Sei sulla pagina 1di 16

Review Article

Duodenal Epithelial Polyps


A Clinicopathologic Review
Katrina Collins, MD; Saverio Ligato, MD

 Context.—Duodenal epithelial polyps are reported in tion based upon immunohistochemical and molecular
1.5% to 3% of individuals referred for upper endoscopy. profiles.
Most duodenal epithelial polyps are asymptomatic and Data Sources.—This review is based on peer-reviewed
nonneoplastic; however, a small subset is neoplastic and literature and the authors’ experiences.
may progress to adenocarcinoma. Recent advances in Conclusions.—In this review we provide an update on
the clinicopathologic, immunohistochemical, and molecu-
immunohistochemical and molecular techniques have
lar features of duodenal epithelial polyps and discuss the
helped further characterize these polyps, shedding light surveillance recommendations and treatment options
on their origin, classification, and risk of progression to available. Particular attention should be placed on
adenocarcinoma. recognizing duodenal adenomas with intestinal, gastric,
Objective.—To provide a comprehensive clinicopatho- and serrated phenotype, as they have an increased risk of
logic review of nonneoplastic and neoplastic duodenal malignant transformation if not completely excised.
epithelial polyps, with particular emphasis on recent (Arch Pathol Lab Med. 2019;143:370–385; doi: 10.5858/
developments in classification schemes and risk stratifica- arpa.2018-0034-RA)

D uodenal epithelial polyps have been reported in


approximately 1.5% to 3.0% of individuals referred
for upper endoscopy. Recent advances in endoscopic
immunophenotypic, and molecular features, and correlation
with clinical outcome.

techniques have increased the detection rate of these polyps NONNEOPLASTIC DUODENAL EPITHELIAL POLYPS
and have allowed removal of lesions up to 2 cm in diameter.1 Nonneoplastic duodenal epithelial polyps include Brunner
Duodenal epithelial polyps can occur as sporadic polyps, gland hyperplastic nodule/polyps (also known as Brunner
usually identified incidentally during upper endoscopy gland hyperplasia), Brunner gland hamartomas, Brunner
performed for other reasons, or in the setting of familial gland cysts, ectopic gastric mucosa, pancreatic heterotopia,
polyposis, in which they are typically multiple in number and hyperplastic polyps, inflammatory polyps, and hamartoma-
often carpet the entire duodenal surface. They may present as tous polyps.
sessile or pedunculated polyps, nodules, excrescences, or
Brunner Gland Hyperplastic Nodules/Polyps and Brunner
subtle abnormalities of the mucosa and can be located either
Gland Hamartomas
in the duodenal bulb, ampullary/periampullary region, or
distal duodenum. Most are benign and associated with peptic The distinction between Brunner gland hyperplastic nod-
injury, duodenitis, or inflammatory lesions; however, a small ules/polyps and Brunner gland hamartomas is arbitrary and
subset is neoplastic with the potential for progression to has been traditionally based mainly on size. Brunner gland
adenocarcinoma. Histologically, duodenal polyps are largely hyperplastic polyps are usually small (0.5 cm–1.5 cm),
divided into nonneoplastic epithelial polyps and neoplastic asymptomatic, and discovered incidentally during upper
epithelial polyps (Table 1). In this article, we review the endoscopy performed for other reasons. Brunner gland
salient clinicopathologic features and treatment options of hamartomas are larger (.2 cm), and when symptomatic,
present as gastrointestinal (GI) hemorrhage or intestinal
duodenal epithelial polyps, and provide an update on their
obstruction, often requiring endoscopic or surgical resection.2–4
classification and risk stratification based on morphologic,
In the past, the terms Brunner gland hyperplasia, Brunner
gland hamartoma, and Brunner gland adenoma have been
Accepted for publication May 18, 2018. used interchangeably causing some confusion in the
Published online October 24, 2018. pathologic classification of these lesions.3,5,6 In fact, prior
From the Department of Pathology & Laboratory Medicine, reports of several large Brunner gland proliferative lesions
Hartford Hospital, Hartford, Connecticut. without definitive evidence of cytologic atypia that were
The authors have no relevant financial interest in the products or
reported as Brunner gland adenomas,5,7 in retrospect,
companies described in this article.
Corresponding author: Katrina Collins, MD, Hartford Hospital, represent examples of Brunner gland hyperplastic polyps
Department of Pathology and Laboratory Medicine, 80 Seymour or Brunner gland hamartomas.2 Brunner gland hyperplastic
Street, Hartford, CT 06102 (email: katrina.collins@hhchealth.org). nodules/polyps and Brunner gland hamartomas are now
370 Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato
Table 1. Histologic Classification of Duodenal adipose tissue and/or smooth muscle intermixed with
Epithelial Polyps multilobulated hyperplastic and sometimes cystically dilated
Brunner glands (Figure 1, C and D).
Category Incidence Immunohistochemically, both lesions are positive for
MUC6 protein (a pyloric gland mucin marker), are negative
Nonneoplastic
for MUC5AC (a foveolar mucin marker), and demonstrate a
Brunner gland hyperplastic nodule/polyp Frequent
very low proliferative activity index by Ki-67/MIB-1.3,4,8
Brunner gland hamartoma Less frequent However, in our experience these 3 markers are not usually
Brunner gland cyst Very rare necessary for the diagnosis of these 2 lesions.
Ectopic gastric mucosa Clinical Significance and Treatment.—As previously
Gastric foveolar metaplasia Frequent mentioned, owing to their larger size, Brunner gland
Gastric heterotopia Less frequent hamartomas may cause GI hemorrhage or intestinal obstruc-
Pancreatic heterotopia Less frequent tion requiring endoscopic and sometimes surgical resection.
Hyperplastic polyp Very rare
Inflammatory polyp Less frequent ‘‘Brunner Gland Adenoma’’
Hamartomatous polyps The concept of Brunner gland adenoma is very contro-
Peutz-Jeghers polyp Rare versial and still a matter of ongoing debate.11 Some authors
Juvenile polyp Rare believe that Brunner gland adenoma is a distinct preneo-
Cowden syndrome polyp Rare plastic lesion with gastric phenotype, nuclear atypia, and
Cronkhite-Canada syndrome Very rare immunohistochemical profile identical to the pyloric gland
adenoma of the stomach.12 Others, however, contend that
Neoplastic there are no well-documented cases of either true glandular
Adenoma, intestinal type dysplasia or carcinoma arising from the proliferation of the
Tubular Less frequent Brunner glands.9 In fact, in reviewing the limited literature
Tubulovillous Less frequent on Brunner gland adenocarcinoma and dysplasia (adeno-
Adenoma, gastric type ma), these authors concluded that the dysplastic or
Pyloric gland adenoma Less frequent neoplastic glandular epithelium in these polyps does not
Foveolar adenoma Very rare originate from Brunner glands, but rather from the ducts or
Serrated adenoma with TSA-like features Rare the surface epithelium overlying the Brunner glands often
Neuroendocrine tumors involved by gastric foveolar metaplasia. Hence, they
Gastrinoma Less frequent
concluded that the term Brunner gland adenoma is mislead-
ing and should be discouraged; instead the term Brunner
Somatostatinoma Rare
gland hyperplasia should be used.9 Although in this review,
Gangliocytic paraganglioma Rare
when we refer to these dysplastic lesions we will use the
Abbreviation: TSA, traditional serrated adenoma. term duodenal pyloric gland adenoma rather than Brunner
gland adenoma, we acknowledge some recent studies, both
believed to represent a morphologic continuum of benign in human and animal models, that have described a new
hyperplastic/proliferative lesions of Brunner glands.8 As class of duodenal adenoma and carcinoma demonstrating
such, the term Brunner gland hyperplasia/hamartoma has dysplastic Brunner glands associated with gastric metapla-
been proposed for describing these nonneoplastic lesions.9 sia.13
Clinical and Endoscopic Findings.—Brunner gland Brunner Gland Cysts
hyperplastic/hamartomatous polyps are benign and are
preferentially located in the duodenal bulb, but may also Brunner gland cysts are very rare polypoid or nodular
extend into the second or third part of the duodenum. lesions of the duodenum characterized by a submucosal
Endoscopically, they may be multiple and present as sessile cystic dilation of the Brunner gland ducts. In the past, they
or pedunculated submucosal nodules and show no sex or have also been referred to as Brunner gland cystadenoma,
mucocele of the Brunner glands, and most recently, Brunner
racial predilection.
gland duct cysts.14 The largest case series reported to date
Etiology and Pathogenesis.—Although the exact etiol-
consists of 25 cases with a mean age of 66.2 years and
ogy is still poorly understood, associations with duodenal
approximately equal sex distribution.15 Endoscopically they
injury secondary to gastric acid hypersecretion, Helicobacter present as single or multiple polypoid/nodular lesions, often
pylori infection, end-stage renal disease, and uremia have detected incidentally in the first or second part of the
been reported.2,10 duodenum. They vary in size between 3 mm and 18 mm,
Histopathology and Immunohistochemistry.—Brunner and owing to their cystic nature, during the biopsy
gland hyperplastic nodules/polyps are characterized by a procedure they may rupture and become flattened.16
lobular proliferation of hyperplastic glands, histologically Histologically, they show unremarkable surface duodenal
indistinguishable from normal Brunner glands. They are mucosa with a unilocular or multinodular cystic dilatation of
predominantly located in the submucosa of the duodenal the Brunner gland ducts immediately underneath the
bulb but can extend through the muscularis mucosa into the muscularis mucosae. The cysts are lined by an undulating
lamina propria (Figure 1, A and B). They can be associated single layer of cytologically bland cuboidal or columnar
with surface villous shortening, gastric foveolar metaplasia, epithelium with clear cytoplasm and basally located nuclei
or peptic injury secondary to peptic duodenitis. The usually without cytologic atypia (Figure 2). However, in
histology of Brunner gland hamartomas is similar to that some cases, a variable papillary architecture involving at
of Brunner gland hyperplastic nodules/polyps, but also least part of the cyst may be identified. Historically, they
shows a minor mesenchymal component composed of were believed to represent benign retention cysts without
Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato 371
Figure 1. A, Brunner gland hyperplastic nodule/polyp. At low power, hyperplastic lobules of proliferating Brunner glands filling the submucosa, and
partially the mucosa, are lined by normal duodenal mucosa. B, At high power, the Brunner glands are composed of benign uniform, mucin-
containing columnar cells. C, Brunner gland hamartoma. At low power, it appears as a multilobular proliferation of Brunner glands (D) separated by
a minor mesenchymal component of smooth muscle fibrous bands and adipose tissue (inset) (hematoxylin-eosin, original magnifications 310 [A],
320 [B, D, and D inset], and 34 [C]).

atypia, developing secondary to local obstruction of the identified in the duodenal bulb. It consists either of gastric
draining duct of the Brunner gland.16 However, some recent foveolar metaplastic cells without oxyntic glands, known as
studies have suggested that at least some of them may gastric foveolar metaplasia, or may be associated with oxyntic
represent an early neoplastic process reminiscent of branch- glands and referred to as gastric heterotopia.2 The endoscopic
duct type intraductal papillary mucinous neoplasms.14,17 In appearance is that of 1 or multiple small (,1.5 cm) patches,
fact, the epithelial lining may demonstrate either a pure nodule(s), or, less commonly, pedunculated lesions (Figure
foveolar (MUC5ACþ) or mixed foveolar/intestinal (MU- 3, A).
C5AC and CDX2þ) phenotype, and some of the cells Etiology and Pathogenesis.—Gastric foveolar metapla-
(especially in the papillary component) may demonstrate sia of the duodenum is generally considered as a reactive/
reparative process usually secondary to acid-peptic injury,
cytologic atypia consistent with a low-grade dysplasia as
duodenal ulcer, H pylori infection, or chronic inflammation.3
supported by expression of S100P (an immunomarker
Gastric heterotopia is indistinguishable from normal oxyntic
consistently expressed in low-grade pancreatic intraepithe- gland mucosa of the stomach and traditionally has been
lial neoplasia, but not in reactive lesions).17 However, considered congenital and benign in nature.
further studies are needed to confirm this hypothesis. Histopathology.—In duodenal gastric foveolar metapla-
Ectopic Gastric Mucosa sia the epithelium consists of gastric foveolar-type columnar
cells containing neutral mucin replacing both absorptive
Clinical and Endoscopic Features.—Ectopic gastric cells and goblet cells of the villi (Figure 3, B). Duodenal
mucosa is a common finding in duodenal biopsies, generally gastric heterotopia consists of oxyntic gastric mucosa with
372 Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato
Histopathology.—Pancreatic heterotopia consists of nor-
mal-appearing pancreatic tissue, and the Von Heinrich
classification recognizes 3 types: type I, the most common,
consists of all 3 pancreatic elements (complete pancreatic
heterotopia) (Figure 3, D); type II contains acini and ducts;
and type III is composed only of ducts and rare acinar cells.23
Gaspar Fuentes et al24 classify ectopic pancreas into 4 types:
type I composed of all pancreatic elements (complete
heterotopia); type II composed of ducts only (canalicular
heterotopia), which when surrounded by smooth muscle is
also referred to as adenomyoma25; type III with only acini
(exocrine heterotopia); and type IV with only clusters of
neuroendocrine cells (endocrine heterotopia).
Clinical Significance and Treatment.—Pancreatic het-
erotopia can undergo pathologic changes similar to those
observed in the pancreas such as pancreatitis, pseudocyst,
abscess formation, and rarely, dysplasia and malignancy.22
Endoscopic or surgical resection is the treatment of choice
when they become large and symptomatic or show evidence
of dysplastic features.
Hyperplastic Polyps
Figure 2. Brunner gland cyst. Cystic dilatations of Brunner glands
immediately underneath the muscularis mucosae. At high power Clinical and Endoscopic Features.—Hyperplastic pol-
(inset), the cyst is lined by a single layer of bland columnar epithelium yps of duodenum are very rare and a total of 16 cases have
without cytologic atypia (hematoxylin-eosin, original magnifications so far been reported.26–30 The largest series consists of 9
310 and 320 [inset]).
cases including 3 male and 6 female patients with a mean
age of 52.2 years (range, 21–72 years).27 Typically, they are
both parietal and chief cells and overlying surface gastric asymptomatic and discovered incidentally and most com-
foveolar metaplastic epithelium (Figure 3, C). The adjacent monly located in the second part of the duodenum. The
duodenal mucosa may be normal or associated with varying endoscopic appearance may reveal a sessile or pedunculated
degrees of inflammation.2 polyp with or without surface ulceration ranging in size
Clinical Significance and Treatment.—Although in between 5 mm and 27 mm. Rarely they may cause
most cases gastric foveolar metaplasia and gastric hetero- symptoms such as anemia or GI bleeding.
topia represent benign lesions, some studies have raised the Etiology and Pathogenesis.—Their pathogenesis has not
possibility that a subset of them may be preneoplastic and been clarified yet; however, most patients with hyperplastic
represent a precursor of adenomas and gastric-type polyps are associated with H pylori infection or other form of
adenocarcinomas.18,19 In fact, in a recent study Matsubara gastroduodenal disease. No evidence of association with
et al20 identified mutations in GNAS and/or KRAS in 55% of serrated polyposis or other chronic inflammatory diseases
cases with duodenal gastric foveolar metaplasia and 28% of has been identified.27
cases with gastric heterotopia. These lesions are usually Histopathology, Immunohistochemistry, and Molecu-
small and asymptomatic; however, when very large they lar Characteristics.—Most duodenal hyperplastic polyps
may cause obstruction or intussusception requiring endo- reported so far in the literature (14 of 16 cases; 87.5%) have
scopic or rarely, surgical excision. the histologic appearance of a microvesicular hyperplastic
polyp of the large bowel, characterized by hyperplastic,
Pancreatic Heterotopia columnar microvesciular mucinous epithelium with luminal
and crypt serration lacking cytologic atypia (Figure 4, A).27
Clinical and Endoscopic Features.—Pancreatic hetero-
However, in a few case reports,28,30 a gastric phenotype with
topia is a developmental abnormality in which normal polypoid hyperplasia of the surface has been described,
pancreatic tissue is found in an abnormal location without raising the possibility that at least some of these lesions may
accompanying anatomic and/or vascular connection to represent examples of inflammatory duodenal polyps with
orthotopic pancreas.21 Pancreatic heterotopia can be ob- gastric foveolar metaplasia rather than hyperplastic polyps.
served in any GI tract organ. It is most common in the Similar to the colorectal hyperplastic polyps, MUC6 is
duodenum but can also be found in the jejunum, liver, expressed only at the crypt bases and MUC5AC on the
biliary tract, and omentum.22 The endoscopic appearance of superficial hyperplastic epithelium. MUC2 is identified only if
pancreatic heterotopia is that of a small nondescriptive goblets cells are present and CDX2 usually shows negativity.
nodule covered by normal duodenal mucosa, often with The latter is particularly helpful in differentiating hyperplastic
central umbilication corresponding to the opening of the duodenal polyps, usually CDX2, from duodenal serrated
main duct. Such lesions when large may become symp- adenomas with traditional serrated adenoma (TSA)–like
tomatic, most commonly presenting with abdominal pain, features, which are usually CDX2þ and have a more
obstruction, intussusception, stricture, or bleeding. aggressive behavior. The proliferative activity index by Ki-
Etiology and Pathogenesis.—Although the pathogene- 67/MIB-1 is low and no p53 overexpression is identified. In
sis is unknown, the most likely hypothesis is that it one study,27 BRAFV600E mutations were described in 2 of 6
originates from embryonic migration of pancreatic buds cases (33%) and KRAS mutations in another 2 cases (33%).27
into the duodenal wall. These findings support the hypothesis that these duodenal
Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato 373
Figure 3. Ectopic gastric mucosa. A, Gross appearance of multiple duodenal nodules (arrow) located in the duodenal bulb. B, Gastric foveolar
metaplasia. Papillary projections lined by gastric foveolar-type epithelium (inset). C, Gastric heterotopia. Low-power view composed of a polypoid
proliferation covered by gastric foveolar metaplastic epithelium. At high power (inset), normal-appearing parietal and oxyntic cells are seen. D,
Pancreatic heterotopia. Normal duodenal mucosa with submucosal pancreatic tissue composed of acini associated with pancreatic ducts and nests
of islet cells, similar to the parenchyma of the normal pancreas (inset) (hematoxylin-eosin, original magnifications 310 [B through D], 320 [B and D
insets], and 340 [C inset]).

polyps may be similar to the microvesicular hyperplastic associated with Crohn disease, ulcerative colitis, or other
polyps of large bowel, which also harbor BRAFV600E or KRAS inflammatory processes such as peptic duodenitis, or rarely
mutations in 70% and in 10% to 15% of cases, respectively.31 with primary immunodeficiency.32 They are usually small
Clinical Significance and Treatment.—Although duo- and asymptomatic; however, when large, they may result in
denal hyperplastic polyps are rare, benign, and usually heme-positive stools, GI bleeding, or recurrent abdominal
asymptomatic, Rosty et al27 have raised the possibility that pain due to intermittent obstruction. The most common
some of them, especially those harboring KRAS mutation, inflammatory polyps of duodenum are the pseudopolyps
may represent a precursor lesion of the duodenal serrated found associated with Crohn disease.
adenoma with TSA-like features. In fact, it has been shown Histopathology.—These lesions are characterized either
that both hyperplastic polyps and duodenal serrated by polypoid proliferations of elongated glands, resembling
adenomas with TSA-like features harbor KRAS mutations hyperplastic polyps of stomach, or polypoid growths of
more frequently (33% and 38% of cases, respectively) than granulation tissue with acute and chronic inflammation of
hyperplastic polyps of the colon (15%). However, this the lamina propria, and ulceration of the surface epithelium
hypothesis needs further validation and until then the risk of (Figure 4, B). These lesions may also show granulomas
recurrence and progression of hyperplastic polyps of when associated with Crohn disease.
duodenum should be considered as uncertain and treated Treatment.—Snare polypectomy for small lesions, endo-
with complete endoscopic resection.27 scopic mucosal resection, or submucosal endoscopic dissec-
tion for larger lesions is the treatment of choice.
Inflammatory Polyps
Clinical and Endoscopic Features.—Nonneoplastic Hamartomatous Polyps
inflammatory polyps are inflammatory proliferations of the Hamartomatous polyps of the duodenum represent a
duodenal mucosa (inflammatory pseudopolyps) usually heterogeneous group of inflammatory polypoid lesions
374 Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato
Figure 4. A, Hyperplastic polyp. At low power, elongated fronds of surface epithelium with a sawtooth appearance and crypts composed of small
crowded mucinous glands. At high power (inset), the hyperplastic epithelium shows microvesicular mucinous cells and rare goblet cells with apical
mucin vacuoles. No cytologic atypia is seen. B, Inflammatory polyp. At low power, a central polypoid epithelial proliferation surrounded on both
sides by ulcerated duodenal mucosa (pseudopolyp). C, Peutz-Jeghers polyp. Lower power: the polyp is characterized by an arborizing network of
smooth muscle bundles surrounded by benign glandular epithelium and normal lamina propria. D, Juvenile polyp. Polypoid mucosa with areas of
surface epithelial erosion showing cystically dilated glands embedded in an expanded lamina propria with areas of dense stroma and vascular
congestion (hematoxylin-eosin, original magnifications 310 [A through D] and 320 [A inset]).

often occurring in the setting of familial polyposis syn- Specific Hamartomatous Duodenal Polyps
dromes33,34 or very rarely as isolated sporadic polyps.2
Peutz-Jeghers Polyps.—Peutz-Jeghers polyposis is an
Clinical and Endoscopic Features.—Syndromic hamar- autosomal dominant syndrome characterized by multiple
tomatous polyps may involve the entire GI tract; however, benign GI hamartomatous polyps, mucocutaneous pigmen-
the duodenum is usually much less commonly involved. tation, and tumors of ovary or testis resulting from
Endoscopically, they may present as small polyps or nodules mutations in the serine-threonine kinase (STK) 11 tumor
or as large lesions that may produce symptoms of intestinal suppressor gene located on chromosome band 19p13.3 in
obstruction or intussusception. approximately 50% of the families with Peutz-Jeghers
Histopathology.—Microscopically, they are usually polyposis.2 Additional genes are yet to be identified. They
characterized by a variable mixture of epithelial and can be located throughout the GI tract and especially in the
stromal elements featuring epithelial hyperplasia, cystic jejunum and duodenum, where they are identified in 78% of
dilation of glands, stromal overgrowth, edema, acute and cases.
chronic inflammation, surface erosion, and Brunner gland Endoscopically, they may appear sessile or pedunculated
hyperplasia. The correct identification and classification of with a smooth multilobulated appearance varying in size
these lesions is critical to the management of these from a few millimeters up to several centimeters. Histolog-
patients because of their association with the develop- ically, they are characterized by branching bands of smooth
ment of GI malignancies as well as other extraintestinal muscle surrounded by glandular epithelium and normal
lesions. lamina propria (Figure 4, C). Sometimes, they may have foci
Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato 375
of adenomatous mucosa and very rarely progress to TSA-like features resembling the TSA of the large bowel.
adenocarcinoma.34 Polypectomy is recommended in order This polyp represents a new distinct category of duodenal
to achieve an accurate diagnosis and because these polyps dysplastic polyp characterized by a significant risk for
have a risk of malignant transformation.35 progression to adenocarcinoma.40,41
Juvenile Polyps.—These polyps may be sporadic or
associated with juvenile polyposis syndrome (JPS), which is Duodenal Adenoma With Intestinal Phenotype
an autosomal dominant syndrome with high penetrance Clinical and Endoscopic Features.—Duodenal adeno-
linked to abnormalities in the signaling of transforming mas with intestinal phenotype account for approximately
growth factor-b (TGF-B) via genetic abnormality of SMAD4 25% of benign neoplasms of the small intestine. They are
or BMPR1AI genes in about 50% to 60% of patients with classified according to their location: ampullary, periampul-
JPS.2 Endoscopically, they typically appear as smooth, lary, or distal to the ampulla. They are also classified
pedunculated, or sessile mucosal lesions that may be clinically into sporadic duodenal adenomas and adenomas
associated with surface ulceration. The histology consists associated with genetic syndromes such as familial adeno-
of cystically dilated glands mixed with a markedly edema- matous polyposis (FAP), attenuated FAP, or MUTYH-
tous/inflamed stroma, erosions of surface epithelium, and associated adenomatous polyposis (MAP). Sporadic duode-
underlying granulation tissue (Figure 4, D). Juvenile nal adenomas account for approximately 40% of all
polyposis syndrome is associated with increased risk of intestinal-type duodenal adenomas and are usually identi-
cancer (~10%), particularly in colon and stomach; however, fied in elderly men (60–80 years of age). Most are
the rarity of these lesions in the duodenum precludes an asymptomatic, usually arising in the second part of the
adequate assessment of their neoplastic risk in this location. duodenum, and endoscopically appearing as sessile or flat
Polyps in Cowden Syndrome.—This syndrome, also rather than pedunculated polyps.19,42 In a study evaluating
known as PTEN (phosphatase and tensin homolog) the risk of adenocarcinoma in nonampullary sporadic
hamartoma tumor syndrome, is a rare autosomal dominant duodenal adenomas, Okada et al43 concluded that lesions
disorder that has been linked to germline mutations in the with low-grade dysplasia and smaller than 20 mm have a
PTEN gene located on chromosome band 10q23.3. It is low risk of progression to adenocarcinoma (4.7%) and some
characterized by multiple hamartomas and hyperplastic risk of progression to high-grade dysplasia (HGD), whereas
lesions of the skin, mucous membranes, brain, thyroid, and adenomas that are larger than 20 mm, or have HGD, have a
entire GI tract.2 In the GI tract, these hamartomatous polyps higher rate of progression to adenocarcinoma (approxi-
may occur in the duodenum more commonly than mately 54.5%). However, most duodenal adenomas with
previously believed (36.5%).36 Endoscopically, they appear intestinal phenotype are found in individuals with FAP
as small (2–5 mm) lesions with a color similar to the (60%) and in approximately 17% to 25% of individuals with
surrounding duodenal mucosa. Histologically, they may MAP. Endoscopically, they present as multiple sessile
present as multiple hamartomatous polyps of differing types polyps with a predilection for the distal duodenum and
featuring juvenile-type polyps, ganglioneuromas, lipomas, periampullary region, and owing to their small size may be
and adenomas.37 If a PTEN mutation is identified, the risk of missed during upper endoscopy. However, with the aid of
adenocarcinoma of breast, thyroid, and endometrium chromoscopic techniques, the number of detected polyps
appears increased; however, an excess risk of GI cancer may increase considerably.
has not been well established. Etiology and Pathogenesis.—Familial adenomatous
Polyps in Cronkhite-Canada Syndrome.—Cronkhite- polyposis is an inherited autosomal dominant syndrome
Canada syndrome is a rare acquired nonfamilial form of due to a germline mutation in 1 copy of the adenomatous
diffuse GI polyposis that typically affects adults in their fifth polyposis coli (APC) gene, a tumor suppressor gene located
and sixth decade of life. These polyps are usually identified on the long arm of chromosome 5 (5q21-22). This condition
in the stomach but may also involve the duodenum.38 In is characterized by the formation of hundreds of intestinal-
isolation, they may be indistinguishable from juvenile or type adenomas in the colon and small bowel with
hyperplastic polyps, and are usually diagnosed in the approximately 65% of people with this condition developing
presence of dramatic GI symptomatology characterized by duodenal adenomas.44 The pathogenesis of both sporadic
weight loss, diarrhea, and nutritional deficiencies.2 Owing to duodenal adenomas and FAP-associated duodenal adeno-
the accompanying findings of hypoalbuminemia, hypocal- mas is not well elucidated; however, it is believed that it may
cemia, anemia, severe electrolyte deficiencies, and severe be analogous to that proposed for colorectal carcinoma in
malabsorption, these patients have a poor prognosis with a which there is progressive accumulation of abnormalities in
mortality rate of 60%. Histologically, these polyps appear the APC pathway. Instead, MAP is an autosomal recessive
similar to juvenile polyps, but in contrast with the classical condition caused by biallelic germline mutations of the
juvenile polyps, the intervening mucosa, both in the MUTYH gene. Carriers of 1 MUTYH gene mutation have a
stomach and duodenum, is also abnormal featuring severe milder clinical presentation and not much is known about
edema, congestion, and inflammation. the impact on the development of duodenal polyps.
Histopathology, Immunohistochemistry, and Molecu-
NEOPLASTIC DUODENAL EPITHELIAL POLYPS lar Characteristics.—Sporadic duodenal adenomas, FAP-
At present, duodenal adenomatous polyps are classified related adenomas, and MAP-related adenomas are mor-
according to the mucin phenotype into intestinal (89.1%) phologically indistinguishable and are characterized by
and gastric type (10.9%). The intestinal-type polyps are adenomatous transformation of the small intestinal epithe-
morphologically subdivided into tubular and tubulovillous lium, similar to that observed in the colonic adenomas. They
adenomas and the gastric-type into pyloric gland adenomas are classified as tubular adenoma, composed of small
and foveolar adenomas.19,39 The most recently described tubular glands lined by eosinophilic absorptive epithelium
neoplastic duodenal polyp is the serrated adenoma with with pseudostratified hyperchromatic nuclei (Figure 5, A),
376 Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato
Figure 5. Duodenal adenoma with intestinal phenotype. Tubular adenoma (A) and tubulovillous adenoma (B) composed of neoplastic epithelium
with crowded columnar cells demonstrating hyperchromatic and pseudostratified nuclei without loss of nuclear polarity. The intestinal phenotype is
confirmed by positivity for CD10 (C) and CDX2 (D) (hematoxylin-eosin, original magnification 320 [A and B]; original magnification 320 [C and D]).

and tubulovillous adenoma demonstrating a villous archi- Clinical Significance and Treatment.—Individuals with
tecture covered by dysplastic epithelium with cytologic FAP-related duodenal adenomas have a 100- to 300-fold
features similar to those described in tubular adenomas increased lifetime risk for developing duodenal or periam-
(Figure 5, B). When they progress to HGD both architectural pullary carcinoma.9 Spigelman et al46 developed a 4-stage
complexity, characterized by distorted and/or cribriform classification (0–IV) system for evaluating the severity of
glands, and cytologic atypia with large hyperchromatic duodenal adenomatosis in individuals with FAP (Table 2),
nuclei with prominent nucleoli and loss of nuclear polarity, and based on the degree of risk of developing carcinoma,
are observed. Immunophenotypically, they are defined by recommendations on the frequency of surveillance and
the expression of CD10 (Figure 5, C), CDX2 (Figure 5, D), treatment options were issued (Table 3).
and/or MUC2 proteins but not MUC5AC and MUC6.
This staging system has been validated by other stud-
However, in a recent study, 76.9% of duodenal adenomas
ies47,48 and stratifies the risk of developing duodenal
with intestinal phenotype demonstrated focal gastric phe-
carcinoma from the number, size, histology, and degree of
notype, which is supported by MUC5AC and MUC6
positivity.19 Emerging evidence shows that both sporadic dysplasia of the polyps. Based on this system, approximately
duodenal adenomas and FAP-related adenomas show
molecular alterations similar to those observed in colorectal
adenoma characterized by frequent genetic alterations, Table 2. Modified Spigelman Score
involving the APC and KRAS genes. DNA mismatch repair Factor 1 Point 2 Points 3 Points
abnormalities and TP53 mutations are identified only rarely
and no BRAF mutations are present.45 Instead, duodenal Polyp number ,4 5–20 .20
cancer developing in MAP-related adenomatous polyps Polyp size, mm 1–4 5–10 .10
demonstrates a specific somatic KRAS mutation variant (c.34 Histology Tubular Tubulovillous Villous
G.T in codon 12), which is found in approximately 64% of Dysplasia Low-grade — High-grade
cases. Abbreviation: —, not applicable.
Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato 377
Table 3. Spigelman Stage: Recommended Duodenal originate from Brunner glands. According to these authors,
Surveillance Frequency when the duodenal mucosa is damaged, a process of
mucosal regeneration ensues in the Brunner glands, giving
Spigelman Total origin to generative cell zones (‘‘neo-G zones’’) with
Stage Points Frequency of Surveillance
proliferation of pluripotent stem cells within Brunner glands
0 0 Every 4 y and ducts.
I 4 Every 2–3 y These cells may differentiate toward the surface, forming
II 5–6 Every 1–3 y areas of gastric foveolar metaplasia or downwards giving
III 7–8 Every 6–12 y rise to Brunner gland hyperplasia or Brunner gland
IV 9–12 Expert surveillance every 3–6 mo hamartoma. According to this hypothesis, metaplastic
Surgical evaluation foveolar epithelium originating from the ‘‘neo-G zone’’ is
Complete mucosectomy or prone to mutation errors leading to the generation of
duodenectomy or Whipple dysplastic polyps, such as pyloric gland adenoma, and
procedure if duodenal papilla is ultimately carcinoma.53,56 In support of this hypothesis is the
involved observation that in all the sites of origin pyloric gland
adenoma is often, but not always, associated with gastric
foveolar metaplasia and/or gastric heterotopia.11,54,57,58 Ad-
70% to 80% of patients with FAP have stage II or III ditionally, in a recent molecular study a significant number
duodenal disease and 20% to 30% have stage I or IV disease. of cases of gastric foveolar metaplasia (55%) and gastric
Although surgery is considered the standard treatment for heterotopia (28%) of the duodenum were found to be
FAP patients with severe polyposis (stage IV), in less associated with GNAS and/or KRAS mutations which,
advanced cases, depending on the size and number of the interestingly, represent the same type of mutations usually
polyps, snare polypectomy or advanced endoscopic tech- identified in the duodenal pyloric gland adenoma and
niques like endoscopic mucosal resection or endoscopic adenocarcinoma with gastric phenotype.20
submucosal dissection may be used.35 Histopathology, Immunohistochemistry, and Molecu-
Because for approximately 4% of individuals with MAP lar Characteristics.—Most duodenal pyloric gland adeno-
their condition may progress to invasive duodenal carcino- mas present as polypoid lesions (Figure 6, A) composed of
ma, surveillance and management recommendations anal- tightly packed pyloric tubules (Figure 6, B) or cystically
ogous to those used for individuals with FAP, using the dilated glands lined by a monolayer of cuboidal to low
Spigelman criteria, have been proposed. columnar cells with basally located nuclei, demonstrating a
As previously mentioned, sporadic duodenal adenomas very subtle atypia and slightly enlarged nucleoli. The
with low-grade dysplasia have a low risk of progression to cytoplasm is abundant, glassy, and slightly eosinophilic
adenocarcinoma and consequently can be closely followed and some of the glands may contain mucinous cells and
up with biopsies. However, HGD lesions and nonampullary clear cells. Occasionally, rare goblets cells or Paneth cells
sporadic duodenal adenomas larger than 40 mm in diameter may be identified; however, they represent most likely
show high risk of progression to adenocarcinoma and residual native intestinal epithelial cells rather than a
therefore should be treated by snare polypectomy, advanced component of pyloric gland adenoma.50,57,58 Other times,
endoscopic techniques, or surgery.43 they may present as submucosal nodules (Figure 6, C) or as
Duodenal Adenomas With Gastric Phenotype flat lesions (Figure 6, D).
When these adenomas progress to low-grade dysplasia,
Duodenal adenomas with gastric phenotype are subclas- the glands become irregularly shaped, with enlarged and
sified into pyloric gland adenoma and foveolar adenoma.19,49 hyperchromatic nuclei, inconspicuous nuclei, and rare
In contrast to the duodenal adenomas with intestinal mitotic figures (Figure 6, C [inset]). Instead, in HGD, the
phenotype, they are preferentially located in the proximal glands are more complex with some cribriform architecture,
duodenum19 and very often have a pedunculated appear- enlarged nuclei with loss of polarity, mild membrane
ance.39 irregularity, nuclear pleomorphism, scattered enlarged
Clinical and Endoscopic Features.—Duodenal pyloric nucleoli, and increased mitotic activity50,53 (Figure 6, D
gland adenoma is a rare neoplasm demonstrating morpho- [inset]). The immunoprofile of duodenal pyloric gland
logic, immunohistochemical, and molecular similarities to adenomas is defined by diffuse expression of pyloric gland
the pyloric gland adenoma of the stomach. Lesions with mucin MUC6 (Figure 6, E) and selective expression of
similar morphology and immunohistochemical phenotype MUC5AC (Figure 6, F) along the superficial gastric foveolar
have also been described in the pancreas, biliary tree, epithelium and only focally in the underlying glands. This
esophagus, and rarely in the rectum; however, their most last finding is useful in differentiating a pyloric gland
frequent localization is in the duodenum and in the adenoma from Brunner gland lesions, especially in small
stomach.50–53 They usually occur in older individuals with biopsy specimens. In fact, MUC5AC shows positivity in the
a mean age of 74 years (range, 52–87 years). Additionally, superficial and focally in the glandular component of pyloric
they may be associated with syndromic polyposis such as gland adenomas but negativity in Brunner glands. In
FAP54 and according to some reports, with Lynch syn- addition, CDX2, CD10, and MUC2 (intestinal markers)
drome.55 Endoscopically, they may present as submucosal usually show negativity and this is helpful in differentiating
nodules, flat lesions, or more often as protruding polypoid duodenal adenomas with pyloric phenotype from those with
lesions varying in size between 5 mm and 28 mm with a intestinal phenotype. An increased proliferative activity
median diameter of 15.3 mm. index by Ki-67/MIB-1 and overexpression for p53 may also
Etiology and Pathogenesis.—The histogenesis of duo- be found especially in those polyps with HGD.50 Adeno-
denal pyloric gland adenomas remains unclear. Kushima et carcinoma arising in these polyps is either of gastric or
al53 was the first to suggest that pyloric gland adenoma may mixed (gastric and intestinal) phenotype as supported by
378 Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato
Figure 6. Duodenal pyloric gland adenoma. A, Exophytic pattern of duodenal pyloric gland adenoma. B, In this example, the polyp is composed of
villous fronds of surface epithelium and tightly packed, variably dilated glands lined by cuboidal cells (inset). C, Duodenal pyloric gland adenoma
with a submucosal/nodular morphology featuring glands with round contours and enlarged/hyperchromatic nuclei consistent with low-grade
dysplasia (inset). D, Flat pyloric gland adenoma with most glands demonstrating round contours focally becoming complex and cribriform with large
nuclei, some prominent nucleoli, and loss of nuclear polarity consistent with high-grade dysplasia (inset). E, Most of the glands are positive for MUC6,
whereas (F) the surface foveolar epithelium and some of the glands are positive for MUC5AC (hematoxylin-eosin, original magnifications 310 [A and
C], 320 [B and D], and 340 [B, C, and D insets]; original magnification 310 [E and F]).

Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato 379
molecular alterations and a more aggressive clinical
behavior.40,41
Clinical and Endoscopic Features.—Review of the
literature has identified approximately 73 serrated adenomas
with TSA-like features involving the upper GI tract
(esophagus [1], stomach [35], duodenum [35], pancreas
[1], and gallbladder [1]) with almost 53.4% demonstrating
HGD or association with adenocarcinoma.41,63 Currently,
approximately 36 serrated adenomas with TSA-like features
have been reported in the duodenum,40,63–66 with the largest
series consisting of 13 patients (12 serrated adenomas and 1
serrated adenocarcinoma) with a median age of 71 years and
a similar male to female ratio.40 Although slow growing,
these polyps have been associated with an aggressive
behavior, with almost 28.6% progressing to adenocarcino-
ma.40 Endoscopically, they appear as papillary lesions
Figure 7. Duodenal adenoma with gastric foveolar phenotype. varying in size from a few millimeters to a few centimeters.
Protruding polyp composed of cells resembling gastric foveolar-type Etiology and Pathogenesis.—The etiology and the
epithelium with nuclear pseudostratification. All tumor cells are positive causes for their aggressive behavior are unknown; however,
for MUC5AC (inset) (hematoxylin-eosin, original magnification 320; some studies have raised the possibility that hyperplastic
original magnification 310 [inset]). Courtesy of Takehiro Mitsuishi, MD. polyps of the duodenum, especially those with KRAS
mutation, may represent a precursor lesion of serrated
adenomas with TSA-like features.27 More recently, Rubio41
maintained coexpression of MUC6, MUC5AC, and CDX2 has also proposed an alternative pathway of carcinogenesis
immunostains.19,59–61 The most frequent molecular abnor- in which CpG island methylation phenotype (CIMP-high)
malities detected in duodenal pyloric gland adenomas would play a pivotal role in the development of these polyps
include mutations in oncogenes KRAS (70%) and GNAS independently of BRAF mutation.
(50%), and less frequently, CTTNB1 and tumor suppressor Histopathology, Immunohistochemistry, and Molecu-
genes SMAD4 and TP53. APC mutations are usually lar Characteristics.—Histologically, they are similar to
absent.58,59 TSAs of the colorectum, featuring a prominent serration in
Clinical Significance and Treatment.—Duodenal pylo- more than 50% of the polyp, ectopic crypt foci, and
ric gland adenomas are neoplastic lesions with a reported predominance of cells with tall eosinophilic cytoplasm and
malignant transformation rate between 12% and 30% of pencillate/pseudostratified nuclei41 (Figure 8, A and B). They
cases. In a series of 19 pyloric gland adenomas of can demonstrate low- or high-grade dysplasia and may
duodenum, Chen et al50 found low-grade dysplasia in progress to adenocarcinoma with serrated morphology63
10.5% of these cases, HGD in 42.1%, and invasive (Figure 8, C and D). Furthermore, several examples with a
carcinoma in 10.5%. Furthermore, Vieth et al51 showed mixed TSA-like and adenomatous morphology have also
focal transition to adenocarcinoma in 30% of cases. It been described.65 Immunohistochemically, they demon-
appears that there is a correlation between size increase of strate an intestinal phenotype with expression of CDX2,
the lesion (20 mm) and occurrence of HGD and abnormal nuclear staining for b-catenin in 23% of cases, and
adenocarcinoma.39 However, regardless of the degree of abnormal p53 expression in 31% of cases. MUC2 expression
atypia and mucin phenotype, if these polyps are completely is observed only in serrated adenomas containing goblets
resected, no cases of recurrence or progression have been cells (58%), and high Ki-67/MIB-1 expression is seen in 62%
of cases with HGD. As previously mentioned, expression of
reported. Instead, in cases where the polyps were not
CDX2 may be clinically useful in distinguishing benign
completely excised, progression to adenocarcinoma with
hyperplastic polyps (CDX2) from potentially aggressive
gastric phenotype has been documented.60
traditional serrated adenomas (CDX2þ).40 The molecular
Duodenal foveolar-type adenomas are very rare le- profile is characterized by KRAS mutation in 38% of cases,
sions,19,39,62 representing approximately 2.7% of duodenal CpG island methylation phenotype (CIMP-high) in 50% of
adenomas with gastric phenotype and have a median tumor cases, and MGMT (O(6)-methylguanine-DNA methyltrans-
diameter of 9.7 mm (range, 8–20 mm). All of these lesions ferase) in 8% of cases. Interestingly, no BRAFV600E mutation
are polypoid and histologically have a tubulovillous or loss of expression of MLH1 was identified in any case.40
architecture with tall columnar epithelium resembling This molecular profile differs from that reported in TSAs of
foveolar gastric epithelium with various degrees of dyspla- the colon, in which BRAF mutation is identified in 67% of
sia19 (Figure 7). Immunohistochemically, they are composed cases and KRAS mutation in 22% of cases.67 These
of MUC5AC-positive (Figure 7 [inset]) cells, rare MUC6- preliminary results suggest that TSAs of duodenum do not
positive cells, and complete absence of intestinal-type mucin develop through the same serrated neoplasia pathway as in
phenotype.19 Owing to their rare occurrence, little is known the large bowel, in which CIMP-high and BRAF mutation
about their molecular abnormalities. evolve concomitantly,68 but rather from an alternative TSA
pathway of carcinogenesis in which CIMP-high represents
Duodenal Serrated Adenoma With TSA-like Features
an early and common molecular event independently of
‘‘Traditional Serrated Adenoma of Duodenum’’
BRAF mutation.40 Summarized in Tables 4 and 5 are the
Duodenal serrated adenoma is a very rare type of main immunohistochemical and molecular features of the
adenoma morphologically resembling serrated adenomas above-discussed clinically significant duodenal epithelial
of colorectum, but reportedly demonstrating different polyps.
380 Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato
Figure 8. Duodenal serrated adenoma with traditional serrated adenoma (TSA)–like features. A, Polyp with elongated fronds demonstrating a
serrated architecture with sawtooth pattern. B, On intermediate power, ectopic crypt formation (arrow) and low-grade pseudostratified nuclei with
eosinophilic cytoplasm resembling a TSA of colon are seen. C, Low-power view of duodenal serrated adenoma with high-grade dysplasia. D, High-
power view highlights the prominent pseudostratification and hyperchromasia of nuclei with loss of nuclear polarity (hematoxylin-eosin, original
magnifications 310 [A and C] and 320 [B and D]).
Figure 9. Duodenal neuroendocrine tumor. A, Submucosal tumor nests composed of tightly packed round cells with a rosette-like architecture
(inset). B, High-power view of another duodenal neuroendocrine tumor composed of small to medium-sized glands with eosinophilic and finely
granular cytoplasm, nuclei with stippled chromatin, and intraluminal microcalcifications. An immunostain for somatostatin (inset) shows positivity,
suggesting the diagnosis of somatostatinoma (hematoxylin-eosin, original magnifications 310 [A], 340 [A inset], and 320 [B]; original magnification
340 [B inset]).

Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato 381
Table 4. Immunohistochemical Profile
MUC6 MUC5AC CD10 CDX2 MUC2
Brunner gland hyperplasia/hamartoma þþ (diffuse)    
Adenoma with pyloric gland phenotype þþ (diffuse) þ (patchy)a   þ (goblet cells)
Adenoma with foveolar gastric phenotype þ/ þþ (diffuse)   
Adenoma with intestinal phenotype   þ þ þ (goblet cells)
Hyperplastic polyps þþ (crypts) þ (surface)   þ (goblet cells)
Serrated adenoma with TSA-like features þþ (crypts) þ/ (surface) þ/ þþ þ (goblet cells)
Abbreviations: TSA, traditional serrated adenoma; þ, positive; , negative.
a
Surface epithelium and focally underlying glands.

Prognosis and Treatment.—Because of their rarity, the an aggressive behavior with lymph node metastasis (5%–
natural history, prognosis, and appropriate clinical man- 10% of cases), especially if functional.72
agement is unclear. However, the high frequency of HGD Histopathology.—Duodenal gastrinoma exhibits rib-
suggests that these serrated adenomas may represent bons, insular, or trabecular patterns of growth, and
aggressive lesions with high malignant potential. Conse- occasional rosetting. The nuclei are usually centrally located
quently, the recommendation is for a complete removal by and demonstrate a fine stippled ‘‘salt and pepper’’
either endoscopic or surgical procedures to rule out the chromatin (Figure 9, A, and inset). Nucleoli are infrequent
possibility of a synchronously growing invasive carcinoma and usually only rare mitoses are observed. The cells are
or to prevent cancer progression. immunoreactive for chromogranin A and synaptophysin is
positive only in about 50% of cases.
NEUROENDOCRINE TUMORS Duodenal somatostatinoma is the second most common
Clinical and Endoscopic Features.—Intestinal neuro- (18%) neuroendocrine tumor of the duodenum and is
endocrine tumors (carcinoids) are most commonly encoun- associated with neurofibromatosis type 1 in one-third of
tered in the ileum,69 with only less than 5% localized in the cases. In contrast to pancreatic somatostatinoma, the
duodenum and ampulla.70 duodenal somatostatinoma is usually nonfunctioning,
They present usually as subepithelial polypoid lesions presents with abdominal pain, nausea, or obstructive
ranging from 1 to 2 cm in diameter. Endoscopically, they jaundice. However, rarely it may be functional and
appear as a white or yellow polyp-like lesion with central associated with the clinical triad of diabetes mellitus,
dimpling or ulceration.71 They are often clinically ‘‘silent’’ cholelithiasis, and steatorrhea.73 Histologically, it demon-
tumors but hormonal expression can be identified by strates a glandular or tubular architecture with intraluminal
immunohistochemistry. Occasionally, they can be func- psammomatous calcifications and can be mistaken for a
tional and associated with clinical syndromes due to metastatic carcinoma. The cells are immunoreactive for
hormone hypersecretion, such as the gastrinoma.71 Based chromogranin A, synaptophysin, and somatostatin74 (Fig-
on the World Health Organization 2010 classification ure 9, B, and inset).
scheme, neuroendocrine tumors of duodenum are classi- Duodenal/ampullary gangliocytic paragangliomas are rare
fied as malignant tumors and graded as low grade (grade (9%) neoplasms also included as neuroendocrine tumors of
1), intermediate grade (grade 2), neuroendocrine carcino- the duodenum. In a review of the literature, Okubo et al75
mas (grade 3), and mixed adenoneuroendocrine carcino- identified 192 cases with an age range from 15 years to 84
mas. The grading system of these neoplasms is based on years (mean, 52.3 years) and a size range from 5.5 to 100
the mitotic rate and Ki-67 labeling index, as described in mm (mean, 25 mm). They may be asymptomatic or present
Table 6.72 with GI bleeding (45.1%), abdominal pain (42.8%), and
Gastrin-producing neuroendocrine tumors (gastrinomas) anemia (14.5%).75 Endoscopically, they may present as
are the most common neuroendocrine tumors of the sessile submucosal nodules or as subepithelial pedunculated
duodenum (62%). They may occur sporadically or in polyps (Figure 10, A). Histologically, they are triphasic
association with multiple neuroendocrine neoplasia type 1 tumors containing a mixture of spindle cells (often positive
(MEN-1) syndrome and are often associated with Zollinger- for S100, chromogranin, and synaptophysin), epithelial cells
Ellison syndrome. They may be multicentric, deeply organized in nests (staining for chromogranin and synapto-
infiltrating and even when small (,1 cm) can demonstrate physin), and ganglion-type cells, which stain for neuro-

Table 5. Molecular Profile


BRAF KRAS GNAS MGMT CIMP-H APC
Ectopic gastric mucosa  þ/ þ   
Adenoma with pyloric gastric phenotype  þþ þþ Unknown Unknown 
Adenoma with foveolar gastric phenotype Unknown Unknown Unknown Unknown Unknown Unknown
Adenoma with intestinal phenotype  þþ  þ/ þ/ þþ
Hyperplastic polyps þ þ Unknown Unknown Unknown Unknown
Serrated adenoma with TSA-like features  þ Unknown þ þþ Unknown
Abbreviations: APC, adenomatous polyposis coli; CIMP-H, CpG island methylator phenotype-high; MGMT, O(6)-methylguanine-DNA
methyltransferase; TSA, traditional serrated adenoma; þ, positive; , negative.
382 Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato
Table 6. World Health Organization 2010 regional lymph nodes in 25% to 50% of cases, mainly
Classification of Neuroendocrine Tumors (NETs) in attributable to the endocrine component of the lesion.76
the Gastrointestinal and Pancreatobiliary Tract Ampullary neuroendocrine tumors appear to have a more
aggressive behavior than nonampullary neuroendocrine
Mitotic Count/ Ki-67 Labeling
Grade 10 HPFs Index, % tumors; in fact they generally are higher-grade tumors with
a clinical presentation often characterized by obstructive
NET, grade 1 ,2 ,3 jaundice due to compression or obstruction of the ampulla.76
NET, grade 2 2–20 3–20 Treatment.—Neuroendocrine tumors up to 2 cm in size
NEC, grade 3 .20 .20 that are limited to the submucosa and without lymph node
Abbreviations: HPF, high-power field; NEC, neuroendocrine carcino- metastasis can be removed endoscopically. In situations of
ma.
multifocality or larger duodenal neuroendocrine tumors,
surgery will be required.
markers such as neurofilament and Hu protein, but also for In conclusion, it is our hope that this review will be a
many neuroendocrine markers75 (Figure 10, B through E). useful resource for the clinician and surgical pathologist in
Although most duodenal gangliocytic paragangliomas the diagnosis, risk stratification, and treatment of common
have a good prognosis, if they involve the muscular wall and relatively uncommon duodenal epithelial polyps en-
and have a size greater than 2 cm, they can metastasize to countered in daily practice.

Figure 10. Duodenal gangliocytic paraganglioma. A, Endoscopic view showing a pedunculated polyp (arrow). B, Low-power microscopic view of
submucosa multinodular lesion. C, Higher-power view: the lesion consists of a spindle cell component (left side) with isolated ganglion cells (arrow)
and clusters of epithelioid cells (right side). D, Immunostaining for S100 shows positivity in the spindle cells. E, Chromogranin highlights the
epithelioid and ganglion cells (arrow) (hematoxylin-eosin, original magnifications 34 [B] and 310 [C]; original magnification 34 [D and E]).
Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato 383
References polyp originating from islands of gastric mucosa. Dig Dis Sci. 1989;34(9):1468–
1. Maruoka D, Matsumura T, Kasamatsu S, et al. Cold polypectomy for 1472.
duodenal adenomas: a prospective clinical trial. Endoscopy. 2017;49(8):776– 33. Liu X, Chen D, Dugum M, Horvath B, Yuan L, Xiao S-Y. Syndromic and
783. sporadic inflammatory/hyperplastic small-bowel polyps: a comparative study.
2. Riddell R, Jain D. Lewin, Weinstein, and Riddell’s Gastrointestinal Pathology Gastroenterol Rep. 2015;3(3):222–227.
and Its Clinical Implications. Vol 2. 2nd ed. Philadelphia, PA: Wolters Kluwer/ 34. Sekino Y, Inamori M, Hirai M, et al. Solitary Peutz-Jeghers type
Lippincott Williams & Wilkins Health; 2014:1331–1335. hamartomatous polyps in the duodenum are not always associated with a low
3. Terada T. Pathologic observations of the duodenum in 615 consecutive risk of cancer: two case reports. J Med Case Rep. 2011;5:240.
duodenal specimens: I, benign lesions. Int J Clin Exp Pathol. 2012;5(1):46–51. 35. Gaspar JP, Stelow EB, Wang AY. Approach to the endoscopic resection of
4. Jung SH, Chung WC, Kim EJ, et al. Evaluation of non-ampullary duodenal duodenal lesions. World J Gastroenterol. 2016;22(2):600–617.
polyps: comparison of non-neoplastic and neoplastic lesions. World J Gastro- 36. Saito K, Nomura E, Sasaki Y, et al. Characteristics of small bowel polyps
enterol. 2010;16(43):5474–5480. detected in Cowden syndrome by capsule endoscopy. Case Rep Gastrointest
5. Gupta V, Gupta P, Jain A. Giant Brunner’s gland adenoma of the duodenal Med. 2015;125:Article 475705.
bulb presenting with ampullary and duodenal obstruction mimicking pancreatic 37. Huber AR, Findeis-Hosey JJ, Whitney-Miller CL. Hereditary gastrointestinal
malignancy. JOP. 2011;12(4):413–419. polyposis syndromes: a review including newly identified syndromes. J Gastroint
6. Gao Y-P, Shu J-S, Zheng W-J. Brunner’s gland adenoma of duodenum: a Dig Syst. 2013;3:155.
case report and literature review. World J Gastroenterol. 2004;10(17):2616–2617. 38. Slavik T, Montgomery EA. Cronkhite–Canada syndrome six decades on: the
7. So CS, Jang HJ, Choi YS, et al. Giant brunner’s gland adenoma of the many faces of an enigmatic disease. J Clin Pathol. 2014;67(10):891–897.
proximal jejunum presenting as iron deficiency anemia and mimicking 39. Hijikata K, Nemoto T, Igarashi Y, Shibuya K. Extra-ampullary duodenal
intussusceptions. Clin Endosc. 2013;46(1):102–105. adenoma: a clinicopathological study. Histopathology. 2017;71(2):200–207.
8. Kim K, Jan SJ, Song HJ, Yu E. Clinicopathologic characteristics and mucin 40. Rosty C, Campbell C, Clendenning M, Bettington M, Buchanan D, Brown I.
expression in Brunner’s gland proliferating lesions. Dig Dis Sci. 2013;58(1):194– Do serrated neoplasms of the small intestine represent a distinct entity:
201. pathological findings and molecular alterations in a series of 13 cases.
9. Odze RD, Goldblum JR. Surgical Pathology of the GI Tract, Liver, Biliary Histopathology. 2015;66(3):333–342.
Tract, and Pancreas. 3rd ed. Philadelphia, PA: Saunders; 2014:582. 41. Rubio C. Traditional serrated adenomas of the upper digestive tract. J Clin
10. Paimela H, Tallgren LG, Stenman S, von Numers H, Scheinin TM. Multiple Pathol. 2016;69(1):1–5.
duodenal polyps in uraemia: a little known clinical entity. Gut. 1984;25(3):259– 42. Lim C-H, Cho Y-S. Nonampullary duodenal adenoma: current understand-
263. ing of its diagnosis, pathogenesis, and clinical management. World J Gastro-
11. Vieth M, Montgomery EA. Some observations on pyloric gland adenoma: enterol. 2016;22(2):853–861.
an uncommon and long ignored entity. J Clin Pathol. 2014;67(10):883–890. 43. Okada K, Fujisaki J, Kasuga A, et al. Sporadic nonampullary duodenal
12. Suda K, Hamada S, Takahashi, et al. A true Brunner’s gland adenoma. adenoma in the natural history of duodenal cancer: a study of follow-up
Endoscopy. 2007;39(suppl 1):E37–E38. surveillance. Am J Gastroenterol. 2011;106(2):357–364.
13. Wang Y, Shi C, Lu Y, Poulin EJ, Franklin JL, Coffey RJ. Loss of Lrig1 leads to 44. Bulow S, Björk J, Christensen IJ, et al. Duodenal adenomatosis in familial
expansion of Brunner glands followed by duodenal adenomas with gastric adenomatous polyposis. Gut. 2004;53(3):381–3816.
metaplasia. Am J Pathol. 2015;185(4):1123–1134. 45. Wagner PL, Chen Y-T, Yantiss R. Immunohistochemical and molecular
14. Pehlivanoglu B, Xue Y, Reid M, et al. ‘‘Brunner gland/duct cysts’’ with features of sporadic and FAP-associated duodenal adenomas of the ampullary
dysplasia: further characterization of a distinctive lesion in the duodenum of and nonampullary mucosa. Am J Surg Pathol. 2008;32(9):1388–1395.
probable precursor nature (abstract No. 828). Mod Pathol. 2018(S2);31:295–296. 46. Spigelman AD, Williams CB, Talbot IC, Domizio P, Phillips RK. Upper
15. Brown IS, Miller GC. Brunner’s gland cyst: a clinicopathological study of 25 gastrointestinal cancer in patients with familial adenomatous polyposis. Lancet.
cases highlighting an underappreciated lesion. Pathology. 2017;49(5):476–478. 1989;2(8666):783–785.
16. Powers M, Sayuk GS, Wang HL. Brunner gland cyst: report of three cases. 47. Saurin JC, Gutknecht C, Napoleon B, et al. Surveillance of duodenal
Int J Clin Exp Pathol. 2008;1(6):536–538. adenomas in familial adenomatous polyposis reveals high cumulative risk of
17. Alpert L, Whitcomb E, Westerhoff M, Hart J, Xiao S-Y. Brunner gland duct advanced disease. J Clin Oncol. 2004:22(3):493–498.
adenoma mimichking pancreatic intraductal papillary mucinous neoplasm 48. Groves CJ, Saunders BP, Spigelman AD, Phillips RKS. Duodenal cancer in
(abstract No. 572). Mod Pathol. 2015;28(S2):145A–146A. patients with familial adenomatous polyposis (FAP): results of a 10 year
18. Rubio CA. Gastric duodenal metaplasia in duodenal adenomas. J Clin prospective study. Gut. 2002;50:636–641.
Pathol. 2007;60(6):661–663. 49. Xue Y, Vanoli A, Balci S, et al. Non-ampullary-duodenal carcinomas:
19. Mitsuishi T, Hamatani S, Hirooka S, et al. Clinicopathological character- clinicopathologic analysis of 47 cases and comparison with ampullary and
istics of duodenal epithelial neoplasms: focus on tumors with a gastric mucin pancreatic adenocarcinomas. Mod Pathol. 2017;30(2):255–266.
phenotype (pyloric gland-type tumors). PLoS One. 2017;12(4):e0174985. 50. Chen ZM, Scudiere JR, Abraham SC, Montgomery E. Pyloric gland
20. Matsubara A, Ogawa R, Suzuki H, et al. Activating GNAS and KRAS adenoma: an entity distinct from gastric foveolar type adenoma. Am J Surg Pathol.
mutations in gastric foveolar metaplasia, gastric heterotopia, and adenocarcino- 2009;33(2):186–193.
ma of the duodenum. Br J Cancer. 2014;112(8):1398–1404. 51. Vieth M, Kushima R, Borchard, Stotle M. Pyloric gland adenoma: a clinic-
21. Armstrong CP, King PM, Dixon JM, Macleod IB. The clinical significance of pathological analysis of 90 cases. Virchows Arch. 2003;442(4):317–321.
heterotopic pancreas in the gastrointestinal tract. Br J Surg. 1981;68(6):384–387. 52. Vieth M, Kushima R, de Jonge JPA, Borchard F, Oellig F, Stolte M. Adenoma
22. Jun S-Y, Son D, Kim M-J, et al. Heterotopic pancreas of the gastrointestinal with gastric differentiation (so-called pyloric gland adenoma) in a heterotopic
tract and associated precursor and cancerous lesions: systemic pathologic studies gastric corpus mucosa in the rectum. Virchows Arch. 2005;446(5):542–545.
of 165 cases. Am J Surg Pathol. 2017;41(6):833–848. 53. Kushima R, Rüthlein HJ, Stolte M, Bamba M, Hattori T, Borcahrd F. ‘Pyloric
23. Von Heinrich H. Ein peitrang zur histrologie des sogen akzessorischen gland-type adenoma’ arising in heterotopic gastric mucosa of the duodenum,
pancreas. Virchows Arch. 1909;198:392–401. with dysplastic progression of the gastric type. Virchows Arch. 1999;435(4):452–
24. Gaspar Fuentes A, Campos Tarrech JM, Fernandez Burgui J, et al. Ectopias 457.
pancreáticas. Rev Esp Enferm Apar Dig. 1973;39(1):255–268. 54. Wood LD, Salaria SN, Cruise MW, Giardiello FM, Montgomery EA. Upper
25. Bromberg SH, Camilo Neto C, Borges AFA, et al. Pancreatic heterotopias: GI tract lesions in familial adenomatous polyposis (FAP) enrichment of pyloric
clinical pathological analysis of 18 patients. Rev Col Bras Cir. 2010;37(6):413– gland adenomas and other gastric and duodenal neoplasms. Am J Surg Pathol.
419. 2014;38(3):389–393.
26. Roche HJ, Carr NJ, Laing H, Bateman AC. Hyperplastic polyps of the 55. Lee SE, Kang SY, Cho J, et al. Pyloric gland adenoma in Lynch syndrome.
duodenum: an unusual histological finding. J Clin Pathol. 2006;59(12):1305– Am J Surg Pathol. 2014;38(6):784–792.
1306. 56. Sakurai T, Sakashita H, Honjo G, Kasyu I, Manabe T. Gastric foveolar
27. Rosty C, Buchanan DD, Walters RJ, et al. Hyperplastic polyp of the metaplasia with dysplastic changes in Brunner gland hyperplasia: possible
duodenum: a report of 9 cases with immunohistochemical and molecular precursor lesions for Brunner gland adenocarcinoma. Am J Surg Pathol. 2005;
findings. Hum Pathol. 2011;42(12):1953–1959. 29(11):1442–1448.
28. Franzin G, Novelli P, Fratton A. Hyperplastic and metaplastic polyps of the 57. Vieth M, Vogel C, Kushima R, Borchard F, Stolte M. Pyloric gland
duodenum. Gastrointest Endosc. 1983;29(2):140–141. adenoma—how to diagnose? Cesk Patol. 2006;42(1):4–7.
29. Sarbia M, Jüttner S, Bettstetter M, Berndt R. Serrated polyps of the 58. Chlumská A, Waloschek T, Mukenšnabl P, Martı́nek P, Kašpı́rková J,
duodenum: three cases with immunohistological and molecular pathological Zámečnı́k M. Pyloric gland adenoma: a histologic, immunohistochemical and
findings. Pathologe. 2013;34(4):347–351. molecular genetic study of 23 cases. Cesk Patol. 2015;51(3):137–143.
30. Katawaratumi H, Tsujimoto T, Nishimura N, et al. A case of lobulated and 59. Hida R, Yamamoto H, Hirahashi M, et al. Duodenal neoplasms of gastric
pedunculated duodenal hyperplasia polyp treated with snare polypectomy. Case phenotype: an immunohistochemical and genetic study with a practical
Rep Gastroenterol. 2011;5(2):404–410. approach to the classification. Am J Surg Pathol. 2017;41(3):343–353.
31. O’Brien MJ, Yang S, Mack C, et al. Comparison of microsatellite instability, 60. Ushiku T, Arnason T, Fukayama M, Lauwers GY. Extra-ampullary duodenal
CpG island methylation phenotype, BRAF and KRAS status in serrated polyps and adenocarcinoma. Am J Surg Pathol. 2014;38(11):1484–1493.
traditional adenomas indicates separate pathways to distinct colorectal carcino- 61. Reid MD, Balci S, Ohike N, Xue Y, et al. Ampullary carcinoma is often of
ma end points. Am J Surg Pathol. 2006;30(12):1491–1501. mixed or hybrid histologic type: an analysis of reproducibility and clinical
32. Remmele W, Hartmann W, von der Laden U, et al. Three other types of relevance of classification as pancreatobiliary versus intestinal in 232 cases. Mod
duodenal polyps: mucosal cysts, focal foveolar hyperplasia, and hyperplastic Pathol. 2016;29(12):1575–1585.

384 Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato
62. Fujisawa T, Horimatsu T, Sakaguchi K, et al. Duodenal adenoma of gastric 69. Maggard MA, O’Connell JB, Ko CY. Updated population-based review of
foveolar phenotype in the second portion of the duodenum. Dig Endosc. 2006: carcinoid tumors. Ann Surg. 2004;240(1):117–122.
18(1):62–66. 70. Modlin IM, Sandor A. An analysis of 8305 cases of carcinoid tumors.
63. Park YK, Jeong WJ, Cheon GJ. Slow-growing early adenocarcinoma arising Cancer. 1997;79(4):813–829.
from traditional serrated adenoma in the duodenum. Case Rep Gastroenterol. 71. Grin A, Streutker CJ. Neuroendocrine tumors of the luminal gastrointestinal
2016;10(2):257–263. tract. Arch Pathol Lab Med. 2015;139(6):750–756.
64. Srivastava A, Rege TA, Kim KM, et al. Duodenal serrated adenomas: 72. Bosman FT, Carneiro F, Hruban RH, Theise ND, eds. WHO Classification of
evidence for serrated carcinogenesis in the proximal small intestine (abstract No. Tumours of the Digestive System. 4th ed. Lyon, France: IARC Press; 2010:102.
705). Mod Pathol. 2000;13:103–106.
World Health Organization Classification of Tumours; vol 3.
65. Taggart M, Rashid A, Estrella J, et al. Serrated polyps of the extracolonic
73. Krejs GJ, Orci LO, Conlin JM, et al. Somatostatinoma syndrome—
gastrointestinal tract. Histologic findings and genetic alterations (abstract No.
biochemical, morphologic and clinical features. N Engl J Med. 1979;301(6):285–
753). Mod Pathol. 2000;13:212–214.
66. Rubio CA. Serrated adenoma of the duodenum. J Clin Pathol. 2004;57(11): 292.
1219–1221. 74. Randle RW, Ahmed S, Newman NA, Clark CJ. Clinical outcomes for
67. Bettington ML, Walker NI, Rosty C, et al. A clinicopathological and neuroendocrine tumors of the duodenum and ampulla of Vater: a population-
molecular analysis of 200 traditional serrated adenomas. Mod Pathol. 2015; based study. J Gastrointest Surg. 2014;18(2):354–362.
28(3):414–427. 75. Okubo Y, Wakayama M, Nemoto T, et al. Literature survey on epidemiology
68. Weisenberger DJ, Siegmund KD, Campan M, et al. CpG island methylator and pathology of gangliocytic paraganglioma. BMC Cancer. 2011;11:187.
phenotype underlies sporadic microsatellite instability and is tightly associated 76. Park HK, Han HS. Duodenal gangliocytic paraganglioma with lymph node
with BRAF mutation in colorectal cancer. Nat Genet. 2006;38(7):787–793. metastasis. Arch Pathol Lab Med. 2016;140(1):94–98.

CAP19 Abstract Program


Submission Deadline Approaching
Abstract and case study submissions to the College of American Pathologists (CAP) 2019 Abstract
Program are now being accepted. Pathologists, laboratory professionals, and researchers in related fields
are encouraged to submit original studies for possible poster presentation at the CAP19 meeting.

Submissions will be accepted until 5 p.m. Central Friday, March 8, 2019. Accepted abstracts and case
studies will appear on the Archives of Pathology & Laboratory Medicine Web site as a Web-only
supplement to the September 2019 issue.

Visit the CAP19 Web site (www.thepathologistsmeeting.org) or the Archives Web site (www.
archivesofpathology.org) for additional abstract program information including a link to the submission
site.

Arch Pathol Lab Med—Vol 143, March 2019 Duodenal Epithelial Polyps: A Review—Collins & Ligato 385

Potrebbero piacerti anche