an ulcerated phase. Crypts become angulated; yps is that mucosal prolapse changes adjacent
some have used the term "diamond-shaped" to carcinomas have identical appearances to
(fig. 8-11) (5,6). Frequently such polyps have those of isolated mucosal prolapse, so multiple
serrated features, which has resulted in confu- biopsies of large "solitary rectal ulcers" can be
sion with sessile serrated adenomas/polyps (SSA; suggested to exclude sampling error (13). "Coli-
discussed below) but sessile serrated adenomas/ tis cystica profunda" is part of the same process
polyps lack smooth muscle proliferation in the and indicates that glands have prolapsed into
lamina propria. However, some hyperplastic pol- the submucosa or muscularis propria.

173
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-12
HYPERPLASTIC POLYP WITH
ASSOCIATED PERINEURIOMA
A: The lamina propria is replaced by
a proliferation that is less eosinophilic
than the smooth muscle seen in figures
8-10, 8-11 .
B: This image shows the bubbly micro-
vesicular cytoplasm in the serrated portion
of the hyperplastic polyp at the upper left
and a nodule of the perineurioma. The
latter is essentially a benign incidental
finding that can present as a polyp or in
association with serrated polyps.
C: The stromal proliferation is far less
eosinophilic than the lamina propria
smooth muscle seen in figures 8-10, 8-11.

POLYPS: CONVENTIONAL ADENOMAS, matic than those of normal colonic epithelial


HYPERPLASTIC POLYPS, AND cells (fig. 8-13). The dysplasia is usually low-
. OTHER SERRATED POLYPS grade with regular nuclei showing maintained
Colorectal polyps are an important compo- nuclear polarity (their long axes are perpendic-
nent of gastrointestinal pathology, and most ular to the basement membrane) and the nuclei
can be easily diagnosed, using a few simple are elongated rather than round. Generally
concepts as seen in Table 8-1. when there is only low-grade dysplasia, pathol-
ogists diagnose "adenoma" without mention
Tubular Adenomas
of dysplasia, which is only mentioned if there
About half of Western people are believed to is high grade dysplasia. Apoptotic bodies are
be destined to develop a colorectal adenoma usually easily found in sporadic adenomas.
(14). Adenomas have dysplasia by definition, Some variant features include finding clear
meaning that their nuclei are more hyperchro- cell change (fig. 8-14) (15), squamous-like

174
Colon Polyps and Neoplasms

Figure 8-13
TUBULAR ADENOMA
A: The adenoma is composed of tubules and is on a long stalk. The tubules have lost their orientation with respect to th e
surface of the polyp and with respect to the muscularis mucosae.
B: Colorectal adenomas originate near the surface and, as they grow, the adenoma cells fill in the mucosa below the
surface. This early lesion shows only sur face involvement, but th e adenomatous glands have already lost their orientation
with th e surface and begin to grow la terally. Their nuclei are larger and more elongated than those of the crypts below.
C: These polyps have a "top-down" growth pattern. At this stage, the la mina propria appears relatively unremarkable.
D: Thi s example shows nonneoplastic crypts between the adenomatous ones. The neoplastic nuclei are elongated but
oriented with their long axes perpendicular to the basemen t membranes.

175
Survival Guide to Gastrointestinal Mucosal Biopsies

Table 8-1
CHARACTERISTICS OF CONVENTIONAL ADENOMAS AND SERRATED POLYPS
Polyp Type · Description Illustration
Tubular Tubular adenomas have their growth
Adenoma initiation at the surface and begin to
form tubules that lose their orientation
to the surface; the adenomatous nuclei
towards the surface are larger than the
nuclei at the base; as the adenoma
grows, the base fills in with the lesion.
The architecture of tubular adenomas
is abnormal.
The cytology is dysplastic.

Hyperplastic Hyperplastic polyps essentially have


Polyp the same architecture as colorectal
mucosa such that there is a connection
between the lumen and the base of
each crypt, which lands on the
muscularis mucosae.
A line can be drawn that communicates
from the lumen to the muscularis
mucosae (see arrow).
The architecture of hyperplastic polyps
is normal.
The surface shows serrations with
"bubbly" microvesicular cytoplasm; the
bases of the crypts are narrow and have
the appearance of normal crypts.
The cytology is nondysplastic.

morules that have varying amounts of neuro- to pre-existing normal crypts, eventually over-
endocrine differentiation (figs. 8-15, 8-16) (16), taking them.
and Paneth cell differentiation. Sometimes these
High-Grade Dysplasia and
variant patterns are seen in invasive carcinomas
Villous Features in Adenomas
that arise in association with the adenomas (15) .
Some reports have noted some degree of endo- High-grade dysplasia (fig. 8-17) is equiva-
crine differentiation in the squamous morules lent to Tis but in the colon, invasion into the
and referred to them as "microcarcinoids" (17). lamina propria is also considered as Tis (19).
Such squamous morules are often found in large Validated criteria to diagnose high-grade dys-
adenomas with high grade dysplasia. plasia in colon adenomas are not established
Adenomas originate near the surface of the even though finding it places the patient into
mucosa; early adenomas only affect the upper a more intensified surveillance protocol (20).
half of the mucosa and grow in a "top-down" The 2012 American Gastroenterological As-
fashion (18) . The adenoma cells in the superfi- sociation (AGA) guidelines for management
cial portions of the mucosa spread laterally and after diagnosis of adenomas: persons with 3 to
downward to form new crypts that first connect 10 adenomas detected at initial colonoscopy,

176
Colon Polyps and Neoplasms

Table 8-1, continued


Polyp Type Description Illustration
Sessile Sessile serrated adenomas have essentially
Serrated the same architecture as colorectal mucosa . • .......
Adenoma such that there is a connection between
the lumen and the base of each crypt,
which lands on the muscularis mucosae.
A line can be drawn that communicates
from the lumen to the muscularis mucosae
(see arrow).
The overall architecture of sessile serrated
adenomas is close to normal.
There are serrations that extend all the way
to the bases and some of the crypts branch
laterally at the bases to form L or inverted
T shaped crypts.
The cytology is nondysplastic.

Traditional Traditional serrated adenomas have a dis-


Serrated organized growth pattern such that the
Adenoma lesion loses its orientation to the mus-
cularis mucosae; no line can be drawn
as for the hyperplastic polyp and sessile
serrated adenoma.
The architecture of traditional serrated
adenomas is abnormal.
There are serrations throughout; the
glands have small outpouchings that
are indicated by stars.
The cytology is dysplastic but the
nuclei are smaller than those seen in
conventional tubular adenomas.

persons with adenomas more than 10 mm (1 In general, however, high-grade dysplasia


cm), villous adenomas, or adenomas with high and large adenoma size generally go hand in
grade dysplasia should have surveillance at 3 hand so unvalidated diagnostic thresholds for
years rather than the 5 to 10 years suggested for high-grade dysplasia are less important than
1 or 2 small typical adenomas (20) . they might seem. However, high-grade dysplasia

177
Survival Guide to Gastrointestinal Mucosal Biopsies

in colorectal adenomas can be diagnosed in


adenomas with cribriform architecture and/or
loss of nuclear polarity (figs. 8-35, 8-36). When
carcinomas arise in adenomas of the colon, lam-
ina propria invasion is considered biologically
equivalent to high-grade dysplasia.
The question of separating tubular versus tu-
bulovillous versus villous adenoma is subjective
and usually does not matter on clinical grounds.
Adenomas with "villous features" are said to
warrant closer surveillance than those without
but there are no real criteria for when an ade-
noma has "villous features," however, villous
features tend to correlate with large size-often
the adenoma size is not known when the ade-
noma is reviewed (21). If the lesion has plentiful
structures that resemble small bowel villi but
are clearly neoplastic, this can be reported as
villous adenoma (fig. 8-17).
Pseudoinvasion (Mucosal
Prolapse Change) in Adenomas
Figure 8-14
Adenomas can have striking mucosal pro-
lapse changes, especially those in .the sigmoid TUBULAR ADENOMA WITH CLEAR CELL CHANGE
colon. The neoplastic glands prolapse through This example shows prominent clear cell change. This is
essentially a histologic curiosity but sometimes carcinomas
the muscularis mucosae, often through small arise in association with adenomas with this appearance and
breaks in areas with lymphoid aggregates. This these carcinomas have a clear cell appearance.

Figure 8-15
TUBULAR ADENOMA WITH SQUAMOUS MORULES
Left: Note the more solid nests in the low magnification image.
Right: The nests in question have a slightly infiltrative appearance but the cells have bland cytologic fea tures.

178
Colon Polyps and Neoplasms

Figure 8-16
TUBULAR ADENOMA WITH SQUAMOUS MORULES
Left: Sometimes these lesion display neuroendocrine differentiation and some observers have used the term
"microcarcinoids" to describe them. Overall they are an incidental finding, but they are usually encountered in large adenomas.
Right: Note the cytologic feature compared to those of the conventional adenoma component.

Figure 8-17
HIGH DYSPLASIA IN TUBULAR ADENOMA
Left: The glands have complex cribriform (sieve-like) contours and some have luminal necrosis. The lamina propria can
be seen between them
Right: Note the luminal necrotic debris.

179
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-18
COLORECTAL TUBULOVILLOUS ADENOMA
Left: In this resection specimen the lesion has arisen in otherwise normal mucosa.
Right: The adenoma has prominent structures reminiscent of small intestinal villi coated by dysplastic (adenomatous)
epithelium.

Evaluation of Adenomas that Contain


is concerning for invasion into the submucosa
Carcinomas in Polypectomy Specimens
(Tl), especially if the glands become obstructed
("Malignant Polyp")
and inspissated mucus dissects into the sur-
rounding connective tissue (figs. 8-19, 8-20) Carcinomas develop in about one in 25 ade-
(22). The prolapsed glands often carry their nomas left in situ. When an invasive carcinoma
lamina propria but in distal polyps (sigmoid), is found in an endoscopically removed polyps
the investing lamina propria can be replaced by (defined as invading into the submucosa; T1),
smooth muscle from mucosal prolapse. a decision concerning whether to perform a
Some clues for separating invasive carcino- follow-up colectomy is made and the pathologic
ma from pseudoinvasion are: 1) in "pseudoin- evaluation is the key factor. To justify offering
vasion," lamina propria is "pulled" with the the patient a colectomy, the risk of the patient
neoplastic glands into the submucosa and thus having a metastasis must be higher than the risk
invests the misplaced glands; 2) accompanying of the patient undergoing surgery to remove a
hemosiderin in often present in pseudoinva- segment of the colon. Polyps containing invasive
sion; 3) prolapsed (pseudoinvading) glands carcinoma (figs. 8-21, 8-22) account for about
tend to be rounded (gland angulation is more 5 percent of all adenomas. The likelihood of
commonly seen in carcinoma); and 4) the sub- finding an invasive component increases with
mucosal component has the same appearance polyp size, and the incidence of invasive carci-
as the adenoma that is clearly in the mucosa- noma in adenomas more than 2 cm ranges from
sometimes there is even high-grade dysplasia. 35 to 53 percent. When technically feasible, it

180
Colon Polyps and Neoplasms

Figure 8-19
ADENOMA WITH PSEUDOINVASION
(MUCOSAL PROLAPSE INTO THE SUBMUCOSA)
A: The polypectomy margin at the right has been painted
with blue ink and the adenoma can be seen coating the
surface at the left of the image. The adenoma has prolapsed
into the submucosa. At the left it has obviously brought the
lamina propria with it. Some of the glands have lost their
connection to the surface but continued to secrete mucin
which has accumulated and extruded into the surrounding
tissue. This is not an indication of a mucinous carcinoma.
B: The adenoma at the top has extended into a lymphoid
aggregate akin to the normal epithelium in figure 8-1.
C: This is a higher magnification of B. The adenoma
glands appear angulated suggestive of carcinoma but several
of the lamina propria glands have the same appearance. It
is important to realize that high grade dysplasia can also
undergo pseudoinvasion.

is recommended that these polyps be removed in the literature. The following risk factors must
intact, rather than in fragments, with as great a be assessed (23- 28): 1) Is there a poorly differen-
margin as possible at the base or stalk. Identifica- tiated component? 2) Is the tumor excised and
tion of the resection margin allows determina- at least 1 mm from the margin (some authors
tion of both the adequacy of the excision and suggest 2 mm)? This can be assessed by making
the closest approach of the tumor, a parameter two small dots (one at the leading edge of the
that predicts the risk of tumor recurrence. tumor and the other at the nearest cauterized
Many malignant polyps often constitute a margin) and measuring the distance between
form of early carcinoma (pathologic T category them with a ruler; 3) Is there vascular space
pTl) can be managed adequately by endoscopic invasion? (figs. 8-21, right and 8-22, right) and
polypectomy alone but he incidence of an un- 4) Is there tumor budding? (fig. 8-23).
favorable outcome (i.e., lymph node metastasis Tumor budding simply means single cells
or local recurrence from residual malignancy) invading into the stroma at the invasive front of
for malignant polyps treated by polypectomy the carcinoma. In some ways this is equivalent
alone varies from 0 percent to about 20 percent to a poorly differentiated component. There

181
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-20
ADENOMA WITH PSEUDOINVASION (MUCOSAL PROLAPSE INTO THE SUBMUCOSA)
A: Note that the surface contains an adenoma and part of it has herniated into the submucosa. The entrapped glands
continue to produce mucin but have lost their connection with the lumen such that the mucin is extruded into the submucosal
tissue, sometimes with a few adenoma cells. However, the low magnification appearance is especially important, since there
is a lobular configuration.
B: This is a higher magnification image of the lesion seen in A. There is lamina propria associated with the glands at the
top of the mucus pool, a clue that this is a benign process, but the mucin in the center of the image has burst into the tissue
and elicited an inflammatory response.
C: This is a very high magnification image of the mucin pool seen in A. In the context of the overall architecture, the
glands can be regarded as sloughed rounded up adenomatous tissue.
D: There is prominent hemosiderin deposition in an area of pseudoinvasion, and the glands in the area have associated
lamina propria even though they are in the submucosa.

182
Colon Polyps and Neoplasms

Figure 8-21
ADENOCARCINOMA ARISING IN A TUBULAR ADENOMA
Left: The adenoma can be seen at the upper left and lower right and the adenocarcinoma, characterized by glands
containing abundant necrotic debris, appears at the right. The carcinoma invades the submucosa very superficially and is
present on a long stalk.
Right: The presence of vascular space invasion in the early carcinoma shown on the left is an indication for an operation
to recover lymph nodes in many cases.

are many complicated definitions for tumor absence of high-risk features, the chance of ad-
budding but Irish colleagues reported that as- verse outcome is very low such that polypecto-
sessing this "by eyeball" at low magnification my alone is considered curative. Some societies
correlated with poor outcome (29,30). If these have begun to advocate measuring the depth
colleagues noted budding at 4X magnification of invasion from the bottom of the muscularis
and confirmed it at 10X, the tumor is designat- mucosae (27) but this has not been consistently
ed as having budding. The 8th edition of the advocated in the US (probably because most
A] CC staging manual did not suggest reporting colleagues require a 2 mm margin rather than
budding (19) for cancers in polyps but it can be a 1 mm margin.
subsumed in a high grade component if it is a
Serrated Polyps
prominent feature to achieve the same outcome.
If one or more of the above features is pres- Serrated polyps can be divided into several
ent, the risk of an adverse outcome following types: 1) hype1plastic polyps (HPs) (fig. 8-24); 2)
polypectomy is estimated to be about 10 to 25 sessile serrated adenomas (SSAs, the term sessile
percent. Management is decided on an individ- serrated polyp is equally acceptable) (fig. 8-25)
ual case basis, but segmental resection of the traditional serrated adenomas (TSAs) (fig. 8-26);
involved colonic segment, local excision (e.g., and 4) sessile serrated adenomas with cytological
transanal disk excision for a low rectal lesion), dysplasia (formerly termed mixed hyperplastic/
or radiation therapy may be considered. In the adenomatous polyps [MHAPs]) (fig. 8-27).

183
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-22
ADENOCARCINOMA ARISING IN A TUBULAR ADENOMA
Left: Note the complex architecture of the glands and effacement of the normal loose lamina propria.
Right: There is subtle vascular space invasion in this image, taken from the same lesion as that shown on the left.

Figure 8-23
ADENOCARCINOMA
Left: This example shows prominent stromal desmoplasia and highly angulated glands.
Right: This image is from the leading edge of the invasive front of the tumor. There are single malignant cells "sprayed"
into the stroma at the right side of the image, an appearance termed "tumor budding," which is associated with a host of
adverse factors .

184
Colon Polyps and Neoplasms

Figure 8-24
HYPERPLASTIC POLYP
Left: There are open serrated glands towards the lumen but the bases of the crypts are slender. The basement membrane
at the top is somewhat thickened such that the surface epithelium has partly "chipped off" in processing. The serrations,
associated with eosinophilic bubbly cytoplasm, extend only about halfway down the crypts. The essential architecture of
the colon is maintained in that the glands communicate between the lumen and the muscularis mucosae.
Right: This example has numerous endocrine cells in the bases of the crypts. Endocrine cells are typically lacking in
sessile serrated adenomas.

It had been well recognized that classic ade- Sessile serrated adenomas account for 15 to
nomatous polyps are premalignant lesions (31), 20 percent of colorectal polyps (33,34) using the
but the importance of serrated lesions was not criterion of accepting them as such with only a
recognized until much later. single aberrant crypt, in line with the frequency
Hyperplastic polyps comprise about 75 of MSI-H colorectal cancer. SSAs usually arise
percent of all serrated polyps. They are an inci- in the right colon, are broad-based, and may
dental finding during screening colonoscopy. become large (several centimeters) . The endo-
They may arise singly or multiply, typically in scopic appearance is often subtle. These polyps
the rectosigmoid colon, and are small(< 5mm). are characterized by serrated crypt architecture
The serrated architecture is limited to the up- that extends to the deep crypts, and dilated
per crypt with glands that become slender at crypt bases oriented parallel to the muscularis
the base and have prominent neuroendocrine mucosae. Although the 2010 World Health Or-
cells. The cytoplasm has a bubbly appearance ganization (WHO) initially arbitrarily suggested
(microvesicular). Microvesicular hyperplastic that three "boot shaped" or "L shaped" or "T
polyps have frequent BRAF mutations while shaped crypts were required for a diagnosis (32),
KRAS mutations are identified more frequently a single altered crypt suffices (33,34). We have
in the goblet cell-rich type (32). Rarer types are also encountered peculiar oncocytic changes
the goblet cell and mucin poor types, which in SSAs. As they progress, some of these polyps
are uncommon. may develop conventional-appearing low-grade

185
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-25
SESSILE SERRATED ADENOMA (POLYP)
A: The essential colorectal architecture is maintained in that the lumina of the glands communicate with the muscularis
mucosae but the crypts themselves have sideways extensions. Serrations extend to the bottoms of the crypts. However, note
also that some of the crypts have slender bases, such as those at the right of the image. If a scant biopsy fails to demonstrate
the unusual crypt contours, this does not exclude sessile serrated adenoma. Because of such issues, many colleagues regard
essentially any right sided serrated polyp as a potential sessile serrated adenoma.
B: One of the crypts has a shape reminiscent of that of a sail boat and another like a boot or sock. Regardless, these crypts
communicate between the lumen and the muscularis mucosae.
C: The nuclei Jack the conventional dysplasia appearance seen in tubular adenomas.
D: This is the base of an oddly shaped crypt. There are no endocrine cells. ·

186
Colon Polyps and Neoplasms

Figure 8-26

TRADITIONAL SERRATED ADENOMA


A: These lesions usually arise in the left colon. Note the disorganized architecture. The adenoma cells are dysplastic but
their cytoplasm is far more eosinophilic and their nuclei far less basophilic than those of typical adenomas.
B: Note the eosinophilic cytoplasm and the outpouchings from the gland in the center.
C: This example shows especially prominent tiny crypt-like outpouchings from the main glands. These outpouchings
have been termed ectopic crypt formations and are characteristic of traditional serrated adenoma.
D: This is a very high magnification image of some ectopic crypt formations.

187
Survival Guide to Gastroin.testinal Mucosal Biopsies

Figure 8-27
SESSILE SERRATED ADENOMA WITH CYTOLOGIC DYSPLASIA
Left: The lesion is tangentially embedded so the architecture is subtle but there is a sessile serrated adenoma in the lesion
lacking dysplasia and a dysplastic component on the bottom, which resembles a typical tubular adenoma. Such lesions were
termed "mixed hyperplastic polyps/adenomas" in the past but they are simply sessile serrated adenomas in the process of
progressing to adenocarcinoma .
Right: This is a high magnification image from the dysplastic component of the polyp seen on the left. It is essentially
indistinguishable from a conventional adenoma.

dysplasia (previously termed mixed hyperplastic ready disorganized crypts are termed "ectopic
adenomatous polyps [MHAPs]), characterized crypt formations" (ECF) and they define the
on immunohistochemistry by loss of expression entity. TSAs are composed of by epithelial cells
of mismatch repair proteins (MLH1) in about with abundant, eosinophilic cytoplasm and
half of cases. Remember that loss of MLH1 cigar-shaped nuclei that are paler than the
expression at the surface of a nondysplastic dysplastic nuclei characteristic of tubular ade-
serrated polyp of colon mucosa is meaningless; nomas and they lack significant enlargement,
the surface cells are not engaging in mismatch prominent nucleoli, and apoptosis.
repair because they are not dividing so nega- Some polyps are difficult to classify but some
tive surface nuclei is normal. SSAs often have comments follow: 1) If the overall morphology
activating mutations in the BRAF gene, which is that of a hyperplastic polyp but the lesion
interferes with cellular apoptosis and, as a con- is located on the right or transverse colon, err
sequence, epithelial cells "pile up," producing on the side of diagnosing SSA; 2) Although
serrated areas characteristic of these polyps. SSAs can be encountered on the left, they are
Traditional serrated adenomas (TSAs, also uncommon in that site. Some hyperplastic
known in the literature as "serrated adenomas") polyps may display reactive epithelial changes
usually arise in the distal colon. They feature and mild, uniform crypt dilation but finding
complex villiform architecture with crypts neuroendocrine cells is a clue to a diagnosis of
that lose their orientation to the muscularis hyperplastic polyp; and 3) Some observers use
mucosae. Small outpouchings from these al- the term "serrated polyp with features of SSA"

188
Colon Polyps and Neoplasms

Figure 8-28
ADENOCARCINOMA ARISING IN ASSOCIATION
WITH A SESSILE SERRATED ADENOMA
A: The sessile serrated adenoma component is
on the left and a dysplastic component can be seen
on the right whereas the invasive carcinoma is in
the center.
B: This is a high grade dysplasia component of the
lesion shown A. Note that this component has many
lymphocytes in the epithelium. This is a feature
associated with mismatch repair defects.
C: This is the adenocarcinoma component from
the lesion seen in A.. Note all the tu m or infiltrating
lymphocytes.

or "serrated polyp, not further classified" for endoscopically worrisome lesions, segmental
examples that do not entirely fulfill SSA criteria. resection could be justified in some cases; 4) Tra-
Guidelines exist for the management of ditional SAs: complete polypectomy; follow-up
adenomatous polyps, but guidelines for the in 3 years; and 5) Serrated polyposis: annual
serrated family of polyps are less established colonoscopy with polypectomies
(20): 1) Small (<1 cm), banal-appearing HPs:
Next colonoscopy in 10 years; 2) SSA, <10 mm COLORECTAL CANCER
(1 cm): Complete polypectomy; follow-up in 5 Colorectal carcinomas are usually not difficult
years; 3) Large (:2:10 mm (1 cm) SSAs or SSAs with to recognize on colon mucosal biopsies (figs.
cytological dysplasia: Complete endoscopic pol- 8-28-8-31). Invasive carcinoma shows angulat-
ypectomy is desirable, but often the adequacy of ed glands and single cells set in a desmoplastic
excision is uncertain endoscopically because of stroma. The key issue is that mucosal biopsies
frequent poor demarcation of these lesions and are-biopsies of the mucosa. Colorectal carcino-
uncertain histology, due to the sessile nature of ma is not regarded as invasive (Tl) unless there
these polyps. If incompletely excised, follow-up is submucosal invasion resulting in occasional
with endoscopic debulking seems reasonable, doubt in samples that do not contain submucosa.
but segmental resection can be considered. For As such, if well-developed desmoplasia can be

189
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-29
ADENOCARCINOMA
ARISING IN THE CECUM
The appendix and ileum
can be seen at the right side
of the image.

Figure 8-30
POORLY-DIFFERENTIATED ADENOCARCINOMA ARISING IN
ASSOCIATION WITH COLITIS-ASSOCIATED LOW-GRADE DYSPLASIA
Left: The carcinoma is present in the center of the image.
Right: This is a cytokeratin immunostain.

190
Colon Polyps and Neoplasms

identified, even in the lamina propria, there is


nearly always an underlying invasive carcinoma
(into at least submucosa); in this situation it is
impmtant to correlate with the endoscopic ap-
pearance of the lesion. It is also important to
report whether there is an associated adenoma
component to help affirm that the lesion is
primary (associated with a precursor).
COLITIS-ASSOCIATED NEOPLASIA
Dysplasia in ulcerative colitis (figs. 8-32, 8-33)
is separated into low and high grades, and cases
in which the findings are unclear are diagnosed
as "indefinite for dysplasia ." A diagnosis of
low-grade dysplasia requires nuclear alterations
(adenoma-like) that extend onto the mucosal
surface without loss of nuclear polarity; in
contrast, high-grade dysplasia displays surface
loss of nuclear polarity. There are also unusual
cases of serrated colitis-associated dysplasia and
serrated polyps in patients with inflammatory
bowel disease. Patients with inflammatory
bowel disease can also develop adenomas and
serrated polyps that are genetically the same as Figure 8-31
sporadic adenomas. There are no perfect criteria ADENOCARCINOMA
to separate sporadic adenomas from colitis-as- This example has a micropapillary pattern, a pattern
sociated polypoid dysplasia, but if there is no that is associated with aggressive behavior. There is also
flat dysplasia, polypoid dysplasia can generally abundant necrosis, a general feature of colorectal carcinoma.

Figure 8-32
COLITIS-ASSOCIATED LOW-GRADE DYSPLASIA (LEFT) AND SERRATED EPITHELIAL
CHANGE OF THE TYPE SEEN IN LONG-STANDING INFLAMMATORY BOWEL DISEASE
The lesion at the left is essentially indistinguishable from a sporadic adenoma, but in the context of the fragment at the right,
which is damaged and highly abnormal, it can be assumed to be a colitis-associated process. The epithelium in the fragment at
the right appears similar to that of a hyperplastic polyp or sessile serrated adenoma but differs by featuring marked architechtral
distortion. Slight serration is common in colonoscopic biopsies from asymptomatic persons and should not be reported but
the findings in the fragment to the right are worth reporting as they can be regarded as equivalent to "indefinite for dysplasia."

191
SwYival Guide to Gastrointestinal Mucosal Biopsies

These polyps are hamartomatous so, in the


colon, they display colonic type mucosa. They
are formed by irregularly shaped and dilated
glands, accompanied by lamina propria that is
expanded with edematou·s granulation tissue.
Dysplasia is rare in sporadic juvenile polyps.
On the other hand, juvenile polyposis is
essentially a cancer syndrome. Diagnostic cri-
teriainclude finding: 1) more than five juvenile
polyps of the colorectum; 2) juvenile polyps
throughout the GI tract; or 3) any number of
juvenile polyps in a patient with a family histo-
ry of juvenile polyposis. Small juvenile polyps
from syndromic patients have appearances that
are identical to those of typical sporadic ones
but larger ones have an increase in the relative
amount of epithelium compared to stroma, are
are multilobulated with projecting rounded or
elongated lobes, and are more likely to have
dysplasia. The dysplasia in juvenile polyps re-
sults in an appearance indistinguishable from
that of sporadic adenomas.
NEUROENDOCRINE TUMORS
Neuroendocrine tumors of the colon and
Figure 8-33
rectum appear to be increasingly detected; they
COLITIS-ASSOCIATED LOW-GRADE DYSPLASIA are essentially limited the rectum (38). Neuro-
This focus is indistinguishable from a sporadic adenoma endocrine tumors of the colon and rectum have
and can be diagnosed as such only in context of the sur- been divided into well- differentiated (carcinoid
rounding mucosal findings. If adjoining mucosa has features
of inflammatory bowel disease, colitis-associated dysplasia tumor) and high grade categories. Well-differen-
can be diagnosed. When a lesion such as this is polypoid and tiated tumors are further divided into low (G1,
there is no flat dysplasia in a person with inflammatory bowel previously known as "carcinoids") and intermedi-
disease, management by polypectomy is safe. ate (G2, previously known as "well differentiated
endocrine carcinoma") grades based on their pro-
be managed as per sporadic adenomas in both liferative rate. High grade (G3) neuroendocrine
ulcerative colitis and Crohn's disease, with carcinomas are aggressive and include tumors
polypectomy (35,36) . Some colitis patients with small and large cell type morphology. The
have serrated changes in their colonic mucosa WHO grading system (32) is as follows: 1) G1 =
reminiscent of hyperplastic polyps but no polyp <2 mitoses/10 high power fields or <2 percent
is seen and there is striking architectural distor- Ki-67 index; 2) G2 = mitotic count 2-20 per 10
tion. Serrated epithlelial change without overt high power fields or 3 to 20 percent Ki-67 index;
dysplasia probably merits intensified follow-up and 3) Grade 3 tumors are high-grade neuroen-
similar to that for dysplasia although our patient docrine carcinomas (large cell or small cell types)
base is enriched for high-risk persons (37). and are characterized by >20 mitoses/10 high
power fields or >20 percent Ki-67 index.
JUVENILE POLYPS WDNETs of the colorectum (fig. 8-35) have a
Juvenile polyps are the most common col- spectrum of appearances, but these are unified
orectal polyps in children, and up to half of the by cells that contain small round nuclei with
time, more than one juvenile polyp is found. "salt and pepper" chromatin. An important pit-
Most sporadic juvenile polyps have a spherical fall is that they express PSAP (prostate-specific
lobulated surface, often with erosions (fig. 8-34). acid phosphatase) (39) and this can lead to an

192
Colon Polyps and Neoplasms

Figure 8-34
JUVENILE POLYP
A: Note the flat surface and the prominent dilation of the glands.
B: These polyps often have an eroded surface.
C: Note the lamina propria expansion and cystically dilated nondysplastic glands.
D: There is prominent reactive surface change. Several neutrophils are present in the surface epithelium.

193
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure8-35
RECTAL WELL-DIFFERENTIATED
NEUROENDOCRINE (CARCINOID) TUMOR
A: The lesion shows no surface component, consisting
of cells with small round nuclei.
B: The nuclei are rounded with dappled ("salt and
pepper") chromatin. A mitosis is shown in the center right.
Most such tumors are Grade 1 with rare mitoses but this
one is a Grade 2 lesion.
C: This is a ki-67 immunostain. Labeled nuclei are easy
to identify.

,.,_,. . ,
.... ~ .-
.,,
.. ... -~ ' I ,,
.'
;.
.•. -~

. ~-

. -.
.
~
•,
..
.. :::
. . .... ·
..•.... ,
, ,, · -::~; I '

~ • . 0 .- ; .. • .~::...~ .
..
t..... . .. .. . .. , , ~• ,, . - ..•w.. - ~- ' • '

' .:
·.. ~- ......:
...,.,. r:
\..:. "' .. ~~-
~· • •;
·,~~· . -: ' 'D .. .
...·... , ' .. . J... :--
t"
.
... • ... .
. . .. , -. ,., .
•:. •. .. .
.. ......',:.,t-:,.,
... :.l!:i ·
. - . ·~

.. ~· ~ · - ·- ....,' _I . _.'!

erroneous diagnosis of spread from prostate can- are safely managed by endoscopic polypectomy.
cer in both low-grade tumors (carcinoids) and Patients with small tumors (1 to 2 cm) showing
high grade ones. Most rectal WO NETs are small muscularis propria invasion and without lymph
and most are indolent with a 5-year survival rate node metastases on endoscopic ultrasound, may
of nearly 90 percent. They are only rarely asso- be managed with transanal excision. A small
ciated with adenomas (fig. 8-36) (whereas high percentage (12 percent) of GI tract neuroendo-
grade neuroendocrine carcinomas are associated crine tumors is reported in the proximal colon
with adenomas; fig. 8-37). Tumor size and mus- (mostly the cecum), and these tend to be larger
cularis propria invasion are the key predictors of more aggressive lesions (38,40).
malignant behavior in well differentiated neu- Neuroendocrine carcinomas (G3) (fig. 8-38)
roendocrine (carcinoid) tumors. Rectal WO NETs are highly aggressive tumors with small cell
larger than 2.0 cm or with locoregionallymph or large cell histology characterized by a high
node involvement are managed as per rectal mitotic rate (>20 mitoses/10 high power fields,
adenocarcinomas whereas small tumors (1 to usually much higher) and extensive necrosis.
2 cm) confined to the mucosa and submucosa Median survival time after diagnosis is 1 to 2

194
Colon Polyps and Neoplasms

Figure 8-36
WELL-DIFFERENTIATED NEUROENDOCRINE (CARCINOID) TUMOR
Left: It is unusual for these tumors to arise in association with adenomas, so this is an exceptional case. The area in
question is subtle on hematoxylin and eosin (H&E) staining.
Right: This is a chromogranin immunostain showing the lesion depicted on the left.

Figure 8-37
HIGH-GRADE NEUROENDOCRINE CARCINOMA ARISING IN ASSOCIATION WITH AN ADENOMA
Left: The malignant cells feature nucleoli in keeping with large cell type.
Right: This is a synaptophysin stain. The expression is weaker in the carcinoma than in the endocrine cells in the adenoma
component at the right.

195
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-38
NEUROENDOCRINE CARCINOMA, SMALL CELL TYPE, IN COLONIC MUCOSA
A: The lesion is in the center of the image.
B: The malignant nuclei are crushed against one another and compress one another (nuclear molding).
C: The malignant cells have colonized a pre-existing crypt and are seen in the space between the basement membrane
and the row of nuclei.
0: This is a synaptophysin stain from the carcinoma seen in figures A through C.

196
Colon Polyps and Neoplasms

Figure 8-39
DIFFUSE LARGE B-CELL LYMPHOMA IN COLONIC MUCOSA
Left: This is the most common type of lymphoma to involve the colon .
Right: Diffuse large B cell lymphoma in colonic mucosa. This is a CDZO stain.

years with no significant differences between 8-42, 8-43), and granular cell tumors (figs. 8-44)
large cell and small cell subtypes (41). (42) can be encountered but they are all uncom-
mon. Vascular malformations and angiomas can
OTHER COLORECTAL TUMORS also be seen (fig. 8-45). Lastly, it is important to
Any of the lymphomas can be encountered always consider endometriosis (which has been
in the colon, but the most common type is shown to harbor mutations usually associated
diffuse large B cell lymphoma (fig. 8-39). Simi- with cancer even though it is not a form of
larly, gastrointestinal stromal tumors (fig. 8-40), cancer) (fig. 8-46) (43), as it can be a pitfall.
leiomyomas (fig. 8-41), leiomyosarcomas (figs.

197
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-40
COLORECTAL GASTROINTESTINAL STROMAL TUMOR (GIST)
Left: Such tumors are more likely to behave in a malignant fashion than gastric GISTs. Note the relatively monotonous
cytologic features.
Right: This is a DOG 1 stain.

: .... i ·- ~
.,"ri,..
,;c-:.·~
V
(
\'
:i
.
...
,., .

''·,.:;.
,,·
.•
...
/

Figure 8-41
COLORECTAL LEIOMYOMA
Left: These benign lesions arise in association with the muscularis mucosae and are composed of cells with brightly
eosinophilic cytoplasm. They are usually found in the rectum.
Right: The lesion is quite hypocellular.

198
Colon Polyps and Neoplasms

Figure 8-42
COLORECTAL
LEIOMYOSARCOMA
The lesion is brightly eosin-
ophilic but far more cellular
than the leiomyoma seen in
figure 8-41.

Figure 8-43
COLORECTAL LEIOMYOSARCOMA
Left: Atypical nuclei are present.
Right: The fascicles of tumor cells are oriented perfectly perpendicularly to one another. Some of the cells featur e
paranuclear vacuoles.

199
Survival Guide to Gastrointestinal Mucosal Biopsies

#
. (
(

~~

Figure 8-44
COLONIC GRANULAR CELL TUMOR
A: The esophagus is the most common gastrointestinal site for these. They tend to be detected in the right colon and can
feature enlarged nuclei along with their granular cytoplasm.
B: This image highlights the nuclear enlargement of the granular cells .
C: An S-100 protein stain is strongly reactive, labeling both the nuclei and the cytoplasm.

200
Colon Polyps and Neoplasms

Figure 8-45
DIEULAFOY LESION OF THE COLON
There is a large abnormal artery extending into
the submucosa just beneath the surface. This lesion
was an incidental finding in a resection performed
for another indication but such lesions, which are
essentially vascular malformations, can lead to massive
hemorrhage if they become eroded. Most such lesions
are encountered in the stomach, where they were first
described.

Figure 8-46
VASCULAR MALFORMATION, COLON
Left: It is impossible to determine whether a lesion such as this is a small angioma or the "tip of the iceberg" from a large
vascular malformation. In general, however, large vessels are not normally encountered in the colorectal lamina propria.
When reporting such lesions it is important to point out the need for imaging to determine whether the process in small
and incidental or part of an expansive process.
Right: Vessels this thick are abnormal in the colorectal mucosa. Radiation and amyloidosis can result in thickened vessels
but in a sample such as this, which was from a child, the likely choice is a vascular malformation.

201
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 8-47
ENDOMETRIOSIS
A: This example mimics a neoplasm.
B: The endometriotic glands appear dysplastic but the
nuclei are smaller than those of adenomas.
C: This is a CDlO stain from the area seen in A and B. It
highlights abundant endometrial stroma. In endometriosis
in which the glands are sparse, it is easy to consider a
sarcoma.

REFERENCES

1. Cameselle-Teijeiro ], Abdulkader I, Forteza ]. in inflammatory pseudopolyps of the colon. Arch


Signet-ring cell change in pseudomembranous Pathol Lab Med 1986;110:833-836.
colitis versus signet-ring cell carcinoma. Am ] 4. Shekitka KM, He! wig EB. Deceptive bizarre stro-
Surg Pathol 2004;28: 1111. mal cells in polyps and ulcers of the gastrointes-
2. Wang K, Weinrach D, La! A, et al. Signet-ring cell tinal tract. Cancer 1991;67:2111-2117.
change versus signet-ring cell carcinoma: a com- 5. Warren BF, Dankwa EK, Davies ]D. 'Dia-
parative analysis. Am] Surg Pathol2003;27:1429- mond-shaped' crypts and mucosal elastin:
1433. helpful diagnostic features in biopsies of rectal
3. ]essurun], Paplanus SH, Nagle RB, Hamilton SR, prolapse. Histopathology 1990;17:129-134.
Yardley ]H, Tripp M. Pseudosarcomatous changes

202
Colon Polyps and Neoplasms

6. Warren BF, Davies ]D . Prolapse-induced inflam- and polypectomy: a consensus update by the US
matory polyps of the colorectum and anal tran- Multi-Society Task Force on Colm·ectal Cancer.
sition zone. Histopathology 1994;24:201-202. Gastroenterology 2012;143:844-857.
7. Agalmy A, Stoehr R, Vieth M, Hartmann A. 21. Appelman HD. Con: High-grade dysplasia and
Benign serrated colorectal fibroblastic polyps/ villous features should not be part of the routine
intramucosal perineuriomas are true mixed epi- diagnosis of colorectal adenomas. Am] Gastro-
thelial-stromal polyps (hybrid hyperplastic pol- enterol2008;103:1329-1331.
yp/mucosal perineurioma) with frequent BRAF 22. Pascal RR, Hertzler G, HunterS, Goldschmid S.
mutations. Am] Surg Pathol2010;34:1663-1671. Pseudoinvasion with high-grade dysplasia in a
8 . Eslami-Varzaneh F, Washington K, Robert ME, colonic adenoma. Distinction from adenocarci-
. Kashgarian M, Goldblum ]R, ]ain D. Benign noma. Am] Surg Pathol1990;14:694-697.
fibroblastic polyps of the colon: a histologic, im- 23. Cooper HS. Surgical pathology of endoscopically
munohistochemical, and ultrastructural study. removed malignant polyps of the colon and
Am J Surg Pathol 2004;28:3 74-3 78. rectum. Am J Surg Pathol 1983;7:613-623.
9. Groisman G, Amar M, Alona M. Early colonic 24. Cooper HS. Pathologi.c issues in the treatment
perineurioma: a report of 11 cases. Int ] Surg of endoscopically removed malignant colorectal
Pathol 2010;18:292-297. polyps.] Natl Compr Cane Netw 2007;5:991-996.
10. Groisman GM, Polak-Charcon S. Fibroblastic 25. Hassan C, Zullo A, Risio M, Rossini FP, Morini
polyp of the colon and colonic perineurioma: S. Histologic risk factors and clinical outcome
2 names for a single entity? Am ] Surg Pathol in colorectal malignant polyp: a pooled-data
2008;32:1088-1094. analysis. Dis Colon Rectum 2005;48:1588-1596.
11. Groisman GM, Polak-Charcon S, Appelman HD. 26. Muller S, Chesner IM, Egan M], et al. Signif-
Fibroblastic polyp of the colon: clinicopatho- icance of venous and lymphatic invasion in
logical analysis of 10 cases with emphasis on malignant polyps of the colon and rectum. Gut
its common association with serrated crypts. 1989;30: 1385-1391.
Histopathology 2006;48:431-43 7. 27. Quirke P, Risio M, Lambert R, von Karsa L, Vi-
12. Hornick JL, Fletcher CD. Intestinal perineuri- eth M, International Agency for Research on
omas: clinicopathologic definition of a new C. European guidelines for quality assurance
anatomic subset in a series of 10 cases. Am] Surg in colorectal cancer screening and diagnosis.
Pathol 2005;29:859-865. First Edition-Quality assurance in pathology
13. Li SC, Hamilton SR. Malignant tumors in the rec- in colorectal cancer screening and diagnosis .
turn simulating solitary rectal ulcer syndrome in Endoscopy 2012;44(Suppl 3):SE116-130.
endoscopic biopsy specimens. Am J Surg Pathol 28. Wilcox GM, Beck JR. Early invasive cancer in
1998;22:106-112. adenomatous colonic polyps ("malignant pol-
14. Kinzler KW, Vogelstein B. Lessons from heredi- yps"). Evaluation of the therapeutic options by
tary colorectal cancer. Cell1996;87:159-170. decision analysis . Gastroenterology 198 7;92(Pt
15. Shi C, Scudiere JR, Cornish TC, et al. Clear cell 1):1159-1168.
change in colonic tubular adenoma and corre- 29. Rogers AC, Gibbons D, Hanly AM, et al. Prognos-
sponding colonic clear cell adenocarcinoma is tic significance of turn or budding in rectal cancer
associated with an altered mucin core protein biopsies before neoadjuvant therapy. Mod Pathol
profile. Am] Surg Pathol 2010;34:1344-1350. 2014;27:156-162.
16. Sarlin JG, Mori K. Morules in epithelial tumors 30. Rogers AC, Winter DC, Heeney A, et al. System-
of the colon and rectum. Am J Surg Pathol atic review and meta-analysis of the impact of
1984;8:281-285. tumour budding in colorectal cancer. Br J Cancer
17. Salaria SN, Abu Alfa AK, Alsaigh NY, Montgomery 2016;115:831-840.
E, Arnold CA. Composite intestinal adenoma-ml- 31. Vogelstein B, Fearon ER, Hamilton SR, et al.
crocarcinoid clues to diagnosing an under-rec- Genetic alterations during colorectal-tumor
ognised mimic of invasive adenocarcinoma. ] development. N Engl J Med 1988;319:525-532.
Clin Pathol 2013;66:302-306. 32. Bosman FT, World Health Organization, Inter-
18. Shih IM, Wang TL, Traverso G, et al. Top-down national Agency for Research on Cancer. WHO
morphogenesis of colorectal tumors. Proc Natl classification of tumours of the digestive system,
Acad Sci US A 2001;98:2640-2645. 4th ed. Lyon: IARC Press; 2010.
19. Amin M, EdgeS, Greene F, Byrd DR, Brookland 33. Bettington M, Walker N, Rahman T, et al. High
RK, Washington MK. A] CC Cancer Staging Man- prevalence of sessile serrated adenomas in con-
ual. 8th ed: Switzerland: Springer; 2016. temporary outpatient colonoscopy practice.
20. Lieberman DA, Rex DK, Winawer SJ, et al. Guide- Intern MedJ 2017;47:318-323.
lines for colonoscopy surveillance after screening

203
Survival Guide to Gastrointestinal Mucosal Biopsies

34. Bettington M, Walker N, Rosty C, et al. Critical 39. Kimura N, Sasano N. Prostate-specific acid phos-
appraisal of the diagnosis of the sessile serrated phatase in carcinoid tumors. Virchows Arch A
adenoma. Am] Surg Pathol2014;38:158-166. Pathol Anat Histopathol 1986;41 0:24 7-251.
35. Odze RD, Far-raye FA, Hecht]L, HornickJL. Long- 40. Saha S, Hoda S, Godfrey R, Sutherland C, Raybon
term follow-up after polypectomy treatment for K. Carcinoid tumors of the gastrointestinal tract: a
adenoma-like dysplastic lesions in ulcerative coli- 44-year experience. South MedJ 1989;82: 1501-1505.
tis. Clin Gastroenterol Hepatol2004;2:534-541. 41. Shia ], Tang LH, Weiser MR, et al. Is nonsmall
36. Quinn AM, Farraye FA, Naini BV, et al. Polyp- cell type high-grade neuroendocrine carcinoma
ectomy is adequate treatment for adenoma-like of the tubular gastrointestinal tract a distinct
dysplastic lesions (DALMs) in Crohn's disease. disease entity? Am] Surg Pathol 2008;32:719-
Inflamm Bowel Dis 2013;19:1186-1193. 731.
37. Parian A, Koh], Limketkai BN, et al. Association 42. Singhi AD, Montgomery EA. Colorectal granular
between serrated epithelial changes and colorec- cell tumor: a clinicopathologic study of 26 cases.
tal dysplasia in inflammatory bowel disease. Am] Surg Pathol2010;34:1186-1192.
Gastrointest Endosc 2016;84:87-95 e81. 43. Anglesio MS, Papadopoulos N, Ayhan A, et al.
38. Modlin IM, Lye KD, Kidd M. A 5-decade analysis Cancer-Associated Mutations in Endometriosis
of 13,715 carcinoid tumors. Cancer 2003;97:934- without Cancer. N Engl J Med 2017;376:1835-
959. 1848.

204
9 NON-NEOPLASTIC ANUS

The length of the anus is short, such that a rectal glandular mucosa and there are anal ducts
biopsy from the anatomic (clinical) anus might and glands at the transition zone (figs. 9-1-9-3).
contain glandular, transitional, or squamous
mucosa. A biopsy labeled "anus," can show HEMORRHOIDS
colonic-type mucosa and be correctly labeled! Hemorrhoids are common (1,2). They have
Anatomically, the anal canal begins as the rec- been viewed as varicosities of submucosal
tum enters the puborectalis sling (palpated as veins but current belief is that they result from
the anorectal ring on a digital exam), ending sagging or slippage of normal structures that
at the squamous mucocutaneous junction with is exacerbated by straining at defecation. The
the perianal skin. The upper two thirds are hemorrhoids are normal anatomic structures lo-
endoderm-derived, and the lower one third is cated in the subepithelial space (1-3), consisting
ectodermally derived. The dentate, or pectinate of connective tissue cushions surrounding the
line, is where the two zones meet. direct arteriovenous communications between
Most of the anal canal is lined by squamous the terminal branches of the superior rectal
mucosa. The adjoining nonsquamous lining can arteries and the superior, inferior, and middle
consist of either transitional (urothelium-like) or rectal veins. The hemorrhoidal cushions are

Figure 9-1
ANAL TRANSITIONAL MUCOSA
Left: Note that th e basal cells are similar to those of squamous epithelium whereas the superficial cells contain mucin
droplets.
Right: This is a high magnification image. Note the cytologic features of the basal layers and the sizes of the nuclei
compared to those of the stromal cells beneath the basement membrane.

205
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 9-2 Figure 9-3


HIGH-GRADE ANAL INTRAEPITHELIAL NEOPLASIA ANAL DUCTS/GLANDS
This is a beautiful image to contrast with the transitional These are small tubules that tend to be surrounded by
epithelium in figure 9-1, right. These images were obtained a cuff of lymphoplasmacytic cells akin to a lamina propria
at the same magnification. Compare the nuclear features as in the colon.
and sizes.

essentially a "stopper" that allows the anal canal typically displays prolapse changes (4) . Epithe-
to fully close. lial changes overlying the hemorrhoids can be
Hemorrhoidal vessels are usually above the strikingly reactive and have the appearance of
dentate line on the left lateral, right anterior, "Pagetoid dyskeratosis" (5) which is illustrated
and right posterior aspects of the anal canal. in chapter 10 as a finding in so-called anal tags
Symptomatic hemorrhoids can be found above (fibroepithelial polyps). It is important not to
(internal) or below (external) the dentate line. diagnose this reactive process as human papil-
Hemorrhoids are reported to affect about 4 loma virus- associated viral cytopathic effect.
percent of persons in the United States; most pa- If course, sometimes squamous dysplasia can
tients are between 45 and 65 years of age. While be found in association with hemorrhoids so
there is rio overall gender predilection, symptom- vigilance is always indicated (figs. 9-4, 9-5).
atic hemorrhoids are frequent during pregnancy.
Surgical treatment ofhemorrhoids is suggest- PAPILLARY ENDOTHELIAL HYPERPLASIA
ed for only a subset of patients, including those (VEGETANT INTRAVASCULAR
who: 1) fail medical and nonoperative therapy; H EMANG IOEN DOTH ELIOMA)
2) have symptomatic severe hemorrhoids; 3) Papillary endothelial hyperplasia was first
have symptomatic hemorrhoids in the presence reported in hemorrhoidal vessels, producing a
of a separate anorectal condition that requires gross appearance that reminded Pierre Masson of
surgery; and 4) patient desire (1) . a cherry (6). The condition is wholly benign itself
Microscopically, hemorrhoids demonstrate but the terminology coined over a century ago at
large ectatic vessels, and the associated mucosa the time of the original description is alarming.

206
Non-Neoplastic Anus

Figure 9-4
HEMORRHOIDECTOMY SPECIMEN
Left: Large ectatic blood vessels are present, some stuffed with erythrocytes . This is an internal hemorrhoid sample
because there is both colorectal type mucosa and squamous mucosa. It is important to study such samples carefully; note
the squamous findings indicated by the arrow.
Right: There is high grade squamous dysplasia (anal intraepithelial neoplasia/AIN). This is a high magnification image
of the focus indicated on the left.

Figure 9-5
HEMORRHOIDECTOMY SPECIMEN
Left: This sample is from the anal transitional zone. There is a colorectal type crypt an the left but the top of the image
shows squamous epithelium. A thrombosed hemorrhoidal vessel is seen at the bottom of the image. There is papillary
endothelial hyperplasia in the thrombus towards the top of it.
Right: This is a high magnification of papillary endothelial hyperplasia, a wholly benign finding. Sometimes, however, it
raises the possibility of angiosarcoma . What is now known as papillary endothelial hyperplasia was initially termed vegetant
hemangioendothelioma, a tetm that is no longer used. Hemangioendothelioma is currently used to desGibed a group of vascular neoplasms
that have low grade malignant behavior. In contrast, the lesion depicted is simply a dramatic form of organizing thrombus. Note
that small endothelial cell nuclei form a coating around cores of fibrin and the lesion is restricted to a vessel lumen.

207
Survival Guide to Gastrointestinal Mucosal Biopsies

. .. ..
''
I! -. ·- - ..
~~· ~
. .'•:• ..... ..... ·'
...... -
• •• ; : - :. , ·

':..
--~ ·~·.~
. • • j

•.f - -•
•. .

Figure 9-6
RADIATION PROCTITIS
Left: There is squamous epithelium at the right and colorectal mucosa at the left. The arrow indicates a thickened lamina
propria vessel that is oriented parallel to the surface; the vascular orientation and thickness is typical of the vascular changes
in radiation proctitis. These samples were from a man who had been radiated for prostate carcinoma.
Right: Note the ectatic thick capillaries just under the surface.

This benign reactive process is simply a form else. For all practical purposes, only squamous
of organizing vascular thrombus. It may occur epithelium is affected. It is associated with genital
at any age and involve any vessel, including lesions and penoanal intercourse (7-10). Biopsies
those of neoplasms and vascular malformations. show characteristic multinucleated cells, with
Histologically, the lesion is usually within a smudged intranuclear inclusions in squamous
vessel or area of thrombus and consists of pap- epithelial cells at the edge of ulcers (fig. 9-7).
illary fronds with cores of fibrin or hyalinized
Cytomegalovirus
collagen, lined by a single layer of endothelial
cells. The endothelial cells may have a hobnail Cytomegalovirus (CMV) proctitis is typically
appearance (be plump and protrude a bit) but an infection of HIVI AIDS patients but it should
lack significant atypia or mitotic activity (fig. also be sought in individuals with inflamma-
9-5). Occasionally, the fronds appear to be tory bowel disease taking immunomodulation
"free-floating" within the vessel lumen. Extra- therapy (11). It can manifest as ulcers, polyps,
vascular extension may occasionally be seen, or a mass. The viral cytopathic effect in CMV
although it is usually not extensive. proctitis is identical to that in other sites with
Remember also that radiation proctitis can re- both nuclear and cytoplasmic inclusions that
sult in vascular changes, a possibility to consider enlarge the infected cells (fig. 9-8).
in men with a history of prostate carcinoma and
Syphilis
women with lower genital tract neoplasms such
as cervical carcinoma (fig. 9-6). Syphilis, caused by Treponema pallidum is a
sexually transmitted disease characterized by
INFECTIOUS PROCTITIS primary, secondary, and tertiary stages. The
primary stage features a chancre, a painless firm,
Herpes Simplex Virus
red lesion at the site of inoculation, which heals
He1pes simplex virus (HSV) proctitis has the after 3 to 6 weeks . Of course, in the rectum or
same appearance as HSV infection anywhere anal canal, this lesion is wholly inapparent to

208
Non-Neoplastic Anus

Figure 9-7
ANAL BIOPSY FROM A PATIENT WITH SYPHILIS AND HERPES SIMPLEX VIRUS PROCTITIS
Left: The sample is from the edge of an ulcer. The lymphoplasmacytosis at the left is a feature of syphilis proctitis but the
multinucleated cell at th e center of the image is a clue to consider herpes simplex virus.
Right: Note the multinucleation of th e cell at the upper left. The nuclei have a smudged appearance (ground glass
appearance).

Figure 9-8
CYTOMEGALOVIRUS PROCTITIS
This is a sample from an HIV infected
man who has sex with men (MSM). The
infected cell is an endothelial cells and
it has both cytoplasmic inclusions and
a nuclear inclusion. It is very large
compared to the other endothelial
cells in the image.

209
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 9-9 Figure 9-10


SYPHILIS PROCTITIS WITH CONDYLOMA LATUM SYPHILIS PROCTITIS WITH CONDYLOMA LATUM
At low magnification, the eye-catching feature is This is a high magnification showing the dense plasma
the dense band-like plasmacytic inflammation and the cell rind encircling the vessels.
cauliflower-like appearance of the squamous proliferation.
The appearance if very much like that of a genital wart
(condyloma) but the dense inflammation is very different.
Note that the vessels at the bottom of the image are also
cuffed by plasma cells. tabes dorsalis, and gummas (rubbery nodules
composed of a necrotic center with surrounding
the patient or anyone else. The secondary stage macrophages, fibroblasts, lymphocytes, and
is characterized by a maculopapular rash and plasma cells).
that results from spirochete multiplication with- Endoscopically, syphilitic proctitis may show
in the skin and mucous membranes and occurs granular, friable, erythematous, or ulcerated
2 to 10 weeks after the initial stage. Condyloma mucosa (13-15) or even a mass clinically con-
lata (raised, broad-based plaques) may be seen cerning for malignancy (16,17).
during this stage and may produce lesions that Biopsies show squamous epithelial hyperpla-
clinically masquerade as HPV infection or ma- sia with or without ulceration, granulation tis-
lignancy. Condyloma lata shows acanthosis, sue, and striking, band-like lymphoplasmacytic
surface erosion, broadening and elongation of infiltrate underlying the squamous epithelium
rete ridges, exocytosis and neutrophils in the that is at least focally angiocentric (16). Defin-
most superficial layers, and spongiosis (fig. 9-9) itive diagnosis requires serologic confirmation
(12). Vessels may show endothelial swelling and with rapid plasma reagin (RPR), fluorescent
proliferation and may be encased by plasma treponema! antibody-absorption test (FTA), or
cells (fig. 9-10) (12). immunolabeling (fig. 9-11) but it is important
Tertiary syphilis is now rare since antibiot- to know that a negative immunostain does not
ics are readily available but manifests aortitis, exclude the diagnosis.

210
Non-Neoplastic Anus

Figure 9-11
SYPHILIS IMMUNOSTAIN
Left: When using this preparation in perianal skin and anal orifice biopsies, it is unwise to use DAB (3,3' Diaminobenzidine)
as the indicator of a positive test as this leads to brown staining, which appears similar to normal melanocytic processes. As
such, it is prudent to use AEC (3-Amino-9-Ethylcarbazole) which results in red staining of the organisms. In this case, the
red colored structures are syphilis organisms whereas the delicate brown ones are melanocytic processes.
Right: This is a negative area and the brown structures are melanocytic processes. There are publications that depict such
an appearance and tout a positive immunostain. This image is from the same case as that shown on the left 16 in a focus
that lacked organisms. It is important to realize that a negative immunostain does not exclude an infection with syphilis
and that lymphogranuloma venereum proctitis has essentially identical feature.

Lymphogranuloma Venereum
The diagnosis is established by serology or PCR
Lymphogranuloma venereum (LGV) is caused and the infection is treated with doxycycline.
by Chlamydia trachomatis, serotypes Ll, L2, and The histologic findings in LGV are nonspe-
L3. Other subtypes of this organism can cause cific and often indistinguishable from those of
cervicitis, endometritis, urethritis, and trachoma syphilis proctitis (fig. 9-12) and thus the diag-
(18) . Outbreaks are repmted in men who have sex nosis must be confirmed by laboratory testing.
with men (MSM) (18) . The classically described
Crohn's Disease
presentation of LGV consists of a penile papule
followed by a secondary stage characterized by Patients with Crohn's disease may have ana-
tender inguinal lymphadenitis (Bubos). How- genital skin lesions (anal sldn tags, hemorrhoids,
ever, in MSM patients, the presentation may tumor-like lesions [rare]) (19), anal canal mani-
include rectal discharge and proctitis with a festations (anal fissures, anal ulcers, and anorectal
tertiary stage of anorectal strictures, perirectal strictures), perianal fistulas and abscesses, rectovag-
abscesses, genital elephantiasis, penile defor- inal fistulas, and cancer (20) . However, the risk of
mities, and esthiomene (a chronic ulcer), all of anal cancers is minimally elevated over that of
which mimics inflammatory bowel disease (18). controls if there is indeed an increased risk (21).

211
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 9-12
CHLAMYDIA (LYMPHOGRANULOMA VENEREUM) PROCTITIS
Left: The findings are similar to those seen with syphilis proctitis. The degree of chronic inflammation of out of proportion
to the degree of acute inflammation and the degree of crypt distortion (in the colonic type component of the mucosa).
Right: There is dense chronic inflammation but scant acute inflammation. There are a few neutrophils in the crypt at the
left but abundant lamina propria inflammation.

In patients with ulcerative colitis, involve- chronic inflammation or fissures, fistulas, and
ment of the anal canal is typically nonspecific striking chronic inflammatory infiltrates can be
and unrelated to the inflammatory disease encountered (fig. 9-13). Unfortunately, unless
whereas patients with Crohn disease have anal granulomas are seen, the biopsy features are
involvement about 25 percent of the time if not specific and must be correlated with clini-
they have classic small intestinal disease and up cal ones. Additionally, many colleagues prefer
to 80 percent of the time if they have colonic not to obtain anal canal biopsies since they are
involvement. Biopsies may show patchy, active uncomfortable for the patient.

212
Non-Neoplastic Anus

Figure 9-13
CROHN'S DISEASE PROCTITIS
A: This is seldom biopsies but note that there is dense
inflammation at the right of the image and minimal
inflammation at the left. This biopsy is from the anal
opening since skin structures can be seen at the left. The
inflammation at the right is granulomatous.
B: A granuloma showing a multinucleated giant cell
with numero us nuclei.
C: This granuloma shows multinucleated giant cells but
also epithelioid histiocytes with reniform (kidney-shaped)
nuclei.

REFERENCES

1. Clinical Practice Committee, American Gastro- 3. Hulme-Moir M, Bartolo DC. Hemorrhoids. Gas-
enterological Association. American Gastroenter- troenterol Clin North Am 2001;30:183-97.
ological Association medical position statement: 4. Kaftan SM, Haboubi NY. Histopathological
diagnosis and treatment of hemorrhoids. Gastro- changes in haemorrhoid associated mucosa and
enterology. 2004;126:1461-2. submucosa. IntJ Colorectal Dis 1995;10:15-8.
2. Madoff RD, Fleshman JW, Clinical Practice 5. Val-Bernal JF, Pinto ]. Pagetoid dyskeratosis is
Committee, American Gastroenterological a frequent incidental finding in hemorrhoidal
Association. American Gastroenterological As- disease. Arch Pathol Lab Med 2001;125:1058-62.
sociation technical review on the diagnosis and 6. Masson P. Hemangioendotheliome vegetant in-
treatment of hemorrhoids. Gastroenterology travasculaire. Bull Soc Anat Paris 1923;93:517-23.
2004;126:1463-73.

213
Survival Guide to Gastrointestinal Mucosal Biopsies

7. Evans BA, Kell PD, Bond RA, MacRae KD, Slomka 15. Teitelman M, Wilson ]A, Guy CD. Image of the
M], Brown DW. Predictors of seropositivity to month. Syphilitic proctitis. Clin Gastroenterol
herpes simplex virus type 2 in women. IntJ STD Hepatol 2008;6:xxvi.
AIDS 2003;14:30-6. 16. Gopal P, Shah RB . Primary anal canal syphilis
8. ]acobs E. Anal infections caused by herpes sim- in men: the clinicopathologic spectrum of an
plex virus. Dis Colon Rectum 1976;19:151 -7. easily overlooked diagnosis. Arch Pathol Lab
9. Krone MR, Tabet SR,Paradise M, Wald A, Corey Med 2015;139 :1156-60.
L, Celum CL. Herpes simplex virus shedding 17. Bender Ignacio RA, Koch LL, Dhanireddy S,
among human immunodeficiency virus-negative Charmie Godorrtes B, Lukehart SA, Marrazzo ]M .
men who have sex with men: site and frequency Syphilis? An unusual cause of surgical emergency
of shedding.] Infect Dis 1998;178:978-82. in a human immunodeficiency virus-infected
10. van de Laar M], Termorshuizen F, Slomka M], et man. Open Forum Infect Dis 2015;2:ofv094.
al. Prevalence and correlates of herpes simplex 18. Kapoor S. Re-emergence oflymphogramiloma ve-
virus type 2 infection: evaluation of behavioural nereum. ] Eur Acad Dermatol Venereol2008;22:
risk factors. IntJ Epidemiol 1998;27:127-34. 409-16.
11. Alam I, Shanoon D, Alhamdani A, Boyd A, Grif- 19. Kim BS, Kim HS, Kim MB, Oh CK, Kwon KS.
fiths AP, Baxter ]N. Severe proctitis, perforation, Perianal Crohn's disease with a clenched fist-like
and fatal rectal bleeding secondary to cytomega- appearance.] Eur Acad Dennatol Venereol2009;
lovirus in an immunocompetent patient: report 23:1197-8.
of a case. Surg Today 2007;37:66-9 . 20. Sandborn WJ, Fazio VW, Feagan BG, Hanauer
12. Pandhi RI<, Singh N, Ramam M. Secondary syph- SB, American Gastroenterological Association
ilis: a clinicopathologic study. Int ] Dermatol Clinical Practice C. AGA technical review on
1995;34:240-3. perianal Crohn's disease . Gastroenterology
13. Furman DL, Patel SI<, Arluk GM. Endoscopic and 2003;125:1508-30.
histologic appearance of rectal syphilis. Gastro- 21. Ryan DP, Compton CC, Mayer R]. Carcinoma of
intest Endosc 2008;67:161; commentary 161-2. the anal canal. N Engl] Med. 2000;342:792-800.
14. Nazemi MM, Musher DM, Schell· RF, Milo S.
Syphilitic proctitis in a homosexual. ]AMA
1975;231:389.

214
10 ANAL POLYPS AND NEOPLASMS

Anal neoplastic pathology mostly concerns foci of bridging and tuft formation (figs . 10-1,
squamous carcinoma and its precursors, even 10-2). These tumors express CK5/6, which is
though such tumors are relatively rare. Most helpful in excluding an adenocarcinoma, as
cancers are treated with radiation and chemo- well as express estrogen receptor (ER.
therapy so resections are not very common and
biopsy pathology is key. An important issue POLYPS
in anal pathology is that it is the size, rather
Inflammatory Cloacogenic Polyp
than the depth, that determines the T stage in
anal cancers (T1 is Q cm, T2 is >2 to 5 cm, T3 Inflammatory cloacogenic polyp is a mucosal
is >5 cm, and T4 encompasses invasion into prolapse polyp arising at the anorectal transi-
adjacent organs regardless of the size) (1) . A few tion, thus having both squamous and columnar
skin lesions can be encountered in the skin just mucosa but it is essentially the same process as
adjacent to the anus. solitary rectal ulcer and mucosal prolapse as seen
in chapter 8 (figs. 10-3,10-4) (5,6). Patients com-
HIDRADENOMA PAPILLIFERUM plain of hematochezia, and the polyps generally
This benign apocrine neoplasm arises in the arise on the anterior wall of the anal canal.
perianal skin and vulva and is almost always
Fibroepithelial Polyps
a lesion of women over the age of 30. Most
examples involve the labia majora, perineum, Fibroepithelial polyps are also termed anal
and perianal skin (2- 4) . tags and excising them is generally not per-
Lesions are composed of epithelial-lined cysts formed unless they interfere with personal
in the mid-dermis with intricate papillary in- hygiene. Removal may be painful and, in the
foldings that are formed by fibrovascular cores, case of those associated with Crohn's disease,
lined by two layers of cuboidal epithelium with may result in additional morbidity. They are

Figure 10-1
HIDRADENOMA PAPILLIFERUM
These benign lesions are skin appendage
tumors that arise in the perineum and are
almost always lesions of adult women.
The lesion is well marginated but the
complex low magnification architecture is
concerning for adenocarcinoma.

215
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 10-2
HIDRADENOMA PAPILLIFERUM
Left: These tumors are composed of complex tubules that each have two cell layers (an outer myoepitheliallayer and an
inner epithelial layer).
Right: This high magnification image emphasizes the dual layer of cells encircling each tubule. The tubules are sectioned
longitudinally in this image.

Figure 10-3
INFLAMMATORY
CLOACOGENIC POLYP
This is a mucosal prolapse
polyp at the anal transition zone
so there are both squamous and
columnar components. At this
magnification the replacement
of the lamina propria by strands
of smooth muscle imparts a pink
rather than blue appearance to
the lamina propria .

216
Anal Polyps and Neoplasms

Figure 10-4
INFLAMMATORY CLOACOGENIC POLYP
Left: The colorectal type glands are angulated ("diamond shaped") and separated by strands of smooth muscle. In areas
they herniate beneath th e squamous epithelium.
Right: Note the elongated strands of smooth muscle between colorectal type glands. They seem to mold to one another.

often submitted to the pathologist as though cancers and about 12,900 new cervical cancers
they are hemorrhoids. They are essentially the were estimated for 2016 (7), which suggests a
same as skin tags (acrochordons). very low progression rate for HPV infection. The
Fibroepithelial polyps consist of myxoid or total number of infected persons is believed to
collagenous stroma covered by squamous epi- be on the order of 80 to 110 million (see http:/ I
thelium (fig. 10-5). Some larger ones contain www.cdc.gov/std/hpv/). In human immunode-
atypical but reactive stromal cells There is oc- ficiency virus (HIV)-infected men who have sex
casionally epidermoid metaplasia (squamous with men (MSM), individuals at nearly 40 fold
epithelium with a granular layer in non-skin the risk for anal squamous carcinoma over the
anal mucosa). general population, the data support vaccinat-
ing against HPV as a cost effective policy (8).
ANAL SQUAMOUS Endorsement of HPV vaccination in both young
INTRAEPITHELIAL NEOPLASIA men and young women was offered by the Cen-
Anal squamous intraepithelial neoplastic le- ter for Disease Control (CDC) in 2013 (9).
sions, like those in the uterine cervix, are related The anal canal has a transformation zone
to sexually transmitted human papillomavirus akin to that in the uterine cervix, and this is
(HPV) infections, and these are regarded as the where many precursor lesions are detected .
precursors to invasive anal squamous cell car- The HPV-associated lesions had been classified
cinomas. HPV infections are very common but as anal intraepithelial neoplasia (AIN) I, AIN
only about 8,000 estimated new cases of anal II, and AIN Ill, but now, as for the cervix, most

217
Survival Guide to Gastrointestinal Mucosal Biopsies

. A '
.
~:· ,·

Figure 10-5
ANAL FIBROEPITHELIAL POLYP (ANAL TAG)
A: The surface has a peculiar reactive appearance from
local trauma.
B: Note the unusual reactive appearance of the surface.
This appearance has been termed "Pagetoid dyskeratosis"
when it is found in prolapsed uterine cervical mucosa and
it can mimic squamous dysplasia or Paget disease but it is
a purely reactive finding.
C: The stromal cells within the subepithelial tissue of the
polyp frequently display degenerative atypia.

c
observers prefer to separate low- and high- Immunolabeling with Ki-67 and p16 antibod-
grade lesions, with the AIN II subsumed under ies can be helpful in confirming and grading
high-grade. Increasing degree of loss of nuclear dysplasia but it has limitations (10). Ki-67 is a
stratification and polarity, nuclear pleomor- sensitive marker (when used as validated) for
phism and hyperchromatism (with or without dysplasia in mature squamous epithelium and
keratinization or HPV viral cytopathic changes), is therefore helpful for confirmation of AIN1
and increased mitoses high in the epithelium and condyloma (figs. 10-6A,B, 10-7B,C). Ki-67
are the findings used to grade dysplasia/intra- labeling does not distinguish reparative changes
epithelial neoplasia (figs. 10-6, 10-7). As in the from dysplasia/intraepithelial neoplasia.
uterine cervix, dysplasia may be seen extend- A positive Ki-67 stain, for the purposes of
ing into colonic glands but not penetrating grading dysplasia, can be defined as the pres-
the basement membranes; this should not be ence of a group of at least two strongly labeled
mistaken for carcinoma (fig. 10-8). epithelial nuclei in the upper two-thirds of

218
Anal Polyps and Neoplasms

Figure 10-6
LOW-GRADE ANAL INTRAEPITHELIAL NEOPLASIA
A: The cytologic features are similar to those of a
condyloma except that the lesion is flat and found in the
anal transitional mucosa (the area where the squamous and
columnar epithelium meet). There is koilocytotic atypia at
the surface with large nuclei surrounded by paranuclear
cavities but mitotic activity is found in the basal half of
the epithelium.
B: This is a Ki-67. It shows some collections of nuclei
in the upper half of the epithelium. Of course, this pattern
can also be encountered in reparative conditions, so Ki-67
labeling is interpreted in the ·context of the morphologic
features on hematoxylin and eosin (H&E) staining.
C: This p16 preparation is nonreactive .

.' .
...
..
... .- ·.. . ., fj- : .•
• ~ ,' 1 ¥!' I., I ;

, , ~" 0
.. ,, . . .
c ' • • .. . .. . - •
' ' -.. • '. • c ~ .,

' . . .. ...
... .
t' ,,
. . •I/. • • •

.. . ,
·'• ~ ., ,...
'
0 .• ' .

.. . .
i"l·. '~
,..~ .
.
.i
;M·,.·{:
:,

...: .''
- ,..

..• :. •
. '.:
~ .-.'··~ :·:. 0: · -·~ 1
; ·XG
..·'
(
• 0 : .~ •

p M ... , .. 0 ':·.__·}:

the epithelial thickness. This labeling pattern of the squamous epithelium, with or without
is that is it not useful for separating low- and superficial staining.
high-grade dysplasia/intra-epithelial neoplasia P16 labeling is an excellent marker of high-
and does not separate low-risk from high-risk grade AIN but has only limited value in confirm-
HPV type lesions. ing a diagnosis of low-grade AIN.
Ki-67 labeling is combined with p16 immu- Bowen disease, an eponymous term, indicates
nolabeling: squamous carcinoma in situ at the anal margin
1) absent or discontinuous, patchy nuclear (the outside part that can be seen on clinical
and cytoplasmic staining pattern is a negative examination, in contrast to lesions seen at the
p16 result. transition zone, inside the canal, at the dentate
2) A positive p16 result is diffuse and strong line, which are seen by anoscopy) where it
staining of cells of the basal and parabasal layers presents macroscopically as a brownish plaque.

219
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 10-7 .
HIGH-GRADE ANAL
INTRAEPITHELIAL NEOPLASIA
A: Hyperchromatic nuclei extend to the
surface and the nuclei are oriented with their
long axes perpendicular to the basement
membrane and surface until about two thirds
of the way to the surface, when they become
oriented with their long axes parallel to the
surface (AIN2 of 3). There is a mitosis about
two thirds of the way to the surface on the
upper right of the image. Note that in this type
of dysplasia nucleoli are inconspicuous.
B: This Ki-67 stain labels numerous nuclei.
C: There is prominent labeling with this
p16 antibody.

Bowen's disease is essentially an AIN lesion that with an estimated 8,080 new cases in 2016 and
arises in the perianal skin (11). Bowenoid papu- about 1,080 disease-associated deaths (7). Years
losis was first described in the penis (12) and is ago, anal cancer was regarded as an inflamma-
a clinicopathologic diagnosis. The patients have tion-related neoplasm and treatment was an
multiple papules, each with features of in situ abdominoperineal resection with permanent
carcinoma, but these essentially never progress colostomy. Now most patients have small pro-
to invasive carcinoma and sometimes they cedures and chemoradiation.
resolve. These, too, are HPV-associated lesions. Most tumors arising within the anal canal
distal to the dentate line are keratinizing squa-
ANAL SQUAMOUS CELL CARCINOMA mous cell carcinomas, whereas proximal ones
Carcinoma of the anal canal is uncommon, are nonkeratinizing squamous cell carcinomas.
accounting for only 1 to 2 percent of gastro- The nonkeratinizing subtypes were referred to
intestinal tract cancers in the United States, as transitional cell or cloacogenic carcinomas

220
Anal Polyps and Neoplasms

Figure 10-8
HIGH-GRADE ANAL INTRAEPITHELIAL NEOPLASIA EXTENDING INTO COLORECTAL TYPE GLANDS
Left: This is not an indication of invasive carcinoma.
Right: Note the mitotic activity.

in the past; however, they are now recognized Table 10-1


as variants of squamous cell carcinoma. One HISTORIC CLASSIFICATION OF
type is composed of large cells, and the other ANAL SQUAMOUS CARCINOMA-
is characterized by small cells. Many tumors
show more than one morphologic subtype but Squamous cell (cloacogenic)
Large cell keratinizing
the majority of these neoplasms are diagnosed Large cell nonkeratinizing (transitional)
on small biopsies, which are likely not repre- Basaloid
sentative of the entire tumor morphology. The Giant condyloma (verrucous carcinoma)
bladder and anus share a common embryo logic
' Data from reference 14.
origin, thus giving rise to the similar (i.e., transi-
tional or cloacogenic) morphology. The biology
and prognosis of keratinizing and nonkeratiniz-
ing tumors of the anal canal are essentially the In this classification, nonkeratinizing car-
same, so prior concern about subclassification of cinomas were believed to resemble urothelial
anal tumors is probably not warranted. Tumor carcinomas, and basaloid carcinoma was com-
differentiation, however, should be included in posed of solid sheets of small cells, often with
the report as poorly differentiated neoplasms peripheral palisading (fig. 10-9). Overall, most
are associated with a higher risk of death than anal squamous cell carcinomas have the features
well to moderately differentiated examples of squamous carcinoma in other anatomic sites
(13). It is also important to separate high-grade with striking nuclear alterations with nuclear
neuroendocrine (small cell and large cell) carci- pleomorphism, prominent mitotic activity, and
nomas from squamous cell carcinomas of the scattered squamous intercellular bridges (figs. 10-
anus. Adenocarcinomas of the anal canal are 10-10-12). An exception to the usual epithelial
clinicopathologically like rectal cancer and are changes is the rare verrucous carcinoma, which
treated as such. consists of broad-based squamous papillae that
Anal squamous carcinoma was historically are sheet-like in a sclerotic background (fig. 10-
classified into several types (Table 10-1) (14). 13). Verrucous carcinoma has also been termed

221
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 10-9
SQUAMOUS CELL CARCINOMA, BASALOID TYPE
Left: This type was called "cloacogenic" carcinoma in the past. Some examples can be difficult to diagnose and require
an immunolabeling panel to exclude neuroendocrine (small cell) carcinoma and even mesenchymal tumors.
Right: Squamous cell carcinoma, basaloid type. The individual cells have scant cytoplasm.

Figure 10-10
SQUAMOUS CELL CARCINOMA
Left: The appearance of anal squamous cell carcinoma is similar to that of squamous cell carcinoma in other anatomic sites.
Right: The tumor is eosinophilic and nests of tumor are invested in desmoplastic stroma.

222
Anal Polyps and Neoplasms

Figure 10-11
SQUAMOUS CELL CARCINOMA
A: This example is highly differentiated with a pushing border but the cytologic features are more atypical than those
of so-called verrucous carcinoma.
B: Despite the pushing growth pattern, this example is associated with vascular space invasion as seen at the left of the
image.
C: The vascular invasion in this lesion (same case as that depicted in B) shows "paradoxical maturation in which keratinization
becomes more prominent as the lesion invades. Note the dense keratinization (eosinophilia) at the periphery ofthis lymphatic
space deposit of carcinoma.

223
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 10-12
SQUAMOUS CELL CARCINOMA
Left: This is a subtle example of early (superficial) invasion in high grade dysplasia/intraepithelial neoplasia. The brightly
eosinophilic extension from the bottom center portion of the lesion is the clue that this lesion is invading.
Right: This is a high magnification showing the pocket of paradoxical maturation in a focus of invasion. Note that the
high grade dysplasia from which the invasioh takes off is more atypical than the invasive tongue.

Figure 10-13
VERRUCOUS CARCINOMA
Left: Such lesions are impossible to diagnose on superficial biopsies but the massive thickness and pushing front are the
clues to diagnosis.
Right: Note the bland cytologic features at the base of the lesion.

224
Anal Polyps and Neoplasms

)
'

'I

...
..
'

Figure 10-14
COLORECTAL TYPE ADENOCARCINOMA DIAGNOSED ON AN ANAL BIOPSY
Left: Such tumors have the characteristic of colorectal carcinomas from the rest of the colon . Note the "dirty" necrosis.
Right: A sizable minority of these tumors express CKS/6 as per the internal control (overlying squamous epithelium).
This finding is not relevant to diagnosing these lesions as colorectal type adenocarcinomas.

the "giant condyloma of Buschke-Lbwenstein"


and may involve the external genitalia and PAGET D ISEASE
perianal skin. It resembles a condyloma, but is, if Extramammary Paget disease affects apocrine
anything, cytologically blander with less striking gland-rich sites, including the perianal zone.
viral cytopathic changes. Koilocytes with nuclear It grows slowly, consisting of an erythematous
cavities are not seen. Sometimes lesions that eczematoid plaque that may extend internally
otherwise have the appearance of verrucous car- to the dentate line. It is essentially a sweat type
cinomas display zones of more typical invasive, carcinoma with an intraepithelial growth pat-
well-differentiated squamous cell carcinoma, a tern. Histologically, the squamous epithelium is
feature which should also prompt a diagnosis of infiltrated by large pale cells, some of which may
conventional squamous cell carcinoma. have signet cell features, and some of which
Recognizing the subtypes is not really im- may be large and pink (fig. 10-15). A pitfall is
portant and they have no impact on outcome that when epithelium is crushed, it can be dif-
with the exception of verrucous carcinoma and ficult to separate the Paget cells from atypical
poorly differentiated carcinomas. Providing a di- keratinocytes of AIN, or even from melanoma.
agnosis of invasive carcinoma is all that is clini- Immunohistochemistry can resolve most cases.
cally relevant. Ordinary colorectal carcinoma Anal Paget disease is a lesion with apocrine cell
can also be found on biopsies of the anus and differentiation, so the cells express CAM 5.2,
can express CK5/6 in a subset of cases, which carcinoembryonic antigen (CEA), gross cystic
can be ignored (fig. 10-14). disease fluid protein (GCDFP), and CK7, and

225
Swvival Guide to Gastrointestinal Mucosal Biopsies

Figure 10-15
ANAL/PERIANAL PAGET DISEASE
A: This biopsy was taken from the anal canal so the
granular layer is metaplastic. There are plump intra epithelial
malignant columnar cells. Paget disease is essentially an
intraepithelial carcinoma that differentiated along the lines
of skin appendages.
B: This is a CK7 stain, which labels the malignant cells
but not the squamous epithelium
C: This is a p16 stain. The significance of this labeling
is not clear but this image is included to point out that
immunolabeling is used to confirm interpretations rather
than to make them in the first place. This staining is
irrelevant to evaluation of Paget's disease.

they contain mucin (15). Nonneoplastic so- noma, however, may lack CK20 (16). Therefore,
called Taker cells in skin can also express CK7, the way to determine whether any given case
a feature the pathologist must consider when of anal Paget disease is associated with an inva-
evaluating these stains. Additionally, nonneo- sive cancer is by a careful clinical examination.
plastic Merkel cells can express CK20. Rarely this type of lesion is associated only with
The key issue is to separate cases showing a colorectal adenoma! The "tlue" Paget disease
adenocarcinoma cells in anal squamous mu- cases that are epidermotrophic skin append-
cosa that reflect Pagetoid extension of a nearby age type neoplasms have a proclivity for local
colorectal cancer into squamous epithelium recurrence and can eventually become inva-
(figs . 10-16, 10-17). These lesions lack GCDFP sive. Those that result from epidermotropism
and are often CK20+, CK7-, and CDX2+. Occa- of "ordinary" adenocarcinomas behave as the
sional microsatellite unstable colorectal carci- associated cancers.

226
Anal Polyps and Neop lasms

.-.

-.; . I
~ ~ f
,' yl
' 't) 11J
j ,. ..
,., \\\

..

Figure 10-16
PAGETO ID EXTENSION OF COLORECTAL CARCINOMA INTO ANAL SQUAMOUS EPITHELIUM
A: This pattern is indistinguishable from that of true Paget's disease but accounts for about half of cases of anal intra epithelial
carcinoma. The malignant cells are plump and mucinous.
B: This CDX2 preparation highlights the malignant colorectal carcinoma cells that have spread into the squamous
epithelium.
C: This high magnification shows the cytoplasmic mucin of the malignant cells.
D: An adenomatous precursor is seen in the colorectal mucosa at the right side of the image. In rare cases, no invasive
colorectal carcinoma is seen but colorectal type cells are found in the squamous epithelium in a Pagetoid pattern. In fact
the squamous epithelium at the left harbors intraepithelial malignant cells but, in this case there was also an associated
invasive colorectal adenocarcinoma.

227
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 10-17
PAGETOID EXTENSION OF COLORECTAL CARCINOMA INTO ANAL SQUAMOUS EPITHELIUM
Left: This is an invasive colorectal adenocarcinoma that produced Pagetoid invasion into squamous epithelium.
Right: At first glance, this process has an appearance of squamous dysplasia, but note the cytoplasmic mucin in the cell
in the center.

ADENOCARCINOMA AND ANAL glands (fig. 10-18) . These are called anal gland
GLAND ADENOCARCINOMAS (duct) carcinomas. They are composed of tu-
Adenocarcinomas arising in the anus are usu- bules originating from ducts that open onto
ally of the rectal type and arise in the upper zone the mucosal surface. They lack a luminal in situ
of the anus (which is lined by columnar mucosa). component but some show Pagetoid spread into
These are identical to rectal carcinomas and are the squamous epithelium. They are typically
managed as such. On biopsies, they are also CK7+ and CK20-, as per anal glands and ducts.
identical to rectal adenocarcinomas, and their Anal gland carcinoma lacks immunoreactivity
immunohistochemical profile is identical (CK20+, with CKS/6 and P63 (17). However, some cases
CK7 usually negative; note the pitfall with CKS/6 can express CDX2. Anal duct/gland carcinomas
seen in figure 10-14). Anal adenocarcinomas are have lacked prostate-specific antigen (PSA) and
usually easy to diagnose, although the differen- prostatic acid phosphatase (18) and have also
tial diagnosis is with mucosal prolapse polyps lacked hormone receptor expression and HPV
(inflammatory cloacogenic polyps), which have by in situ hybridization (19). Many have be-
diamond-shaped glands, fibromuscular mucosal haved aggressively (19).
stranding, and bland cytology. A general caveat to remember is that typical
A rare subset of anal canal adenocarcinomas colorectal (and some colorectal neuroendocrine
shows differentiation towards, anal ducts/anal neoplasms) cancer can display PAP expression.

228
Anal Polyps and Neoplasms

Figure 10-18
ANAL DUCT/GLAND CARCINOMA
A: There are numerous malignant tubules in the
subepithelial tissue but no surface component.
..() B: Note the malignant glands beneath the squamous
.I# epithelium.
C: This is a CK7 preparation .

10-19). They may induce overlying squamous


OTHER TUMORS pseudoepitheliomatous hyperplasia that can
mimic invasive squamous cell carcinoma. They
Granular Cell Tumor
express CD68 and S-100 protein.
Most gastrointestinal granular cell tumors
Melanoma and Nevi
arise in the esophagus, followed by the anus.
There is a female predominance and most affect- Melanocytes are normal constituents of the
ed individuals are adults. Granular cell tumors anal canal and anal verge, so both melanomas
generally arise at the anocutaneous junction as and nevi can be encountered. In this location,
a nodule but they are sometimes found within melanomas are more common than nevi and
the anal canal. These infiltrative lesions consist usually affect white adults. However, as is the
of uniform cells with abundant pink granular case with subungual and esophageal lesions,
cytoplasm, just like those of other sites (fig. individuals with pigmented skin can also be

229
Survival Guide to Gastrointestinal Mucosal Biopsies

Figure 10-19
GRANULAR CELL TUMOR
A: The lesion is at the bottom of the field but the eye-
catching feature is the pseudoepitheliomatous hyperplasia
at the top and left of the image, which can mimic squamous
cell carcinoma.
B: The tumor is composed of eosinophilic cells with
ample cytoplasm.
C: This high magnification image highlights the granular
cytoplasmic appearance.

affected. When patients present with a mass nib (22). BRAF mutations are rare in mucosal
or often rectal bleeding, it is often initially melanomas but common in cutaneous ones but
attributed to hemorrhoids (20) . Since anal treatment with vemurafenib has been useful
melanomas often lack pigment, a panel immu- in rare persons harboring such mutations (23).
nohistochemical approach is often required to There are few data on response to PD1-related
exclude carcinomas and lymphomas. Assuring (programmed death 1) targeted immunotherapy
that the lesion is primary is best accomplished (24) in anal melanoma at this writing, but in
by the identification of an in situ component. theory anal melanomas should be amenable to
Anal melanomas often are CD117 /KIT+ on this treatment
immunohistochemistry, which does not always Anal melanomas can be difficult to diagnose
correlate with KIT mutations. Some mucosal if an in situ component is not seen. Melanomas
melanomas (about 20 to 30 percent) do have can be pleomorphic and sarcomatoid as well as
J(]T mutations (21) and can respond to imati- epithelioid (fig. 10-20).

230
Anal Polyps and Neoplasms

Figure 10-20
MELANOMA
A: This highly malignant appearing neoplasm has
spindle cell features and cannot be readily diagnosed as
melanoma on the H&E stain. Usually no in situ component
is detected so immunolabeling is needed to confirm the
diagnosis.
B: Prominent nucleoli are a clue to the diagnosis but
immunolabeling was required.
C: This is an S-100 protein stain.

Melanocytic nevi are rare in this area and the body with collections of bland melanocytes
only sporadic examples are reported in the lit- that show maturation towards the deep aspect
erature. They appear similar to nevi elsewhere in of the lesion (figs. 10-21, 10-22).

231
Survival Guide to Gastrointestinal Mucosal Biopsies

-V
•"'*

Figure 10-21
COMPOUND NEVUS WITH CONGENITAL FEATURES
Left: Anal and perianal nevi are uncommon but occasionally encountered.
Right: Compound nevus with congenital features. Finding the nevus extending along skin appendages (a hair shaft in
this example) suggests that it could be congenital.

Figure 10-22
INTRADERMAL NEVUS WITH CONGENITAL FEATURES
Left: This nevus is highly pigmented.
Right: Intradermal nevus with congenital features. This lesion arose in pigmented perianal skin.

232
Anal Polyps and Neoplasms

REFERENCES

1. Amin M, EdgeS, Greene F, et al. A]CC Cancer the anal canal: analysis of patients from the
Staging Manual, 8th ed. Switzerland: Springer National Cancer Data Base. Dis Colon Rectum
International Publishing; 2017. 2009;52:624-631.
2. Goette DK. Hidradenoma papilliferum. ] Am 14. ]ass], Sobin L. Histological Typing of intestinal tu-
Acad Dermatol 1988;19(Pt 1):133-135. mours, 2nd ed. New York: Springer; 1989:10-11.
3. Handa Y, Yamanaka N, Inagaki H, Tomita Y. Large 15. Goldblum]R, Hart WR. Perianal Paget's disease: a
. ulcerated perianal hidradenoma papilliferum in a histologic and immunohistochemical study of 11
young female. Dermatol Surg 2003;29:790-792. cases with and without associated rectal adeno-
4. Loane ], Kealy WF, Mulcahy G. Perianal hidrad- carcinoma. Am J Surg Pathol 1998;22: 170-179.
enoma papilliferum occurring in a male: a case 16. McGregor DK, Wu TT, Rashid A, Luthra R,
report. Ir] Med Sci 1998;167:26-27. Hamilton SR. Reduced expression of cytokeratin
5. Lobert PF, Appelman HD. Inflammatory cloaco- 20 in colorectal carcinomas with high levels
genic polyp. A unique inflammatory lesion of of microsatellite instability. Am ] Surg Pathol
the anal transitional zone. Am ] Surg Pathol 2004;28:712-718.
1981;5:761-766. 17. Lisovsky M, Patel K, Cymes K, Chase D, Bhuiya
6. Saul SH. Inflammatory cloacogenic polyp: rela- T, Morgenstern N. Immunophenotypic char-
tionship to solitary rectal ulcer syndrome/mu- acterization of anal gland carcinoma: loss of
cosal prolapse and other bowel disorders . Hum p63 and cytokeratin 5/6. Arch Pathol Lab Med
Pathol1987;18:1120-1125. 2007; 131:1304-1311.
7. Siegel RL, Miller KD, ]emal A. Cancer statistics, 18. Ballo MT, Gershenwald ]E, Zagars GK, et al.
2016. CA Cancer] Clin 2016;66:7-30. Sphincter-sparing local excision and adjuvant
8. Deshmukh AA, Chhatwal ], Chiao EY, Nyitray radiation for anal-rectal melanoma.] Clin On col
AG, Das P, Cantor SB. Long-term outcomes of 2002;20) :4555-4558.
adding hpv vaccine to the anal intraepithelial 19. Meriden Z, Montgomery EA. Anal duct carcinoma:
neoplasia treatment regimen in HIV-positive a report of 5 cases. Hum Pathol2012;43:216-220.
men who have sex with men . Clin Infect Dis 20. Felz MW, Winburn GB, Kallab AM, Lee JR. Anal
2015;61:1527 -1535. melanoma: an aggressive malignancy masquer-
9. Advisory Committee on Immunization Prac- acting as hemorrhoids. South MedJ 2001;94:880-
tices. Recommend ed adult immunization 885.
schedule: United States, 2013*. Ann Intern Med 21. Antonescu CR, Busam I<], Francone TD, et al.
2013;158:191-199. L576P KIT mutation in anal melanomas cor-
10. Patil DT, Yang B. Utility of human papilloma virus relates with KIT protein expression and is sensi-
capsid protein L1 and p16 in the assessment and ac- tive to specific kinase inhibition. Int] Cancer
curate classification of anal squamous int:raepithe- 2007;121:257-264.
liallesions. Am] Clin Pathol2015;144:113-121. 22. Carvajal RD, Antonescu CR, Wolchok ]D, et al.
11. Bosman F, Carneiro F, Hruban R, Theise N, eds. KIT as a therapeutic target in metastatic mela-
WHO classification of tumours of the digestive noma. JAMA 2011;305:2327-2334.
system. In: Bosman F, ]affee E, Lakhani S, Ohgaki 23. Martinez-Cadenas C, Bosch N, Penas L, et al.
H, eds. World Health Organization classification Malignant melanoma arising from a perianal
of tumours. Lyon: IARC Press; 2010. fistula and harbouring a BRAF gene mutation: a
12. Wade TR, Kopf AW, Ackerman AB. Bowenoid papu- case report. BMC Cancer 2011;11:343.
losis of the penis. Cancer. 1978;42:1890-1903. 24. Topalian SL, Hodi FS, Brahmer ]R, et al. Safety, activ-
13. Bilimoria KY, Bentrem DJ, Rock CE, Stewart ity, and immune correlates of anti-PD-1 antibody
AI<, Ko CY, Halverson A. Outcomes and prog- in cancer. N Engl] Med 2012;366:2443-2454.
nostic factors for squamous-cell carcinoma of

233
Index*

endoscopic mucosal resection, 36


A
goblet cells, 23
Active chronic colitis, 153 granular cell tumor, 39
ulcerative colitis, 153 intramucosal carcinoma, 31
Acute colitis, 148 leiomyoma, 40
Adenocarcinoma, 74, 128, 189, 228 multilayered epithelium, 21
anal, 228 nondysplastic Barrett's esophagus, 24
colon, 189 squamous cell carcinoma, 37, see also Squamous
gastric, 74 cell carcinoma in Barrett's esophagus
small intestine, 128 treatment issues, 34
Adenoma, 70, 121, 174 Bowen disease, 219
colon, 174 Brunner gland lesions, small intestine, 124
high-grade dysplasia, 176
pseudoinvasion, 178
malignant polyp, 180
c
sessile serrated adenoma, 177 Carcinoid tumors, 83, 132
traditional serrated adenoma, 177 gastric, 83
tubular, 17 4 small bowel, 132
gastric, 70 Celiac disease, 107
intestinal, 73 Chemical gastropathy, 48
pyloric, 73 nonsteroidal anti-inflammatory drug use, 48
small intestine, 121 Crohn's disease, 64, 129, 156, 191
pyloric, 124 and adenocarcinoma, small bowel, 129
tubular/tubulovillous, 121 andcolitis,156,191
Adenomatous polyps, 129 gastric, 64
Ampullary carcinoma, small bowel, 129 Colitis, 148, 191
Anal biopsy, 205 active chronic, 153
hemorrhoids, 205 acute, 148
infectious proctitis, 208 associated neoplasia, 191
normal mucosa, 205 collagenous colitis, 15 7
Anal gland carcinomas, 228 diverticular-associated, 15 7
Anal intraepithelial neoplasia, 206 ischemic, 148
Angiosarcoma, small bowel, 141 lymphocytic colitis, 157
Apoptotic colopathy, 161 radiation, 149
Apoptotic esophageal injury, 14 ulcerative colitis, 153
Autoimmune enteropathy, 111 Colitis-associated neoplasia, 191
Autoimmune gastritis, 55 Collagenous colitis, 15 7
Autoimmune metaplastic atrophic gastritis, 55 Collagenous gastritis, 62
Colon biopsy, 145
active chronic colitis, 153
B
acute colitis, 145
Barrett's esophagus, 21 apoptotic colopathy, 161
adenocarcinoma, 37 collagenous colitis, 15 7
dysplasia, 23 infectious organisms, 148
high-grade dysplasia, 25 ischemic colitis, 148
indefinite for dysplasia, 25 lymphocytic colitis, 157
low-grade dysplasia, 25 normal mucosa, 146

235
Survival Guide to Gastrointestinal Mucosal Biopsies

patterns of disease, 145 G


radiation colitis, 149
Gangliocytic paraganglioma, small bowel, 135
Colorectal cancer, 189
colitis-associated neoplasia, 191 Gastric antral vascular ectasia, 50
juvenile polyps, 192 Gastric cardia, 3
neuroendocrine tumors, 192 Gastric siderosis, 51
Common variable immunodeficiency, small Gastrointestinal stromal tumor, 88, 140
intestine, 110 small bowel, 140
Crohn's disease, anal, 211 stomach, 88
Cytomegalovirus, 61, 101, 148, 208 Giardia Iamblia, small intestine, 102
anus, 208 Giardiasis, small intestine, 102
colon, 148 Goblet cells 23
small intestine, 101 Granular cell tumor, 39, 229
stomach, 61 anus, 229
Cytomegalovirus enteritis, small intestine, 103 esophagus, 39
Cytomegalovirus proctitis, 218 Granuloma, esophagus, 15

D H
Diffuse large B-celllymphoma, colon, 197 Helicobactor pylori, 50, 70, 85
gastric polyps, 70
gastritis, 50
E
mucosa-associated lymphoid tissue lymphomas, 85
Endometriosis, colon, 197 Helicobactor pylori gastritis, 50
Endoscopic mucosal resection, 36 Hemorrhoids, 205
Eosinophilic esophagitis, 5 Herpes simplex proctitis, 208
Eosinophilic gastritis, 61 Hidradenoma papilliferum, anal, 215
Epidermoid metaplasia, esophagus, 18 Human papillomavirus, 38, 217
Erosions, esophagus, 10 in Barrett's esophagus, 38
Esophageal biopsy, 1 Human papillomavirus-associated neoplasia, 217
apoptotic injury, 14 Hyperkeratosis, esophagus, 16
biopsy evaluation, 1 Hyperplastic polyps, 70, 176
epidermoid metaplasia, 18 colon, 176
esophagitis dissecans pattern, 15 gastric, 70
granulomas, 15 Hyperplastic serrated polyps, colon, 183
hyperkeratosis, 16
intraepithelial eosinophils, 5
intraepitheliallymphocytes, 8
reactive epithelial changes, 2 Infectious proctitis, see Proctitis, infectious
ulcers and erosions, 10 Inflammatory cloacogenic polyp, 215
Esophagitis dissecans, 15 Inflammatory fibroid polyp, gastric, 88
Extranodal marginal zone lymphoma, gastric, 85 Intestinal adenoma, 73
Intraepithelial eosinophils, 5
F Intraepitheliallymphocytes, 8
Intramucosal carcinoma, 31
Familial adenomatous polyposis, 69, 126 Iron pill gastritis, 52
and fundic gland polyps, 69 Ischemic colitis, 148
and Peutz-]eghers polyps, 126
Fibroepithelial polyps, anal, 215
Follicular lymphoma, small intestine, 137
Fundic gland polyps, 69 Juvenile polyps, colon, 192

236
Index

L Peutz-Jeghers polyps, 72, 126


gastric, 72
Leiomyoma, esophagus, 40 small intestine, 126
Lichen planus esophagitis, 9 Pit apoptosis, gastric, 60
Lymphangioma, small bowel, 141 Polyps, 69, 167, 215
Lymphocytic colitis, 15 7 anal, 215
Lymphocytic gastritis, 60 fibroepithelial polyps, 215
Lymphogranuloma venereum, 211 inflam matory cloacogenic polyp, 215
Lymphomas, 85, 135 colon, 167
. gastric, 85 hyperplastic polyps, 176
mucosa-associated lymphoid tissue tumors, 85 malignant polyp, 180
small bowel, 135 mucosal prolapse polyps, 169
follicular lymphoma, 13 7 serrated polyps, 183, see also Serrated polyps,
mantle cell lymphoma, 138 colon
T-celllymphoma, 138 tubular adenoma, 17 4
gastric, 69
M adenomas, 70
Malignant polyp, colon, 180 fundic gland polyps, 69
MALT lymphoma, 85 familial adenomatous polyposis associated, 69
Mantle cell lymphoma, small intestine, 138 sporadic, 69
Melanoma, anal, 229 hyperplastic polyps, 70
Mesenchymal tumors, small intestine, 140 juvenile polyp, 71
gastrointestinal stromal tumors, 140 Peutz-Jeghers polyp, 72
Metastatic carcinoma, gastric, 77 small intestine, 121
Mucosa-associated lymphoid tissue tumors, 85 Brunner gland lesions, 124
Mucosal prolapse polyps, colon, 169 pancreatic heterotopia, 124
M)'cobacterium avium, small intestine, 105 Peutz-Jeghers polyp, 126
Mycobacterium avium enteritis, small intestine, 105 pyloric gland adenoma, 124
tubular/tubulovillous adenoma, 121
N Papillary endothelial hyperplasia, anal, 206
Neoplasms, 69, 121, 167, see also individual Portal gastropathy, 49
neoplasms Proctitis, infectious, 208
anal, 217 Crohn's disease, 211
colon, 167 cytomegalovirus, 208
normal colon and carcinoma mimics, 167 herpes simplex virus, 208
small bowel, 121 lymphogranuloma venereum, 211
stomach, 69 syph ilis, 208
Neuroendocrine tumors, 81, 132, 192, see also Pseudomelanosis, 101
individual twnors Pyloric adenoma, 73, 124
colon, 192 Pyloric gland adenoma, small intestinal, 124
gastric, 81
sma ll bowel, 132 R
Nevus, anal, 231 Radiation colitis, 149
Nodular duodenitis, 101 Reflux esophagitis, 3, 5
Nonsteroidal anti-inflammatory drugs, 48 Russell bodies, 64

p s
Paget disease, anal, 225 Sessile serrated adenoma, colon, 177, 183
Pancreatic heterotopia, small intestine, 124 Sessile serrated polyps, colon, 183

237
Swvival Guide to Gastrointestinal Mucosal Biopsies

Serrated polyps, colon, 183 Stongyloides stercora/is, small intestine, 102


hyperplastic polyps, 183 Strongyloidiasis, small intestine, 104
sessile serrated adenoma with cytological ·squamous cell carcinoma, anal, 220
dysplasia, 183 classification, 221
sessile serrated polyps, 183 Squamous cell carcinom'a in Barrett's esophagus, 37
traditional serrated polyps, 183 biopsy findings, 39
Small intestinal biopsy, 95 human papillomavirus, 38
autoimmune enteropathy, 111 precursor lesions, 37
common variable immunodeficiency, 110 Syphilis, anal, 208
infectious organisms, 101
cytomegalovirus enteritis, 103 T
giardiasis, 102
T-celllymphoma, small intestine, 138
Mycobacterium avium enteritis, 105 Traditional serrated adenoma, colon, 177
strongyloidiasis, 104
Traditional serrated polyp, colon, 183
Whipple disease, 106
Trophe1yma whippleii, small intestine, 106
medication-associated injury, 111
Tubular/tubulovillous adenoma, 121, 174
neoplasms, see Neoplasms, small bowel
colon, 174
nodular duodenitis, 101
small intestine, 121
normal mucosa, 96
patterns of injury, 95 u
pseudomelanosis, 101
Spirochetosis, colon, 148 Ulcerative colitis, 153, 191, 212
Squamous intraepithelial neoplasia, anal, 217 Ulcers, esophagus, 10
human papillomavirus, 217
in situ, 219 V
Stomach biopsy, 45 Vascular lesions, small intestine, 141
autoimmune gastritis, 55 angiosarcoma, 141
chemical gastropathy, 48 lymphangioma, 141
Crohn's disease, 64 malformations, 141
eosinophils, 61 Vascular malformations, small bowel, 141
Helicobactor pylori, 50 Vegetant intravascular hemangioendothelioma, 206
inflamed mucosa, 50 Verrucous carcinoma, anal, 221
lymphocytic gastritis, 60
nonsteroidal anti-inflammatory drug use, 48 w
pit apoptosis, 60
Well-differentiated neuroendocrine tumors, 81,
portal gastropathy, 49
132, 192
Russell bodies, 64
colon, 192
type of gastric mucosa, 45
gastric, 81
antral, 45
small bowel, 132
oxyntic, 46
Whipple disease, small intestine, 106
transitional, 47
uninflamed mucosa, 46

238

Potrebbero piacerti anche