Ilovepdf Merged

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 54

GIE GUIDELINE

Diagnosis and management of cancer risk in the


gastrointestinal hamartomatous polyposis syndromes:
recommendations from the U.S. Multi-Society Task Force on
Colorectal Cancer
C. Richard Boland,1,* Gregory E. Idos,2,* Carol Durno,3,4 Francis M. Giardiello,5 Joseph C. Anderson,6,7,8
Carol A. Burke,9 Jason A. Dominitz,10,11 Seth Gross,12 Samir Gupta,13,14,15 Brian C. Jacobson,16
Swati G. Patel,17 Aasma Shaukat,18,19 Sapna Syngal,20,21,22 Douglas J. Robertson6,7

San Diego, California, USA; Duarte, California, USA; Toronto, Ontario, Canada; Baltimore, Maryland, USA; White River
Junction, Vermont, USA; Hanover, New Hampshire, USA; Farmington, Connecticut, USA; Cleveland, Ohio, USA; Seattle,
Washington, USA; New York, New York, USA; San Diego, California, USA; La Jolla, California, USA; San Diego, California,
USA; Boston, Massachusetts, USA; Aurora, Colorado, USA; Minneapolis, Minnesota, USA; Boston, Massachusetts, USA

The gastrointestinal hamartomatous polyposis syndromes are rare, autosomal dominant disorders associated
with an increased risk of benign and malignant intestinal and extraintestinal tumors. They include Peutz-
Jeghers syndrome, juvenile polyposis syndrome, the PTEN hamartoma tumor syndrome (including Cowden’s
syndrome and Bannayan-Riley-Ruvalcaba syndrome), and hereditary mixed polyposis syndrome. Diagnoses
are based on clinical criteria and, in some cases, confirmed by demonstrating the presence of a germline path-
ogenic variant. The best understood hamartomatous polyposis syndrome is Peutz-Jeghers syndrome, caused by
germline pathogenic variants in the STK11 gene. The management is focused on prevention of bleeding and
mechanical obstruction of the small bowel by polyps and surveillance of organs at increased risk for cancer.
Juvenile polyposis syndrome is caused by a germline pathogenic variant in either the SMAD4 or BMPR1A genes,
with differing clinical courses. Patients with SMAD4 pathogenic variants may have massive gastric polyposis,
which can result in gastrointestinal bleeding and/or protein-losing gastropathy. Patients with SMAD4 mutations
usually have the simultaneous occurrence of hereditary hemorrhagic telangiectasia (juvenile polyposis
syndrome–hereditary hemorrhagic telangiectasia overlap syndrome) that can result in epistaxis, gastrointestinal
bleeding from mucocutaneous telangiectasias, and arteriovenous malformations. Germline pathogenic variants
in the PTEN gene cause overlapping clinical phenotypes (known as the PTEN hamartoma tumor syndromes),
including Cowden’s syndrome and related disorders that are associated with an increased risk of gastrointes-
tinal and colonic polyposis, colon cancer, and other extraintestinal manifestations and cancers. Due to the rela-
tive rarity of the hamartomatous polyposis syndromes, recommendations for management are based on few
studies. This U.S. Multi-Society Task Force on Colorectal Cancer consensus statement summarizes the clinical
features, assesses the current literature, and provides guidance for diagnosis, assessment, and management of
patients with the hamartomatous polyposis syndromes, with a focus on endoscopic management. (Gastrointest
Endosc 2022;95:1025-47.)

(footnotes appear on last page of article)

The gastrointestinal hamartomatous polyposis syn- data in affected populations focus increasingly on those
dromes are rare, autosomal dominant disorders associ- with defined cancer susceptibility germline pathogenic
ated with an increased risk of benign and malignant variants leading to less heterogeneity in terms of quanti-
intestinal and extraintestinal tumors. Nevertheless, there fying cancer risk.
has been tremendous progress in recent years, both in The U.S. Multi-Society Task Force on Colorectal
understanding the underlying genetics that underpin Cancer (USMSTF) is a group of colorectal cancer (CRC)
these disorders and in elucidating the biology of associ- content experts appointed by the American College
ated premalignant and malignant conditions. Emerging of Gastroenterology, American Gastroenterological

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1025


Gastrointestinal hamartomatous polyposis syndromes

Association, and American Society for Gastrointestinal placed on the risk for gastrointestinal cancer in these
Endoscopy, supplemented at times by other experts to disorders to gain consensus on rational and reasonable
complement existing expertise. In this USMSTF strategies for management once these diseases are diag-
Consensus Statement, the gastrointestinal hamartoma- nosed in a family. The document was approved by the
tous polyposis syndromes were chosen because of recent governing boards of each of the sponsoring gastroenter-
progress in understanding these diseases. The following ology societies.
entities reviewed are: Peutz-Jeghers syndrome (PJS), ju- The USMSTF approach to an adapted use of Grading
venile polyposis syndrome (JPS), PTEN hamartoma tumor of Recommendations Assessment, Development and
syndrome (PHTS, including Cowden’s syndrome [CS] Evaluation (GRADE) has been described previously.1 In
and Bannayan-Riley-Ruvalcaba syndrome [BRRS]), and brief, the GRADE process categorizes the quality of the
hereditary mixed polyposis syndrome (HMPS). Germline evidence as high, moderate, low, or very low on the
alterations are known to cause each of these disorders, basis of the strength of underlying studies, and that
but the diagnosis can also be made on the basis of clinical categorization can be adjusted on the basis of study
criteria. limitations. For example, randomized trials begin as
Although there are essentially no long-term prospec- high-quality evidence and observational studies as low-
tive controlled studies of comparative effectiveness of quality evidence, but their quality may be adjusted up
management strategies for these syndromes, there have or down on the basis of specific study factors. Although
been consensus statements by expert panels that made the GRADE process entails a formal meta-analysis to
management recommendations for these disorders. The assess the quality of evidence for each recommendation,
goal of this USMSTF Statement was to review the litera- the USMSTF employs a modified, qualitative approach
ture focusing on the most recent data, synthesize both for this assessment. The GRADE process separates eval-
the data and the suggested approaches to diagnosis and uation of the quality of the evidence to support a recom-
management by other expert groups, and present the mendation from the strength of that recommendation.
consensus recommendations of the USMSTF (Table 1). This is done in recognition of the fact that, although
Review of summary tables, conference calls, and the quality of the evidence can influence the strength
revisions of iterative drafts, including recommendation of the recommendation, other factors can influence a
statements, were used to reach consensus, at which recommendation, such as adverse effects, patient prefer-
time documents were forwarded to Governing Boards ences, values, and cost. Generally, strong recommenda-
for approval. As our USMSTF is a group of individuals tions mean that most informed patients would choose
with expertise in gastroenterology and gastrointestinal the recommended management. Weak recommenda-
malignancies, we have reserved our management tions mean that patients’ choices will vary according to
recommendations to these areas and defer to other their values and preferences, and clinicians should
expert groups’ recommendations for other cancers ensure that patient care is in keeping with their values
(which are reviewed here). This document therefore and preferences. When the quality of the evidence to
recommends clinical approaches to diagnosing and support a recommendation is low or very low, or if
managing these conditions that affect children and there is a close balance between desirable and undesir-
adults, focuses on cancer risk, and provides insights into able consequences, then usually only a weak recommen-
future research opportunities. dation would be warranted. Weaker recommendations
are indicated by phrases such as “we suggest,” and
stronger recommendations are typically stated as “we
METHODS recommend.”
However, the relative infrequency of, and absence of
A computer-aided PubMed search was performed controlled prospective trials of the interventions (eg, to
from 2000 to 2018, with additional back searches as prevent cancer) in, these syndromes leave all of the rec-
required, and consisted of the following search terms: ommendations without a robust basis of underlying evi-
hamartoma, hamartomatous polyp, hamartoma syn- dence. Thus, all of the interventional recommendations
drome, Peutz-Jeghers syndrome, juvenile polyp, juve- fall (at best) into the “low quality of evidence” GRADE
nile polyposis, Cowden’s syndrome, Cowden’s disease, category, indicating that the true effect of the interven-
PTEN-hamartoma, Bannayan-Riley-Ruvalcaba syn- tions may be markedly different than estimated at this
drome, hyperplastic polyposis, serrated polyposis, and time, and that further research is likely to impact or
hereditary mixed polyposis syndrome. Only English- change our confidence in the effects. As such, this review
language articles were reviewed. Published articles is intended to establish a starting point for future research
were selected on the basis of relevance to the diagnosis into the care of patients with the hamartomatous polypo-
or clinical management of these diseases. Emphasis was sis syndromes.

1026 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

TABLE 1. Questions and recommendations of best practice


Which individuals with hamartomatous polyps should be referred for genetic evaluation?
We recommend patients with any of the following undergo a genetic evaluation: 2 or more lifetime hamartomatous polyps, a family history of
hamartomatous polyps, or a cancer associated with a hamartomatous polyposis syndrome in first or second-degree relatives. Genetic testing (if
indicated) should be performed using a multigene panel test. (Strong recommendation, low quality of evidence)
Peutz-Jeghers syndrome
Who should undergo a genetic evaluation for Peutz-Jeghers syndrome?
We recommend genetic evaluation for any individual with the following: 1) 2 or more histologically confirmed Peutz-Jeghers polyps, 2) any
number of Peutz-Jeghers polyps in an individual who has a family history of Peutz-Jeghers syndrome in a first-degree relative, 3)
characteristic mucocutaneous pigmentation in a person with a family history of Peutz-Jeghers syndrome, 4) any number of Peutz-Jeghers
polyps in a person with the characteristic mucocutaneous pigmentation of Peutz-Jeghers syndrome. (Strong recommendation, low quality of
evidence)
Which organs should undergo surveillance when caring for a patient with Peutz-Jeghers syndrome?
Patients with Peutz-Jeghers syndrome are at increased risk for cancer in multiple organs including cancer of the breast, small bowel, colon,
stomach, pancreas, ovaries, testes, and lungs.
Given this risk, we recommend a multidisciplinary approach to cancer surveillance in these organs (Strong recommendation, low quality of
evidence)
How and when should small bowel surveillance be performed in Peutz-Jeghers syndrome?
We recommend that baseline small bowel surveillance using video capsule endoscopy or magnetic resonance enterography be performed
between ages 8-10 years or earlier if the patient is symptomatic. If no polyps are found at the initial examination, surveillance should resume
at age 18. Because of the risk of small bowel intussusception, small bowel surveillance in adulthood is recommended to continue throughout
life every 2-3 years. (Strong recommendation, low quality of evidence)
What is the recommended approach to endoscopic surveillance of the colon, stomach, and duodenum in Peutz-Jeghers syndrome?
We suggest a baseline upper gastrointestinal endoscopy between the ages of 8 and 10 years, which could be performed at the time of capsule
placement for small bowel surveillance or if polyps are identified on magnetic resonance enterography. Although the initiation age for
colonoscopy remains uncertain, we also suggest initiation of colonoscopy at same time as esophagogastroduodenoscopy. In those in whom
characteristic polyps are detected, both colonoscopy and esophagogastroduodenoscopy should be repeated every 2–3 years. In those in
whom there are no Peutz-Jeghers polyps at baseline, surveillance is repeated at age 18 years, or sooner should symptoms arise, and then
every 3 years. (Weak recommendation, very low quality of evidence)
What size polyps found on small bowel imaging in Peutz-Jeghers syndrome should be removed?
We recommend polypectomy of small bowel polyps that are symptomatic or 10 mm to prevent intussusception and other complications, such
as bleeding. (Strong recommendation, low quality of evidence)
What is the recommended pancreatic cancer surveillance in Peutz-Jeghers syndrome?
We suggest annual pancreatic cancer surveillance with either magnetic resonance cholangiopancreatography or endoscopic ultrasound starting
at age 35 years. (Weak recommendation, low quality of evidence)
Juvenile Polyposis Syndrome
Who should undergo a genetic evaluation for juvenile polyposis syndrome?
We recommend genetic evaluation for any individual with 1) 5 or more juvenile polyps of the colon or rectum; or 2) 2 or more juvenile polyps in
other parts of the gastrointestinal tract; or (3) any number of juvenile polyps and 1 or more first-degree relatives with juvenile polyposis
syndrome. (Strong recommendation, low quality of evidence)
Which organs should undergo surveillance when caring for a patient with juvenile polyposis syndrome?
Juvenile polyposis syndrome patients are at increased risk for cancer in multiple organs including cancer of the colon and stomach.
Given this risk, we recommend patients with juvenile polyposis syndrome undergo surveillance of the colon and stomach. (Strong
recommendation, low quality of evidence)
At what age should colonoscopic and upper endoscopic surveillance begin in individuals identified with juvenile polyposis syndrome?
We suggest initiating colonoscopic and upper endoscopic surveillance at age 12–15 years, or earlier if symptomatic. Surveillance should be
repeated every 1–3 years depending on polyp burden. (Weak recommendation, low quality of evidence)
Which patients with juvenile polyposis syndrome should undergo screening for hereditary hemorrhagic telangiectasia?
We suggest patients with SMAD4 pathogenic variants be clinically evaluated for HHT at the time of the diagnosis, including screening for and
appropriate management of cerebral and pulmonary AVMs. (Weak recommendation, low quality of evidence)
PTEN hamartoma tumor syndrome
Which gastrointestinal findings should prompt a genetic evaluation for PTEN hamartoma tumor syndrome?
We recommend individuals with multiple gastrointestinal hamartomas or ganglioneuromas undergo genetic evaluation for Cowden’s syndrome
and related conditions. (Strong recommendation, low quality of evidence)
(continued on the next page)

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1027


Gastrointestinal hamartomatous polyposis syndromes

TABLE 1. Continued

Which organs should undergo surveillance for cancer when caring for a patient with PTEN hamartoma tumor syndrome?
In PTEN hamartoma tumor syndrome, patients are at increased risk for cancer in multiple organs, including cancer of the breast, thyroid, kidney,
uterus, colon, and skin.
Given this risk, we recommend a multi-disciplinary approach to cancer surveillance in these organs. (Strong recommendation, low quality of
evidence)
What is the recommended colonoscopic surveillance in individuals identified with PTEN hamartoma tumor syndrome?
We suggest colonoscopy surveillance to begin at age 35 years (or 10 years younger than age of any relative with colorectal cancer), repeated at
intervals no greater than 5 years, depending on polyp burden. (Weak recommendation, low quality of evidence)
NOTE. Specific circumstances may merit modification of the recommendations. In cases where very-early-onset cancers develop, the above statements may be modified to start
surveillance 10 years earlier than the youngest cancer diagnosis in the family.

CANCER FAMILY HISTORY ASSESSMENT AND Genetic counseling is a key component to hereditary
REFERRAL FOR GENETIC TESTING IN cancer risk assessment. The purpose of genetic coun-
GASTROENTEROLOGY PRACTICE seling is to educate individuals about the genetic and bio-
logic factors that are related to a patient’s cancer
Question: Which individuals with hamartomatous diagnosis or risk of disease. Counseling helps an individ-
polyps should be referred for genetic evaluation? ual understand the relevant genetic, medical, and psycho-
Recommendation: We recommend patients with social information to make informed decisions about their
any of the following undergo a genetic evaluation: 2 health care. This includes reviewing and expanding the
or more lifetime hamartomatous polyps, a family history following: family history information, elements of genetic
of hamartomatous polyps, or a cancer associated with a testing, tailored cancer risks associated with a pathogenic
hamartomatous polyposis syndrome in first- or second- variant, impact on medical management, reproductive is-
degree relatives. Genetic testing (if indicated) should be sues and options, confidentiality of results, risks with ge-
performed using a multigene panel test. (Strong recom- netic discrimination, potential significance of test results
mendation, low quality of evidence) for other family members, and other pertinent topics.
Practice guidelines from the American College of Medical
Hereditary cancer syndromes account for approximately Genetics and Genomics and National Society of Genetic
5%–10% of new cancer diagnoses and many of the cancers Counselors are available for details regarding the ele-
that arise in families with undiagnosed hereditary cancer ments and process of genetic counseling.3 Although
syndromes are preventable. The identification of individ- traditional models of genetic evaluation and testing
uals with a hereditary gastrointestinal cancer syndrome re- included a certified genetic counselor, alternative
quires a thorough evaluation of the patient’s personal and models exist and are emerging that include provisions
family history of cancer. The collection and assessment of for pretest counseling to be provided by physicians and
family cancer history is a valuable tool for cancer intercep- other health care providers in order to deal with the
tion and prevention and can be critical in the identification increasing demand for genetic testing. If a patient is
of genetic susceptibility. An accurate family history is one found to be a carrier of a germline pathogenic variant, or
that collects the following information: 1) type of cancer, the results are ambiguous due to the finding of a variant
2) age at diagnosis of each primary cancer, 3) lineage of uncertain significance, the help of a genetics provider
(maternal or paternal), 4) ethnicity (people of some ethnic- for post-test counseling and education is recommended.
ities, such as those with Ashkenazi Jewish ancestry, are at In the current era, the vast majority of genetic testing for in-
greater risk for certain cancers), and 5) results of any pre- herited cancer risk predisposition is performed using a
vious cancer-related genetic testing.2 multigene panel testing approach.4 Some patients or
Features of a patient’s personal history and clinical char- families may elect to decline genetic testing due to
acteristics may suggest an inherited susceptibility to can- concerns about risk to confidentiality and insurance; in
cer. Although it is not rare to identify individuals with an these cases, surveillance may still be indicated in the
isolated hamartomatous polyp (particularly an isolated presence of a concerning clinical and/or family history.
juvenile polyp), other features may prompt further evalua- If a germline pathogenic variant is identified, other fam-
tion for an underlying hereditary syndrome. Features asso- ily members should be offered testing for clarification of
ciated with the hamartomatous polyposis syndromes are their own risk. This testing may facilitate initiation of
outlined in detail in this document and include early age screening for associated cancers before symptomatic man-
at cancer onset, multiple cancers in close relatives, unusual ifestations occur and reduce the morbidity and mortality
numbers of hamartomatous polyps, or associated dermato- associated with the syndrome. For example, early small
logic findings. Genetic evaluation may include genetic bowel surveillance may find a polyp that could be removed
counseling and/or genetic testing. from a child with a pathogenic variant in STK11 before

1028 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

leading to intussusception. It is important to recognize that PEUTZ-JEGHERS SYNDROME


genetic testing may not identify pathogenic variants in
every family suspected of a hereditary syndrome. However, Question: Who should undergo a genetic evaluation
there may be clinical features in the family history that sug- for Peutz-Jeghers syndrome?
gest a familial predisposition to cancer and suggest more Recommendation: We recommend genetic evalua-
intensive surveillance recommendations. Referral to Cen- tion for any individual with the following: 1) 2 or more
ters of Excellence might be particularly helpful when ge- histologically confirmed Peutz-Jeghers polyps, 2) any
netic testing results are ambiguous in the setting of number of Peutz-Jeghers polyps in an individual who
suspicious features and prophylactic surgery is being has a family history of Peutz-Jeghers syndrome in a
considered. Lastly, in the era of multigene panel testing, first-degree relative, 3) characteristic mucocutaneous
there may be a scenario in which a germline variant is pigmentation in a person with a family history of
found incidentally associated with an unsuspected syn- Peutz-Jeghers syndrome, and 4) any number of Peutz-
drome. In these cases, patients may be eligible for cancer Jeghers polyps in a person with the characteristic muco-
screening and surveillance as outlined. However, pheno- cutaneous pigmentation of Peutz-Jeghers syndrome.
type and cancer risk compared with patients with classic fa- (Strong recommendation, low quality of evidence)
milial features are not established and are areas of active
research. Enlisting the assistance of a genetic specialist
may be particularly helpful in interpreting ambiguous re- Clinical features
sults and providing management recommendations in PJS was the first hamartomatous polyposis syndrome
these cases. described, by Peutz in Holland in 1921 and by Jeghers,
When children are identified with a hamartomatous pol- McKusick, and Katz in the United States in 1949.9 The
yposis syndrome, their transition of care to adulthood clinical recognition of PJS was facilitated by the
for cancer surveillance is a unique aspect that bears consid- characteristic mucocutaneous freckling around the mouth
eration. It is imperative to transition adolescents with and multiple cerebriform-appearing polyps due to smooth
life-long medical conditions from child-centered to adult- muscle bands coursing through the polyp (Figures 1A and
centered care. Preparation for this transition takes place 2A–C, and Table 2). Hamartomatous polyps vary in size and
throughout childhood and adolescence to achieve inde- may have a characteristic histologic structure, which makes
pendent health management in adulthood. Steps required it possible to distinguish the PJ polyp. PJ polyps are typically
are individualized based on the developmental needs of composed of branching bands of smooth muscle covered
the patient. Inherited conditions involve generational fac- by hyperplastic glandular mucosa.10 PJS polyps may develop
tors, as multiple family members may be affected. Health in the stomach, small intestine, and colon. Rectal bleeding
care providers can assist in the transition of care by coordi- with anemia is the most common presentation, followed by
nating screening and surveillance to ensure patients abdominal pain, diarrhea, and intussusception. The clinical
receive recommended care.5 management in early life is initially focused on preventing
complications of small bowel polyposis–related obstruction
and bleeding and, in adulthood, the focus is primarily on
GASTROINTESTINAL HAMARTOMATOUS management of cancer risk.
POLYPOSIS SYNDROMES

The hamartomatous polyposis syndromes are rare entities Diagnosis


with an estimated prevalence of 1/100,000–200,000,6,7 but The diagnostic clinical features of PJS include the pres-
this has not been measured directly in any population. The ence of 2 or more histologically confirmed PJ polyps; any
term hamartoma implies a non-neoplastic tumor with a number of PJ polyps in an individual who has a family his-
markedly distorted architecture composed of an abnormal tory of PJS in a first-degree relative; characteristic mucocu-
mixture of cells and tissue normally present in that particular taneous pigmentation in a person with a family history of
area. The diagnosis is based on the presence of a pathogenic PJS; or any number of PJ polyps in a person with the char-
germline variant or meeting clinical criteria for the syndrome. acteristic mucocutaneous pigmentation of PJS.11
The hamartomatous polyposis syndromes are distinct from
Lynch syndrome and the adenomatous polyposis syndromes, Genetics
based on the presence of hamartomas (Figures 1 and 2). In 1997, a genetic locus for PJS was mapped to chromo-
Certain hamartomatous polyps of the gut have a unique some 19p13.3,12 which in 1998 led to the cloning of the
histopathological appearance, such as those associated with STK11 (serine/threonine kinase) gene, which encodes the
PJS, PTEN, JPS, and HMPS.8 Hamartomas are not typically LKB1 (liver kinase B1) protein, and linkage to PJS.13
characterized by dysplasia, but some evidence suggests the STK11 functions like a tumor suppressor gene, regulates
existence of a hamartoma–carcinoma pathway in some of cell growth via adenosine monophosphate–activated pro-
these polyps. tein kinase,14 and negatively regulates mTOR signaling.15

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1029


Gastrointestinal hamartomatous polyposis syndromes

Figure 1. Histologic characteristics of polyps. hamartomatous polyps. Images courtesy of Drs Aaron Pollett and Thomas Plesec. A, Peutz-Jeghers polyp:
pathognomonic broad bands of mucosal smooth muscle seen throughout the lesion. B, Juvenile polyp: characteristic chronic inflammatory infiltrate and
cystic dilatation. C, Adenomatous polyp: characteristic hypercellularity with glandular crowding, enlarged nuclei, increased mitotic activity and reduced
goblet cells. By definition, all tubular adenomas show epithelial dysplasia. D, Ganglioneuroma: benign neuroectodermal tumor composed of ganglion and
Schwann cells.

PJS is inherited in an autosomal dominant fashion with an herited from the unaffected parent in a somatic tissue accord-
inactivating germline pathogenic variant inherited from the ing to the classic “two-hit” model of Knudson.16 However, loss
affected parent. It was initially assumed that the polyps of the wild-type allele (ie, the second hit) is not an obligatory
occurred after the loss of the second, wild-type, allele in- feature of PJ polyps.17 Moreover, recent data in mice indicate

1030 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

Figure 2. Endoscopic images of hamartomatous polyposis syndromes. Endoscopic photos provided courtesy of Swati Patel, MD, Gregory Idos, MD, and
Carol Burke, MD. A, Peutz-Jeghers small bowel polyps. B, Peutz-Jeghers gastric polyps. C, Peutz-Jeghers colon polyps. D, Juvenile polyposis gastric
polyps. E, Juvenile polyposis colon polyp. F, Cowden syndrome associated–esophageal glycogenic acanthosis. G, Cowden syndrome small bowel polyps.
H, Cowden syndrome colon polyps. I, Cowden syndrome gastric polyps.

that the presence of a single inactivating germline pathogenic for the formation of the polyps in these mice. Furthermore,
variant (ie, haploinsufficiency), as occurs in individuals with conditional knockout of the STK11 gene targeted to gastroin-
PJS, promotes the development of gastrointestinal testinal smooth muscle cells yields the same polyposis pheno-
polyposis, and that loss of the wild-type allele is not necessary type in mice.18 Evidence suggests that LKB1 deficiency in

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1031


Gastrointestinal hamartomatous polyposis syndromes

TABLE 2. Clinical features of gastrointestinal hamartomatous polyposis syndromes

“Commercially
Syndrome available gene testing” Polyps Clinical features

PJS STK11 Peutz-Jeghers polyps (pathologically Childhood: Labial pigmentation; gastrointestinal


characteristic) bleeding and intussusception
Adults: Increased risk for multiple cancers
JPS SMAD4 or BMPR1A Juvenile (inflammatory) polyps; juvenile polyps Childhood: Gastrointestinal bleeding, auto-amputation
and inflammatory polyps are pathologically of polyps; anemia
indistinct Adults: CRC and gastric cancer
CS PTEN (inactivation) Hyperplastic polyps; juvenile-like polyps; Childhood: none
WWP1 (gain-of-function) ganglioneuromas; lipomas; Adults: multiple cancer risks
hamartomas;adenomas
BRRS PTEN Same as for CS Developmental delay, hemangiomas, lipomas,
gastrointestinal polyps
HMPS GREM1 (duplication Pathologically mixed with features of adenoma, Increased risk of colonic polyposis and CRC
upstream of promoter) hyperplastic polyps, inflammatory polyps

either T cells19 or mesenchymal cells18,19 leads to immune cell Very high lifetime risks of cancer occur in multiple or-
proliferation in the stroma. Stromal deficiency of LKB1 leads gans in patients with PJS, inside and outside the gut. Intra-
to tumor formation (in mice) via the interleukin-11–JAK/ tubular large-cell hyalinizing Sertoli cell neoplasms, ovarian
STAT3 (Janus kinase/signal transducer and activator of sex cord tumors, and adenoma malignum of the uterine
transcription 3) pathway, and administration of the JAK1/2 cervix, although not common, are linked to PJS25 (Table 3).
inhibitor ruxolitinib dramatically reduces polyposis in The lifetime risk of colon, stomach, and small bowel can-
mice.20 The phenomenon of stromal-driven epithelial polyp cer has been estimated at 12%–39%, 29%, and 13% respec-
development may be referred to as a “landscaper” genetic tively6,26–28 (Table 3). Most CRCs occur after the mid-20s
effect.21 These discoveries raise the possibility of (range, 27–71 years, median age 46 years), but these malig-
the development of novel preventive pharmacological nancies have been reported in teenage years as well.26
interventions for patients. Mean age at diagnosis of gastric adenocarcinoma ranges
Genotype–phenotype relationships reveal that missense between 30 and 40 years and of small intestinal cancer
alterations in STK11 are associated with later onset of symp- between 37 and 42 years.26,27,29
toms than other sequence variations.11,22 Some of the In a meta-analysis of 210 cases reported in 6 publications
germline pathogenic variants in STK11 are Alu-mediated (retrospective cohort studies with kindred-based ascertain-
deletions and inversions.23 Deletions and inversions are ment from the United States, United Kingdom, and The
likely to completely inactivate a gene, whereas a missense Netherlands) on PJS that were based on clinical and histologic
pathogenic variant may have an intermediate effect on criteria with varying types of ascertainment, the relative risk
protein function. Approximately 15% of STK11 pathogenic (RR) for any cancer was 15.2 (95% confidence limits [CL], 2
variants in PJS involve large genetic deletions.24 Therefore, and 19) compared with a variety of time period–specific
genetic diagnostic platforms must include strategies to U.S.-based registries. Significantly increased age-adjusted
detect these types of germline variations; at least some of cancer risks were noted for the small intestine (RR, 520;
the “missing” germline pathogenic variants may involve 95% CL, 220 and 1306; cumulative risk [CR], 13%), stomach
obscure rearrangements of the STK11 gene. (RR, 213; 95% CL, 96 and 368; CR, 29%), pancreas (RR, 132;
Cancer risk 95% CL, 44 and 261; CR, 36%), colon (RR, 84; 95% CL, 47
and 137; CR, 39%), esophagus (RR, 57; 95% CL, 2.5 and 557;
Question: Which organs should undergo surveil- CR, 0.5%), ovary (RR, 27; 95% CL, 7.3 and 68; CR, 21%),
lance when caring for a patient with Peutz-Jeghers lung (RR, 17.0; 95% CL, 5.4 and 39; CR, 15%), uterus (RR,
syndrome? 16.0; 95% CL, 1.9 and 56; CR, 9%), testes (RR, 4.5; 95% CL,
Recommendation: Patients with Peutz-Jeghers syn- 0.12 and 25; CR, 9%), and breast (RR, 15.2; 95% CL, 7.6 and
drome are at increased risk for cancer in multiple or- 27; CR, 54%).26 Mean age at the time of cancer diagnosis
gans, including cancer of the breast, small bowel, was 42.9 years. The absolute risk of developing any cancer
colon, stomach, pancreas, ovaries, testes, and lungs. between the ages of 15 and 64 was estimated to be 93%.
Given this risk, we recommend a multidisciplinary The risk of cancer in PJS was revisited in 2 follow-up
approach to cancer surveillance in these organs. (Strong studies that included some who were members of the orig-
recommendation, low quality of evidence) inal cohort.28,30 The lattermost report included some of
the original 210 cases and expanded to 419 cases, in

1032 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

TABLE 3. Risk of cancer in hamartomatous polyposis syndromes

Site General population risk,* % Syndrome risk, % Mean age at diagnosis, y Reference

PJS
Colorectal 4.3 39 42–46 26, 29
Stomach <1 29 30–40 26, 29
Small bowel <1 13 37–42 26, 29
Breast 12.9 32–54 37–59 26, 28, 29
Ovarian (mostly SCTAT) 1.2 21 28 26
Cervix (adenoma malignum) <1 10–23 34–40 26
Uterus 3.1 9 43 26, 29
Pancreas 1.7 11–36 41–52 26, 28, 29, 32
Testicular (Sertoli cell tumor) <1 9 6-9 26, 29
Lung 6.3 7–17 47 26, 28, 29
JPS
Colon 4.3 39 44 86
Stomach <1 5–21 54 65, 67, 89
CS
Breast 12.9 25–85 38–46 100, 101, 102
Thyroid 1.3 3–38 31–38 100, 101, 102
Uterus 3.1 5–28 25 95, 100, 101, 102
Kidney (renal cell) 1.7 15–34 40 97, 100, 104
Colon 4.3 9–18 35 100–103, 106
Melanoma 2.3 6 3y 100, 101
HMPS
Colon 4.3 Increased d 119–121
SCTAT, Sex cord tumors with annular tubules.
*National Cancer Institute. Surveillance, Epidemiology, and End Results Cancer Statistics Review 1975–2017. Lifetime risk (%) of being diagnosed with cancer by site, 2017.
yYoungest age of onset.

which 297 had confirmed germline pathogenic variants in lower than the American and European data), early age
STK11.28 The cumulative incidences of cancer by decade of onset, and a similar tumor spectrum.32
from age 20 to 70 years were 2%, 5%, 17%, 31%, 60%,
and 85% respectively, confirming the initial estimates and Surveillance of affected individuals
tumor spectrumdpredominantly the gastrointestinal tract The effectiveness of cancer surveillance in PJS has not
and breast. Cancer risks were the same in those with a been evaluated in controlled studies. Consequently, sur-
clinical diagnosis but no detectable germline pathogenic veillance recommendations have been developed by
variant in the STK11 gene. consensus groups and expert opinion analyzing cancer
Several collaborative studies from Europe and the risks and published organ-specific surveillance protocols
United States again found similar risks for cancer.29,31 A (Tables 3 and 4).11,33–35
systematic review of 20 cohort studies looking at 1644
patients with PJS confirmed at least 1 cancer in >90% of Question: How and when should small bowel sur-
patients with PJS at a mean age of 42 years.29 CRC was veillance be performed in Peutz-Jeghers syndrome?
the most commonly diagnosed tumor, followed by Recommendation: We recommend that baseline
cancers of the breast, small intestine, stomach, lung, small bowel surveillance using video capsule endoscopy
pancreas, cervix, ovary, bile ducts, and testicles. A or magnetic resonance enterography be performed be-
multicenter study from Italy of 119 STK11 pathogenic tween ages 8 and 10 years or earlier if the patient is
variant carriers reported an overall RR for cancer of 22.0 symptomatic. If no polyps are found at the initial exam-
in women (in part because of additional risks for cervical ination, surveillance should resume at age 18 years.
cancers) and 8.6 in men, compared with an Italian-based Because of the risk of small bowel intussusception,
small bowel surveillance in adulthood is recommended
general population registry, and a cumulative risk of cancer
to continue throughout life every 2–3 years. (Strong
reaching 89% by age 65 years.27 A more recent study from
recommendation, low quality of evidence)
China confirmed elevated cancer risks (albeit somewhat

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1033


Gastrointestinal hamartomatous polyposis syndromes

TABLE 4. Surveillance guidelines in hamartomatous polyposis syndromes

ACG 2015 NCCN 2020 ESPGHAN 2019 USMSTF 2020


Age of Interval, Age of Interval, Age of Interval, Age of Interval, Evidence
Examination initiation, y y initiation, y y initiation, y y initiation, y y grade

JPS
Colonoscopy 12–15 1–3 15 2–3 12–15 d 12–15 1–3 Low
Upper endoscopy 12–15 1–3 15 2–3 Late teens* d 12–15 1–3 Low
Screen for vascular lesions 6 mo d 6 mo d At diagnosis d 6 moy d d
PJS
Colonoscopy 8, 18z 3 Late teens 2–3 8 3 8–10, 18z 2–3 Very low
z
Upper endoscopy 8, 18 3 Late teens 2–3 8 3 8–10, 18z 2–3 Very low
z z
VCE 8, 18 3 w8–10x 2–3 8 3 8, 18 2–3 Low
CT or MRE of small bowel d d w8–10x 2–3 d d d d d
MRI/MRCP or EUS of 30 1–2 w30–35{ 1–2 d d 35 1 Low
pancreas
MRI and/or mammogram 25 1 w25 1 d d d d d
Physical examination Birth to 1 w10 1 d d d d d
teenage
Pelvic examination and Pap 25jj 1 w18–20 1 d d d d d
smear
Testicular examination Birth to 1 w10 1 d d d d d
teenage
PHTS
Colonoscopy 15 2 35 5 d d 35 5 Low
Upper endoscopy 15 2–3 d d d d d d d
Thyroid examination and Adolescence 1 7 1 d d d d d
US
MRI and/or mammogram 30–35 1 30-35 1 d d d d d
Endometrial sampling 30–35 1 d 1–2 d d d d d
Urinalysis or renal US 18 1 40 1–2 d d d d d
Skin examination w18 1 At diagnosis 1 d d d d d
ACG, American College of Gastroenterology; CT, computed tomography; ESPGHN, European Society for Paediatric Gastroenterology Hepatology and Nutrition; EUS, endoscopic
ultrasound; MRCP, magnetic resonance cholangiopancreatography; NCCN, National Comprehensive Cancer Network.
*Upper gastrointestingal screening in JPS SMAD4 carriers is indicated in asymptomatic patients starting in late teens. For non-SMAD4 JPS patients, upper endoscopic screening is
only indicated if the patient has relevant symptoms or anemia not explained by colonic polyps.
ySMAD4 mutation carriers should be clinically evaluated for HHT at the time of the diagnosis, including screening for, and appropriate management of, cerebral and pulmonary
AVMs.
z
First procedure at 8 years of age; if polyps present, repeat every 3 years; if no polyps, restart at 18 years of age and every 3 years.
xBaseline at age 8–10 years of age with follow-up interval based on findings but at least by age 18 years of age, then every 2–3 years, although this may be individualized by at
least age 18 years, then every 2–3 years, although this may be individualized, or with symptoms).
{Based on clinical judgment, early initiation age may be considered, such as 10 years younger than the earliest age of onset in the family.
jjACG 2015 Guidelines recommend transvaginal ultrasound as part of surveillance beginning at age 25 years.

Gastrointestinal polyposis and cancer surveillance with esophagogastroduodenoscopy (EGD),


Gastric, small bowel, and colorectal polyposis occur in colonoscopy, and small bowel imaging with video
88%–100% of patients, with the majority appearing in capsule endoscopy (VCE) and/or magnetic resonance en-
the small bowel (60%–90%) and colon (50%–64%).36 terography (MRE) is recommended to begin at age 8
Polyps can vary in number (1–100) and size (0.1–3 cm in years.11,33–35
diameter) and age of onset of symptoms may vary. Polyp Intussusception is rare in children younger than 5
growth begins in childhood by age 10 years (33%), with years, and the precise risk of intussusception between 5
most experiencing symptoms such as bleeding, and 18 years of age is unknown. Retrospective registry
abdominal pain, intussusception, or obstruction (68%) data report that 23 of 34 adults with PJS (68%) had under-
by age 18 years.37 In affected or at-risk individuals, early gone laparotomy by the age of 18 years, 70% of which

1034 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

were performed as an emergency. By the age of 10 years, Management of polyposis


30% had required a laparotomy.37 In a single-institution Although the malignant potential of PJ polyps is un-
study of 379 pediatric patients who underwent pneumatic known and the evidence of benefit from gastrointestinal
reduction for intussusception (from all causes), one- surveillance is not robust, endoscopic removal of polyps
quarter required operative management.38 There is a is recommended to prevent polyp-related complications
paucity of studies that assess modalities to evaluate the and to reduce the risk of cancer. In a study of long-term
small bowel in patients with PJS. Retrospective data outcomes of gastrointestinal procedures from a PJ polypo-
comparing VCE to small bowel barium studies have sis registry, investigators tracked the results of 776 proced-
reported VCE as a useful diagnostic tool in PJS. A ures among 63 patients with PJS at a median age of 20
retrospective study from France included 27 children years (range, 3–59 years). A total of 2461 polypectomies
who underwent at least 1 VCE.39 The authors found VCE were performed; more than 1000 polyps were removed
was a useful diagnostic tool, however, findings at VCE may during colonoscopy and more than 400 polyps were
not predict future bowel obstructions. Although VCE may removed during EGD, and the remaining polyps were
not predict future bowel obstructions, it has a greater removed by means of enteroscopy or laparotomy of the
sensitivity in detecting small bowel polyps compared with small bowel.41 A substantial proportion of patients
MRE and provides patients with another alternative if they required intervention for removal of large polyps and the
are unable to undergo MRE. Ten children with PJS with authors concluded that surveillance reduced polyp
polyps >15 mm identified by VCE and MRE underwent burden and likelihood of polyp-related complications,
single-balloon enteroscopy.40 In this small study, single- and provided cancer surveillance. Therefore, we recom-
balloon enteroscopy was effective for treating small bowel mend polypectomy for polyps in the stomach and colorec-
polyps. Further larger, multicenter studies are warranted tum that are >0.5 cm in size on endoscopic surveillance
to accurately determine the safety of therapeutic single- and an attempt to remove all polyps if endoscopically
balloon enteroscopy in children.7 Based on the published feasible.33,35,42,43
data, VCE or MRE is recommended between ages 8 and 10 In the small bowel, balloon enteroscopy and MRE have
years in asymptomatic patients. Investigations should similar diagnostic yields for lesions 15 mm, but endos-
commence earlier if patients have symptoms. If no polyps copy permits polyp removal.43 Consequently, removal,
are found on the baseline investigation, repeat small preferably by enteroscopy, of small intestinal polyps that
bowel evaluation may commence at age 18 years. If polyps are symptomatic or rapidly growing, or asymptomatic
are found, further investigation and surveillance should be polyps that are >1.0–1.5 cm in size, has been
individualized based on polyp size and location. Because recommended.33,35,37,42,44,45 Surgery is often needed
of the risk of small bowel intussusception, small bowel when small bowel intussusception occurs. Some
surveillance in adulthood continues throughout life every authorities recommend an attempt to clear the small
intestine of polyps during laparotomy by means of
2–3 years. If polyps are present in the colon or stomach,
intraoperative endoscopy with polypectomy or, for larger
EGD or colonoscopy is continued as necessary every 2–3
polyps, by means of enterotomy. This “clean sweep”
years.
approach appears to decrease the need for recurrent
small bowel surgery.46 Thus, the early management focus
Question: What is the recommended approach to
is on large hamartomatous polyps and their tendency to
endoscopic surveillance of the stomach, duodenum,
obstruct the gut or bleed. As children grow older and
and colon in Peutz-Jeghers syndrome?
transition into adulthood, the focus shifts to managing
Recommendation: We suggest a baseline upper
the cancer risk.
gastrointestinal endoscopy between the ages of 8 and
10 years, which could be performed at the time of
Question: What size polyps found on small bowel
capsule placement for small bowel surveillance or if
imaging in Peutz-Jeghers syndrome should be removed?
polyps are identified on magnetic resonance enterogra-
Recommendation: We recommend polypectomy
phy. Although the initiation age for colonoscopy remains
of small bowel polyps that are symptomatic
uncertain, we also suggest initiation of colonoscopy at
or 10 mm to prevent intussusception and other com-
the same time as esophagogastroduodenoscopy. In
plications, such as bleeding. (Strong recommendation,
those in whom characteristic polyps are detected, both
low quality of evidence)
colonoscopy and esophagogastroduodenoscopy should
be repeated every 2–3 years. In those in whom there Breast cancer
are no Peutz-Jeghers polyps at baseline, surveillance is Invasive ductal adenocarcinoma poses the greatest risk
repeated at age 18 years, or sooner should symptoms
of early malignancy to patients with PJS (24%–54% lifetime
arise, and then every 3 years. (Weak recommendation,
risk in women) and often presents at a young age (range,
very low quality of evidence)
19–61 years in 1 study).26,47 The risk of breast cancer in

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1035


Gastrointestinal hamartomatous polyposis syndromes

women with PJS is within the same range as women recommended by the Cancer of the Pancreas Screening
affected by BRCA-1 or BRCA-2 pathogenic variants (40%– Consortium. At the current time, the USMSTF awaits
85% lifetime risk for breast cancer).26,48 Therefore, definitive data before making a recommendation
consensus opinion surveillance recommendations by regarding fasting glucose and hemoglobin A1c. Recent
groups that include breast cancer experts49,50 include updates to the Cancer of the Pancreas Screening
monthly breast self-examination starting at age 18 years, Guidelines detail surveillance protocols and management
biannual clinical breast examination starting at age 25 recommendations based on imaging and endoscopic
years, annual breast magnetic resonance imaging (MRI) findings.55
from ages 25–29 years, and mammography with consider-
ation of tomosynthesis (3-dimensional mammography) Gynecological cancers
alternating every 6 months with breast MRI with contrast The lifetime risks for ovarian, uterine, and cervical can-
from ages 30 to 75 years.6,11 The option of cer are estimated at 21%, 9%, and 10%–23%, respectively,
prophylactic mastectomy might be discussed on a case- with mean age at presentation of 28–35 years, 43 years,
by-case basis, factoring in family history. In patients with and 34–40 years, respectively.6,26,27 Of note, almost all
PJS, referral to a breast cancer specialist for management ovarian tumors in patients with PJS are sex cord tumors
of breast cancer surveillance is reasonable, and a multidis- with annular tubules, and rarely cystadenomas or
ciplinary approach including a breast surgeon is recom- granulosa cell tumors. Sex cord tumors with annular
mended when prophylactic mastectomy is being tubule neoplasms are a heterogeneous group of benign
considered. or malignant neoplasms, but the tumors rarely have
lymph node metastasis.26 Also, an unusual percentage of
Question: What is the recommended pancreatic cervical cancers in patients with PJS are adenoma
cancer surveillance in Peutz-Jeghers syndrome? malignum, a rare, well-differentiated, cervical adenocarci-
Recommendation: We suggest annual pancreatic noma that is associated with a poor prognosis and difficult
cancer surveillance with either magnetic resonance to diagnose on Pap smear. A high level of suspicion is
cholangiopancreatography or endoscopic ultrasound required for diagnosis.56 Recommendations for
starting at age 35 years. (Weak recommendation, low gynecological surveillance are pelvic examination with
quality of evidence) Pap smear and transvaginal ultrasound annually starting
at age 25 years.33,35
Pancreas cancer
Pancreatic cancer is the third most frequently occurring Testicles
malignancy in patients with PJS. The lifetime risk of The estimated risk of testicular cancer in male patients
pancreatic ductal adenocarcinoma is between 11% and with PJS is 9%, mean age at diagnosis is 9 years (range,
36%, presenting, on average, at ages 41–52 years, with 3–20 years).6,26,27 These tumors present as testicular
95% of cases occurring after age 24 years.6,26,28,51,52 PJS masses. The tumors are Sertoli cell tumors that can
is associated with the highest relative risk of all cause gynecomastia and other signs of hyperestrogenism
pancreatic cancer syndromes, with the exception of and occasionally virilization and/or accelerated height
hereditary pancreatitis (25%–40% lifetime risk).43 growth.26 Accelerated height velocity can be challenging
Consequently, a high-risk pancreatic surveillance protocol to detect as adolescents have “growth spurts” as part of
is recommended. The consensus recommendations of normal maturation and development. Expert opinion
the International Cancer of the Pancreas Screening Con- recommends annual history and physical examination
sortium53 recommend pancreas MRI/magnetic resonance (including self-examination) with observation for femi-
cholangiopancreatography and/or endoscopic ultrasound nizing changes and examination of the testicles6,34,35;
every 1–2 years starting from age 40 years, for which based on the range of age at diagnosis of this tumor,
evidence suggests that these cancers can be found in examination should start from birth. Ultrasound of the
earlier stages.54 The National Comprehensive Cancer testicles every 2 years from birth to age 12 years has
Network Guidelines recommend initiating pancreatic been suggested.44
cancer surveillance between the ages of 30 and 35 years.
Due to reports of pancreatic cancer diagnoses in patients Lung
with PJS before the age of 40 years,26 the USMSTF The lifetime risk of lung cancer in patients with PJS has
suggests initiating annual surveillance at age 35 years been estimated between 7% and 17%,6,26–28,30 compared
with MRI/magnetic resonance cholangiopancreatography with 0.2%–0.6% in nonsmokers in the general
and/or endoscopic ultrasound. Ideally, these population.57 The cumulative risk of lung cancer in PJS
examinations would alternate on an annual basis, as they surpasses 5% by age 55 years.6 Of note, lung cancer risk
are complementary. In addition, routine fasting glucose in patients with PJS has not been calculated with
and hemoglobin A1c at initiation of screening is adjustment for smoking status. The RRs of lung cancer in

1036 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

patients with PJS compared with nonsmokers are similar to JUVENILE POLYPOSIS SYNDROME
people with a more than 30 pack-year history of smoking
who have quit for 10–15 years (hazard ratio, 14.8).58 Question: Who should undergo a genetic evaluation
Currently, the American College of Chest Physicians, for juvenile polyposis syndrome?
American Society of Clinical Oncology, and American Recommendation: We recommend genetic evalua-
Cancer Society recommend low-dose computed tomogra- tion for any individual with 1) 5 or more juvenile polyps
phy annually for individuals with this level of cumulative of the colon or rectum; 2) 2 or more juvenile polyps in
risk for lung cancer from ages 55 to 74 years.59,60 There other parts of the gastrointestinal tract; or 3) any num-
are no data to show benefit of lung cancer surveillance in ber of juvenile polyps and 1 or more first-degree rela-
patients with PJS. Lung cancer surveillance with annual tives with juvenile polyposis syndrome. (Strong
low-dose computed tomography, as performed in smokers recommendation, low quality of evidence)
at high risk for lung cancer, may be considered in patients
with PJS. Smoking cessation counseling in patients with PJS Clinical features
is advisable to mitigate risk.33,34 JPS is an autosomal dominant inherited condition in
which multiple juvenile polyps are found in the colorectum
(98% of affected patients) (Figure 1B), stomach (14%),
Chemoprevention jejunum and ileum (7%), and duodenum (7%).63–65 The
Currently, there are no known chemopreventive agents incidence of JPS is between 1 in 100,000 and 1 in
in clinical practice that slow or prevent the development of 160,000 individuals.65 The polyps in JPS vary in size from
intestinal polyps and cancers in PJS. Pathogenic variants in small sessile nodules to pedunculated lesions that are 3
the STK11 gene decrease inhibition of mTOR leading to cm in diameter. Histologically, the typical juvenile polyp
the development of intestinal polyps. A trial examining has a distinctive cystic architecture, dilated mucus-filled
the oral selective mTOR inhibitor, everolimus, was glands, a prominent lamina propria, and Paneth cells en-
stopped prematurely because of poor patient accrual. meshed within a dense infiltration of inflammatory cells.
Only 2 patients were enrolled, 1 with pancreatic cancer In patients with germline pathogenic variants in SMAD4,
that progressed and another patient who withdrew from additional features of gastric polyposis, gastric cancer,
the protocol because of severe complications from the and a JPS–hereditary hemorrhagic telangiectasia (HHT)
medication.61 overlap syndrome are common. The management of JPS
In a murine model of PJS, treatment with celecoxib, a is based on decreasing the risk of bleeding and gastric
COX2 inhibitor, resulted in a >50% reduction in polyp and colorectal cancer through polypectomy. Patients with
burden. When used in patients with PJS with diffuse polyp- JPS-HHT overlap syndrome should have lifelong HHT sur-
osis in the body of the stomach (tens to hundreds), 2 of 6 veillance. One study also demonstrated that as many as
had a significant reduction in polyp number, as assessed by 38% may have thoracic aorta abnormalities.66
5 independent evaluators, after administering celecoxib Juvenile polyps are most often solitary and are not syn-
(200 mg twice per day for 6 months).62 dromic, occurring sporadically in infants and children. A
typical history associated with a solitary juvenile polyp is
the asymptomatic passing of a polyp into an infant’s diaper.
Summary Juvenile polyps appear endoscopically to have a smooth
PJS is associated with a very high cumulative lifetime red surface, may be sessile or pedunculated, and patholog-
risk of cancer of multiple organs, including, but not limited ically are characterized by cystic dilatation of mucus-filled
to, the gastrointestinal tract. Intensive surveillance is rec- glands suspended in an inflamed stroma (Figures 1B and
ommended to prevent and manage complications of pol- 2D and E). These lesions may also be called retention
yposis and identify cancer at an early stage. The polyps or inflammatory polyps because of their
development of polyposis and the cancer risks may be a microscopic appearance.
reflection of the effects of haploinsufficiency of the LKB1
protein, for which there are possible medical therapies to Diagnosis
be explored. It is unclear whether the basic mechanisms The diagnosis of JPS is made based on clinical criteria or
responsible for hamartoma formation in younger life are identification of a germline pathogenic variant in SMAD4 or
the same as those that create the risks for cancer later in BMPR1A. The clinical diagnosis of JPS is made when a per-
life. Simulation models and clinical trials are needed to son has any 1 of the following: 1) 5 or more juvenile polyps
optimize endoscopic surveillance frequencies and the use of the colon or rectum; 2) any number of juvenile polyps in
of imaging modalities in adults. In addition, collaborative parts of the gastrointestinal tract other than the colon; or
multicenter consortia may help facilitate chemoprevention 3) any number of juvenile polyps and 1 or more first-
trials in the future. degree relatives with JPS.65

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1037


Gastrointestinal hamartomatous polyposis syndromes

Genetics immune cells in JPS, and may explain the inflammatory


JPS is an autosomal dominant disorder with approxi- appearance of juvenile polyps independent of abnormal
mately 75% of cases inherited from a parent and 25% rep- biology in the epitheliumdanother landscaper
resenting de novo pathogenic variants.64 Approximately mechanism, as discussed above for PJS. These
60% of patients with JPS have a pathogenic variant in the observations may have important clinical implications
BMPR1A or SMAD4 gene.67 Rarely (1 in 1,000,000) for future attempts to halt the appearance or growth of
individuals develop features of JPS and PHTS, known as the inflammatory polyps. However, allelic loss (ie, a
juvenile polyposis of infancy, due to a large deletion somatic variants as the second hit) occurs in at least
encompassing both the BMPR1A and PTEN genes. some polyps of JPS patients.75,76
In a study from the Cleveland Clinic of 35 patients with
JPS, germline pathogenic variants in SMAD4 and BMPR1A Juvenile polyposis syndrome–hereditary
were associated with similar colonic polyposis phenotypes, hemorrhagic telangiectasia overlap syndrome
but SMAD4 pathogenic variant carriers were more likely to HHT occurs in approximately 15%–81% of patients with a
have more gastric polyps, and an 11% risk of gastrointes- germline SMAD4 pathogenic variant.77,78 The clinical
tinal cancer.68 However, no gastric cancers were reported features of HHT, such as epistaxis, obscure gastrointestinal
in patients with BMPR1A pathogenic variants in 8 bleeding, digital clubbing, visceral arteriovenous
patients followed for a mean of 11 years. malformations (AVMs), and mucocutaneous telangiectasias
However, germline pathogenic variants in other genes should be sought in SMAD4 patients. International
may cause a hamartomatous polyp phenotype. In a study guidelines (outlined below) recommend that surveillance
of 49 patients referred to the Cleveland Clinic for unex- and treatment for HHT complications are necessary for all
plained hamartomatous or hyperplastic polyps, germline SMAD4 carriers.79
pathogenic variants were found in multiple genes,
including endoglin (ENG, a gene associated with HHT), Juvenile polyposis of infancy
STK11 (the PJS gene), SMAD4, BMPR1A, and PTEN.69 In a Juvenile polyposis of infancy is a severe form of juvenile
later study from this group of 603 patients with a polyposis. This disease presents in the first 2 years of life
“moderate load of gastrointestinal polyps” with at least 1 with diarrhea, abdominal pain, rectal bleeding, refractory
confirmed to be a hamartoma or hyperplastic polyp, 13% anemia, hypoalbuminemia, and enteropathy. Case reports
were found to have a germline pathogenic variant in at indicate that a large deletion in the long arm of chromo-
least 1 of the genes listed above and 20% of the cohort some 10 (10q23), encompassing the PTEN and BMPR1A
had a personal history of CRC.70 genes, is associated with the development of the dis-
As discussed in the context of PJS, it has been tradi- ease.69,80–83
tionally thought that a germline pathogenic variant asso-
ciated with a hereditary colon cancer syndrome did not Question: Which organs should undergo surveil-
change the biology of the normal tissues and a second lance when caring for a patient with juvenile polyposis
(somatic) hit was required for a tumor to form. In fact, syndrome?
allelic loss of the SMAD4 locus was found in the epithelial Recommendation: Patients with juvenile polyposis
component of juvenile polyps of patients with JPS syndrome are at increased risk for cancer in multiple or-
(together with a germline pathogenic variant in SMAD4) gans, including cancer of the colon and stomach.
using in situ hybridization, but not in the inflammatory Given this risk, we recommend patients with juvenile
or stromal cells. This was compatible with the Knudson polyposis syndrome undergo surveillance of the colon
2-hit model.71 However, another group has reported and stomach. (Strong recommendation, low quality
that SMAD4 haploinsufficiency (ie, a lower dose of the of evidence)
gene product due to the specific type of germline
pathogenic variant) causes the JPS phenotype in Gastrointestinal polyposis and cancer
humans, supported by data from mice.72 However, just In a retrospective chart review study of 257 children
as in PJS, being haploinsufficient for SMAD4 (ie, just the with juvenile polyps at a single-referral center, patients pre-
germline pathogenic variant), is associated with partially sented at a median age of 5.6 years, and at colonoscopy
diminished transforming growth factor–b signaling, at 39% had multiple polyps.84 Among 192 patients who
least in mice,72 as this alters proliferation in T cells, underwent complete colonoscopy at initial diagnosis, 117
which contributes to the development of polyps and (60.9%) had a single polyp and 75 (39.1%) had multiple
cancer.73,74 Specific deletion of SMAD4 in mouse T cells polyps. These lesions recurred in 21 of 47 patients
leads to up-regulation of the Th17-inflammatory pathway (44.7%) after an initial eradication, including 3 of 18
in the stroma, and the growth of large polyps in the presenting with a single polyp, and neoplasia was found
gastrointestinal tracts of mice.73,74 This raises the in 3.9% of lesions. Patients with JPS often have a variable
question of whether there is abnormal regulation of presentation of polyp distribution, which may occur

1038 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

throughout the colon and/or stomach and the cancer risk telangiectasias, an annual history and physical examination
is attributable to the presence of dysplasia in the polyps. and complete blood counts to monitor for rectal bleeding
One study of 78 juvenile polyps from 12 patients with and/or anemia should begin at age 12–15 years in patients
JPS and 34 patients with sporadic juvenile polyps with a germline SMAD4 pathogenic variant.
reported that dysplasia was present in 31% of the polyps Endoscopic polypectomy is recommended for colo-
from patients with JPS but in none of the polyps from rectal polyposis management. Surgery with colectomy
patients with sporadic juvenile polyps.85 Dysplasia in and ileorectal anastomosis is recommend for patients
polyps from patients with JPS was associated with with CRC, endoscopically unmanageable colon polyp
somatic variants in the APC gene.85 In a longitudinal burden, and uncontrolled anemia from colonic bleeding.33
study from St Mark’s Hospital of 44 patients with JPS In some cases, proctocolectomy is necessary for rectal
from 30 kindreds, a total of 787 polyps (juvenile and cancer or advanced polyp burden of the rectum.
adenomatous) were resected, and 65 of 787 (8.3%) Colectomy in patients with JPS should be reserved for
contained mild/moderate architectural dysplasia, and 20 patients with polyp burdens that cannot be managed by
additional polyps (2.5%) were adenomatous.65 polypectomy, persistent blood loss leading to severe
Patients with JPS are at increased risk for cancer princi- anemia or hypoalbuminemia, or cancer. A decision to
pally in the stomach and colon (Table 3). In a small cohort proceed to colectomy should be reviewed with
of patients with JPS (n Z 84) relative to age-, sex-, and gastroenterologists and surgeons with expertise in caring
race-matched controls, the RR of CRC was estimated to for individuals with hereditary polyposis syndromes.
be 34 (95% CL, 14.4 and 65.7), with a cumulative lifetime The risk of gastric cancer is a concern in patients with
risk of CRC reaching 38.7%.86 The CRCs were diagnosed JPS with SMAD4 pathogenic variants and gastric polypo-
at a mean age of 43.9 years. Interestingly, no other sis. There is a paucity of published data evaluating the
gastrointestinal cancers were noted in this cohort, but stomach and small bowel in pediatric patients with JPS.
the numbers were small and the duration of the study Those with upper gastrointestinal symptoms, or with
was short. Individuals with gastric polyposis, usually in anemia not explained by colonic polyps, should undergo
association with pathogenic variants in SMAD4 are also at evaluation with upper endoscopy.92 In pediatric patients
risk for gastric cancer, with the lifetime risk estimated to without SMAD4 pathogenic variants, according to the
be at least 30% and median age of diagnosis is 58 years; existing data, gastroscopy is not indicated unless the
this has not been reported in association with BMPR1A child has symptoms.92 In asymptomatic patients with
pathogenic variants.67,87–91 Estimates of gastrointestinal SMAD4 pathogenic variants, it is prudent to assess the
cancer risk range from 11%68 to 55%,89 but none of the upper tract between the ages of 12 and 15 years. At
studies are prospective long-term studies (which would the time of this publication, it is uncertain as to
lead to underestimates), and all are prone to referral- whether BMPR1A pathogenic variants are associated
based ascertainment bias (overestimates). with gastric cancer risk, and that pending new
evidence, upper endoscopy surveillance is suggested at
Question: At what age should colonoscopic and up- intervals similar to those recommended for SMAD4
per endoscopic surveillance begin in individuals identi- carriers. In adults, partial or complete gastrectomy is
fied with juvenile polyposis syndrome? indicated in patients with gastric cancer, high-grade
Recommendation: We suggest initiating colono- dysplasia, inability to adequately survey or endoscopi-
scopic and upper endoscopic surveillance at age 12– cally control polyposis, persistent anemia or gastrointes-
15 years, or earlier if symptomatic. Surveillance should tinal bleeding from gastric polyposis or angioectasia,
be repeated every 1–3 years depending on polyp symptoms of gastric outlet obstruction, or protein-
burden. (Weak recommendation, low quality of evi- losing gastropathy.63,64
dence)

Management of polyposis Question: Which patients with juvenile polyposis


The goal of surveillance in JPS is to mitigate symptoms syndrome should undergo screening for hereditary
related to the disorder and decrease the risk of complica- hemorrhagic telangiectasia?
tions from the manifestations, including cancer. Colonos- Recommendation: We suggest patients with
copy should be first performed at age 12–15 years and SMAD4 pathogenic variants be clinically evaluated for
repeated every 1–3 years, depending on polyp burden hereditary hemorrhagic telangiectasia at the time of
found, with removal of all polyps when feasible or at least the diagnosis, including screening for and appropriate
all polyps 5 mm. Upper endoscopy should be first per- management of cerebral and pulmonary arteriovenous
formed at age 12–15 years and repeated every 1–3 years malformations. (Weak recommendation, low quality
depending on polyp burden found, with removal of of evidence)
polyps 5 mm. Due to the possible presence of intestinal

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1039


Gastrointestinal hamartomatous polyposis syndromes

Management of juvenile polyposis syndrome– PHTS includes a variety of phenotypic variations known
hereditary hemorrhagic telangiectasia as CS, BRRS, and Proteus syndrome. The clinical diagnosis
As recommended by HHT Foundation International, of the PHTS is made in patients meeting the revised diag-
patients with SMAD4 pathogenic variants should be nostic criteria, which include the presence of hamartomas
screened for vascular findings associated with HHT.79,93 of the skin and gastrointestinal tract (see Figures 1D and
Children with possible or confirmed HHT should be 2F–H), mucocutaneous lesions, macrocephaly, and an
screened for brain AVMs at the time of diagnosis and increased risk of benign and malignant lesions of the
undergo at least 1 follow-up MRI at puberty because brain breast, thyroid, and endometrium.95
AVM development appears to correlate with times of
growth. Lung AVM screening and surveillance is recom- Diagnosis
mended at diagnosis and then every 3–5 years with pulse The genetic diagnosis of PHTS is established with a
oximetry testing and consideration of transthoracic germline pathogenic variant in the phosphatase and
contrast echocardiogram. In adulthood, surveillance tensin homolog (PTEN) gene. The clinical criteria for the
should include annual hemoglobin or hematocrit for all diagnosis of CS is complex, and can be found at the
patients older than 35 years. Transthoracic contrast echo- National Comprehensive Cancer Network website.49 The
cardiogram as a screen for pulmonary AVMs should be per- discussion for this article will focus on the manifestations
formed at the time of diagnosis, within 5 years preceding and management of patients with a confirmed diagnosis
planned pregnancy, after pregnancy, and otherwise every of PHTS.
5–10 years. Physicians should consider referring these pa-
tients to HHT Centers of Excellence for this evaluation. Genetics
Brain MRI with and without contrast should be performed
at birth or at the time of diagnosis to screen for cerebral Question: Which organs should undergo surveil-
vascular malformations. lance for cancer when caring for a patient with PTEN ha-
martoma tumor syndrome?
Chemoprevention Recommendation: In PTEN hamartoma tumor syn-
No known effective chemoprevention strategies exist drome, patients are at increased risk for cancer in mul-
for the development of polyposis in patients with JPS. tiple organs, including cancer of the breast, thyroid,
kidney, uterus, colon, and skin.
Summary Given this risk, we recommend a multidisciplinary
BMPR1A and SMAD4 gene alterations are responsible for approach to cancer surveillance in these organs. (Strong
JPS, yet only 60% of patients will have a pathogenic alter- recommendation, low quality evidence)
ation identified. The clinical overlap syndromes include
JPS-HHT in patients with a SMAD4 pathogenic variant and
juvenile polyposis of infancy in patients with a combined The PTEN gene encodes a dual-function phosphatase
BMPR1A and PTEN deletion. The symptoms of JPS are usu- that negatively regulates the growth-promoting activity of
ally related to bleeding from colorectal polyposis or in the phosphatidylinositol 3-kinase pathway. The PHTS fam-
SMAD4-related JPS to gastric polyposis or manifestations ily of syndromes are caused by inactivating pathogenic
of HHT. Cancer risk in JPS is elevated, mainly in the colon variants in PTEN, making it a classic tumor suppressor
and stomach (39%–68%), and is largely associated with gene.96 Germline pathogenic variants in the PTEN gene
SMAD4 pathogenic variants.94 Excess risk of lead to heterogeneous phenotypes called PHTS, which
nongastrointestinal cancer is not reported in JPS. includes CS, BRRS, and some cases of the PTEN-related
Proteus syndrome (not considered further here).97,98
PTEN-HAMARTOMA TUMOR SYNDROME Germline pathogenic variants in PTEN are associated
with other systemic nonpolyposis phenotypes, including
Clinical features abnormalities of the central nervous system and skeleton,
but these will not be discussed here in detail. Patients
Question: Which gastrointestinal findings should with CS are also at risk for dysplastic cerebellar gangliocy-
prompt a genetic evaluation for PTEN hamartoma tu- toma (Lhermitte-Duclos disease), but the specific genetic
mor syndrome? basis of this and the other protean manifestations of germ-
Recommendation: We recommend individuals line pathogenic variants in PTEN are not yet understood.
with multiple gastrointestinal hamartomas or ganglio- Interestingly, gain-of-function germline pathogenic variants
neuromas undergo genetic evaluation for Cowden’s in the WWP1 gene, an E3 ubiquitin ligase commonly up-
syndrome and related conditions. (Strong recommen- regulated in cancers, inhibits the activity of PTEN, causes
dation, low quality of evidence) a CS-like syndrome (oligopolyposis and cancer-prone
phenotype), and provides some insight into what may be

1040 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

responsible for PHTS in the absence of germline variants in lungs. An earlier report from this group on 156 patients
PTEN.99 reported that benign gastrointestinal polyps were found
in 31% of patients at a mean age of 38 years (range, 18–
Cancer risk 62 years) and most were considered “hamartomas.”105
Cancer risks for patients with germline pathogenic vari- The cumulative risk of developing polyps was 70% by
ants in PTEN are very high. The International Cowden Con- age 60 years. The cumulative risk of developing CRC was
sortium reported in 2012 on 368 individuals with germline 18% by age 60 years (occurring between ages 53 and 62
pathogenic variants in PTEN. Elevated standardized inci- years), suggesting that surveillance colonoscopy might
dence ratios (SIRs) were found for carcinomas of the not be necessary in early adult life.
breast (SIR, 25.4; 95% CL, 19.8 and 32.0), thyroid (SIR,
51.1; 95% CL, 38.1 and 67.1), endometrium (SIR, 42.9; Gastrointestinal polyps and polyposis
95% CL, 28.1 and 62.8), colorectum (SIR, 10.3; 95% CL, Gastrointestinal polyps are frequently found in patients
5.6 and 17.4), kidney (SIR, 30.6; 95% CL, 17.8 and 49.4), with germline pathogenic variants in PTEN, with variable
and melanoma (SIR, 8.5; 95% CL, 4.1 and 15.6). This led prevalence. Estimates suggest that 35%–93% of patients
to cumulative lifetime risks for cancer of the breast at with CS have gastrointestinal polyps, but in some reports
85.2% (95% CL, 71.4% and 99.1%), thyroid 35.2% (95% the germline basis of the disease was not known and a uni-
CL, 19.7% and 50.7%), endometrium 28.2% (95% CL, form interpretation of nonadenomatous polyps had not
17.1% and 39.3%), colorectum 9.0% (95% CL, 3.8% and been established.103,106 The wide range of estimated polyp
14.1%), kidney 33.6% (95% CL, 10.4% and 56.9%), and mel- prevalence is probably a reflection of the heterogeneity of
anoma 6% (95% CL, 1.6% and 9.4%).100 the disease and challenges in making definitive
A multicenter study from France estimated cancer risks diagnoses outside of germline tests. Gastrointestinal
in 154 patients with a germline pathogenic variant in polyps include hyperplastic polyps, inflammatory polyps,
PTEN.101 SIRs for female breast cancer were 39.1 (95% ganglioneuromas, lipomas, adenomas, and the nonspecific
confidence interval [CI], 24.8–58.6), thyroid cancer 43.2 term hamartomas.
(95% CI, 19.7–82.1) in women and 199.5 (95% CI, 106.39– A prospective study of 127 PTEN pathogenic variant car-
342.03) in men, melanoma 28.3 (96% CI, 7.6–35.4) in riers from 2548 subjects in an International Cowden Con-
women and 39.4 (95% CI, 10.6–100.9) in men, and sortium study in which 69 underwent endoscopic studies
endometrial cancer 48.7 (95% CI, 9.8–142.3). Estimated found that 64 (93%) had gastrointestinal polyps.103 Of
cumulative lifetime risks by age 70 years were 85% for any that group, one-half had hyperplastic polyps, but all of
cancer, 77% for female breast cancer, and 38% for thyroid the above-listed polyps (plus adenomas) were also found.
cancer. Median age for developing cancer was 36 years. The number of polyps ranged from 1 to “innumerable,”
Although 85% were reported to have colonic polyps of and were distributed throughout the gut.
any variety, risks for gastrointestinal cancer were not BRRS is caused by germline pathogenic variants in the
reported to be elevated in this series. However, a review PTEN gene.98,107 It is a pediatric condition associated
of 211 patients with CS from the Mayo Clinic, including a with macrocephaly, developmental delay, gastrointestinal
review of the literature, reported a 16% lifetime risk for hamartomatous polyps, and pigmented macules on the
CRC. In this study, only 46% had a proven germline toes and glans penis. It has also been called the
pathogenic variant in PTEN, and the cumulative risk for Bannayan-Zonana or Ruvalcaba-Riley-Smith syndrome.
any cancer by age 70 years was 89%.102 Inexplicably, members of the same family may have fea-
In a study of 2548 patients who met “relaxed” Interna- tures of either BRRS or CS,108 and some cases of BRRS
tional Cowden’s Consortium criteria for CS with 5 or do not have detectable pathogenic variants in PTEN.109
more gastrointestinal polyps in which at least 1 was consid- Confusing overlap between CS and JPS can occur when,
ered hyperplastic or hamartomatous, germline pathogenic as mentioned above, a chromosomal deletion occurs on
variants in PTEN were found in 127 (5%).103 At endoscopy, chromosome 10q22.3-q24.1. This leads to the loss of
gastrointestinal polyps were found throughout the gut, and both PTEN and BMPR1A28; a situation that can also look
one-half were considered “hyperplastic.” In this group, like BRRS, also known as juvenile polyposis of infancy
13% of those who underwent colonoscopy developed (see section on JPS).110,111
CRC (7.1% of the entire series of patients), all before age In a smaller study of 19 patients with CS (in which only
50 years (the youngest was 35 years old), with a SIR for 12 were shown to have germline pathogenic variants in
CRC of 224.1 (95% CI, 109.3–411.3). PTEN), pan-colonic polyps were found in 58%, pan-
A multicenter study from 9 countries of 180 carriers of gastrointestinal polyps in 42%, and the pathological inter-
germline pathogenic variants in PTEN estimated the cumu- pretation of the polyps included inflammatory polyps in
lative risk of any cancer or Lhermitte-Duclos disease by age 95%, but also adenomas, lipomas, and ganglioneuromas.112
60 years was 56% for men and 87% for women.104 Moreover, there was more than 1 pathological variety in
Increased risk was reported for cancers of the breast, 79% of patients, indicating the clinical heterogeneity in
thyroid, endometrium, skin, kidneys, colorectum, and this entity. Esophageal glycogenic acanthosis is also

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1041


Gastrointestinal hamartomatous polyposis syndromes

found in PHTS, which is a benign condition and there is no Endometrial cancer


reported increased risk for esophageal cancer.113 The management of endometrial cancer risk begins be-
tween the ages of 30 and 35 years. Endometrial cancer can
Question: What is the recommended colonoscopic often be detected early on the basis of symptoms, and
surveillance in individuals identified with PTEN hamar- women should be educated regarding seeking medical atten-
toma tumor syndrome? tion on the basis of symptoms, including abnormal uterine
Recommendation: We suggest colonoscopy sur- bleeding or postmenopausal bleeding. Endometrial biopsy
veillance to begin at age 35 years (or 10 years younger is sensitive and specific for endometrial cancer and surveil-
than age of any relative with colorectal cancer), lance via biopsy every 1–2 years can be considered.33,49
repeated at intervals no greater than 5 years, depending
on polyp burden. (Weak recommendation, low quality Thyroid cancer
of evidence) An annual thyroid ultrasound should be performed,
beginning at the time of PHTS diagnosis (including in
childhood).33,49
Surveillance of affected individuals
Surveillance recommendations are provided in Table 4. Renal cancer
The assessment at diagnosis of CS/PHTS should include a The recommended management of kidney cancer risk is
complete (and especially dermatologic and neurologic) an annual renal ultrasound and/or renal MRI starting at age
clinical examination, mammography and breast MRI, 40 years if there is a family history of renal cancer, or every
thyroid ultrasound, transvaginal ultrasound, upper 2 years if not.33,49
gastrointestinal endoscopy, colonoscopy, and renal
ultrasound. Although there are no data regarding risk- Melanoma
reduction surgery in women with CS, the option for risk- The management of melanoma risk includes an annual
reducing mastectomy and hysterectomy should be dis- clinical skin examination beginning at age 18 years.33
cussed on a case-by-case basis.
The risk of CRC in patients with CS is estimated to be up Summary
to 9%–18% lifetime risk with mean age at diagnosis of 44 Germline pathogenic variants in PTEN are associated
years, but ranging from 35 to 49 years.100,102,103,105,114,115 with variety of gastrointestinal hamartomatous polyps
Consequently, expert opinion recommends colonoscopy and a markedly increased risk of cancer, but largely in
starting at age 35 years unless symptomatic or if a close nongastrointestinal organs. There is a moderate increase
relative has had colon cancer before age 40 years, then in the risk of CRC, but it may be that the age of risk is
start 10 years before the earliest known colon cancer in such that colonoscopic surveillance can be withheld until
the family and repeat every 5 years or more frequently if age 35 years or later or 10 years younger than the age
the patient is symptomatic or polyps are found. This of the youngest relative with CRC. However, the reports
recommendation differs from a recent American College of CRC risk have been variable,95,116 and no surveillance
of Gastroenterology Guideline,32 which recommends recommendation has been rigorously evaluated. The
initiating colonoscopy at age 15 years. Recent evidence principal organs at risk for cancer include the breast,
suggesting later onset of significant colon cancer risk thyroid, endometrium, and colorectum.
informed our recommendations.
Colectomy is rarely required in patients with CS and is HEREDITARY MIXED POLYPOSIS SYNDROME
reserved for patients with CRC or in whom surveillance
and clearing of premalignant lesions is not endoscopically The genetic basis of the disease and cancer risk
feasible. HMPS is a rare autosomal dominant disease reported in
only a few families. It is characterized by attenuated colonic
polyposis. The polyps include adenomas, hyperplastic, and
Breast cancer a particular polyp with an admixture of variable histologies
The management of breast cancer risk begins at age 25 including adenomatous, hyperplastic, juvenile, and mixed
years with clinical breast examination every 6–12 months; polyps.117 In the original HMPS kindred described by
annual mammography and breast MRI surveillance starting Whitelaw et al,118 13 members were diagnosed with CRC
between ages 30 and 35 years or 5–10 years before the and 23 developed multiple polyps of several different
earliest known breast cancer in the family.49 In patients histologic types (adenomatous, hyperplastic, and
with PHTS, referral to a breast cancer specialist for juvenile). HMPS has been associated with large
management of breast cancer surveillance is reasonable duplications of the promoter region or entire GREM1
and a multidisciplinary approach, including a breast gene.119–121 These unusual pathogenic variants increase
surgeon, is recommended when prophylactic mastectomy the expression of the gene product, which then inhibits
is being considered. the bone morphogenetic protein (BMP) pathway.119 This

1042 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

TABLE 5. Areas requiring further investigation in hamartomatous polyposis syndromes


1. Discover the germline variants in families with clinically recognizable syndromes who do not have germline mutations in the genes known to be
associated with these syndromes.
2. Understand the effects of haplo-insufficiency of STK11 in PJS and the SMAD4 in JPS on the development of polyposis.
3. Identify safe and effective pharmacological interventions for the inhibition of polyp formation children and adults with the hamartomatous pol-
yposis syndromes.
4. Determine whether pharmacological intervention can mitigate cancer risk in PJS and JPS in adults (independent of the polyposis risk).
5. Develop simulation models in adults with a hamartomatous polyposis syndrome to determine the optimal endoscopic frequency.
6. Design and implement studies to determine the best imaging modalities and surveillance intervals in adults with a hamartomatous polyposis
syndrome.
7. Determine whether the hamartomatous phenotype in pediatric patients predicts the cancer risk in adulthood.
8. Perform outcomes studies evaluating the transition of care in pediatric polyposis patients entering adult life, including outcomes such as compli-
ance with surveillance.
9. Determine difference in phenotype and cancer risk in individuals who meet clinical criteria, but do not have an identifiable pathogenic variant
compared to those with a germline pathogenic variant in a hamartomatous polyposis syndrome.
10. Determine the lifetime cancer risks in patients with a pathogenic variant, but without clinical manifestations (those identified incidentally on multi-
gene panel testing).
11. Quantify the frequency of mosaicism as a cause of the hamartomatous polyposis syndromes.
12. Determine the optimal modality of small bowel evaluation in individuals with PJS.

promoter duplication was found in 0.7% of Ashkenazi Jews myeloproliferative disease) may have therapeutic poten-
in Israel who met clinical criteria for Lynch syndrome.120 tial in patients with PJS.21 Also, LKB1 activity inhibits the
The largest series reported 4 families with 16 affected activation of adenosine monophosphate–dependent pro-
members; the onset of polyposis starts in the late 20s, tein kinase. Loss of LKB1 activity results in adenosine
which is when colonoscopic surveillance should begin.122 monophosphate–dependent protein kinase activation
There are not enough data to know the optimal with up-regulation of mTORC1 signaling contributing
surveillance intervals or whether extraintestinal neoplasia to growth of PJS polyposis. Targeting of adenosine
is a risk. The underlying genetic basis of the majority of monophosphate–dependent protein kinase activation is
HMPS families is unknown. currently being investigated.123

FUTURE RESEARCH CONSIDERATIONS Juvenile polyposis syndrome


Of note is the mouse model of juvenile polyposis
The hamartoma syndromes are rare and it is difficult created by a conditional knockout of the SMAD4 gene
for single institutions or even collaborative centers to only in T-cell populations.73 These animals spontaneously
accumulate enough cases and follow them prospectively developed massive polyps within the gastroduodenal
long enough to develop robust conclusions about cancer region. The epithelium in the polyps contained increased
risk. Modeling, simulation, and collaborative multicenter expression of pro-inflammatory cytokines, particularly
clinical studies can be used to help clarify the benefits interleukin-11, a cytokine known to promote gastric
and risks of various interventions and surveillance pro- epithelial cell survival and hyperplasia with evidence of
grams. The management of these diseases changes increased TH17 cell activity. This suggests possible thera-
dramatically when the patient matures from childhood peutic targets for the future chemoprevention of juvenile
to adulthooddwhich is where almost all of the cancer polyposis.
risk lies. Important knowledge gaps are listed in Table 5
and expanded upon below. SUMMARY AND CONCLUSIONS

Peutz-Jeghers syndrome Among the gastrointestinal hamartomatous polyposis


As indicated above, STK11/LKB1 pathogenic variants syndromes, PJS is the best understood. The polyps are
result in up-regulation of inflammatory cytokines and readily identified as PJ polyps pathologically, and only 1
promotion of overgrowth of both stromal and normal known gene (STK11) is associated with this entity. The
gastrointestinal epithelium, driving polyp formation phenotype is clinically distinct. Not all patients (or fam-
(Table 5). Up-regulation of cytokines is associated with ilies) have detectable germline pathogenic variants in
hyperactivation of JAK-STAT, which contributes to STK11, so additional genes in this pathway are possibly
inflammation and cancer. Inhibition of JAK-STAT signifi- contributors to this entity. The polyps may evolve
cantly reduced polyp growth in mice. Consequently, the through an expansion of elements of the gut stroma
JAK inhibitor ruxolitinib (already clinically approved for due to haploinsufficiency of STK11. In children, the

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1043


Gastrointestinal hamartomatous polyposis syndromes

main risks are for gastrointestinal obstruction and 3. Hampel H, Bennett RL, Buchanan A, et al. A practice guideline from
bleeding. Later in adult life, a very high risk of intestinal the American College of Medical Genetics and Genomics and the Na-
tional Society of Genetic Counselors: referral indications for cancer
and extraintestinal cancers exists. The major organs at predisposition assessment. Genet Med 2015;17:70-87.
risk for cancer (in order of decreasing relative risk) 4. Heald B, Hampel H, Church J, et al. Collaborative Group of the Amer-
include the small intestine, stomach, pancreas, colon, icas on Inherited Gastrointestinal Cancer Position statement on multi-
esophagus, ovary, lung, uterus, and breast, with estimated gene panel testing for patients with colorectal cancer and/or
lifetime risks for any cancer reaching >90%. The ovaries polyposis. Fam Cancer 2020;19:223-39.
5. Attard TM, Burke CA, Hyer W, et al. ACG clinical report and recom-
and testes are also at risk for rare variant tumors. The mendations on transition of care in children and adolescents with he-
screening and surveillance strategies change dramatically reditary polyposis syndromes. Am J Gastroenterol 2021;116:638-46.
when the children reach adulthood. 6. van Lier MG, Wagner A, Mathus-Vliegen EM, et al. High cancer risk in
JPS has principal risks for obstruction and bleeding in Peutz-Jeghers syndrome: a systematic review and surveillance recom-
the pediatric ages, but the management in adults shifts mendations. Am J Gastroenterol 2010;105:1258-64; author reply 1265.
7. Latchford AR, Phillips RK. Gastrointestinal polyps and cancer in Peutz-
to cancer risks in the stomach and colon. Importantly, un- Jeghers syndrome: clinical aspects. Fam Cancer 2011;10:455-61.
like PJS, the gastrointestinal cancers tend to arise within 8. Shaco-Levy R, Jasperson KW, Martin K, et al. Morphologic character-
the juvenile polyps, suggesting that polyp removal might ization of hamartomatous gastrointestinal polyps in Cowden syn-
prevent cancer. Cancer risk is linked to germline patho- drome, Peutz-Jeghers syndrome, and juvenile polyposis syndrome.
genic variants in the SMAD4 gene rather than the BMPR1A Hum Pathol 2016;49:39-48.
9. Menko FH. LKB1/ STK11, Peutz-Jeghers syndrome and cancer. Fam
gene. The polyps are histologically inflammatory in nature, Cancer 2011;10:413-4.
and their growth may be driven by haploinsufficiency of 10. Jass JR. Colorectal polyposes: from phenotype to diagnosis. Pathol
SMAD4 in the immune cells of the polyp stroma. The man- Res Pract 2008;204:431-47.
agement focuses on the polyps in children and cancers 11. Beggs AD, Latchford AR, Vasen HF, et al. Peutz-Jeghers syndrome: a
later in life, when endoscopic approaches are the mainstay systematic review and recommendations for management. Gut
2010;59:975-86.
of surveillance. 12. Amos CI, Bali D, Thiel TJ, et al. Fine mapping of a genetic locus for Peutz-
The hamartoma syndromes associated with germline Jeghers syndrome on chromosome 19p. Cancer Res 1997;57:3653-6.
pathogenic variants in PTEN raise a completely different 13. Hemminki A, Markie D, Tomlinson I, et al. A serine/threonine kinase
clinical challenge. Although nearly all patients with PHTS gene defective in Peutz-Jeghers syndrome. Nature 1998;391:184-7.
have a variety of different hamartomatous gastrointestinal 14. Hardie DG. The LKB1-AMPK pathway-friend or foe in cancer? Cancer
Cell 2013;23:131-2.
polyps, studies suggest an increased risk of cancer in this 15. Shaw RJ, Bardeesy N, Manning BD, et al. The LKB1 tumor suppressor
setting.100,103 The extracolonic cancer risks are greatest negatively regulates mTOR signaling. Cancer Cell 2004;6:91-9.
for the breast, thyroid, melanoma, and endometrium. 16. Knudson AG Jr. Mutation and cancer: statistical study of retinoblas-
The absolute risk for CRC appears to be increased, toma. Proc Natl Acad Sci U S A 1971;68:820-3.
ranging from 9% to 18% during a lifetime, and the rest of 17. Hernan I, Roig I, Martin B, et al. De novo germline mutation in the
serine-threonine kinase STK11/LKB1 gene associated with Peutz-
the gastrointestinal tract does not have an established Jeghers syndrome. Clin Genet 2004;66:58-62.
increase in cancer risk. The age for beginning 18. Katajisto P, Vaahtomeri K, Ekman N, et al. LKB1 signaling in mesen-
surveillance for CRC remains to be determined, but the chymal cells required for suppression of gastrointestinal polyposis.
early onset of CRC provides some suggestions. Be aware Nat Genet 2008;40:455-9.
that the PTEN gene and BMPR1A are located near one 19. Poffenberger MC, Metcalfe-Roach A, Aguilar E, et al. LKB1 deficiency
in T cells promotes the development of gastrointestinal polyposis.
another on chromosome 10q, and that large-scale chromo- Science 2018;361:406-11.
somal deletions could adversely affect both genes, compli- 20. Ollila S, Domenech-Moreno E, Laajanen K, et al. Stromal Lkb1 defi-
cating the clinical picture. Long-term prospective studies of ciency leads to gastrointestinal tumorigenesis involving the IL-11-
mutation carriers are still needed to further clarify the risk JAK/STAT3 pathway. J Clin Invest 2018;128:402-14.
of cancer and the role of surveillance in these syndromes. 21. Hollstein PE, Shaw RJ. Inflamed T cells and stroma drive gut tumors.
Science 2018;361:332-3.
With increases in genetic testing and evaluation, future 22. Amos CI, Keitheri-Cheteri MB, Sabripour M, et al. Genotype-pheno-
studies will be conducted with more robust cohorts of type correlations in Peutz-Jeghers syndrome. J Med Genet 2004;41:
genetically characterized, less heterogeneous populations. 327-33.
However, there is also a need to study patients and families 23. Borun P, De Rosa M, Nedoszytko B, et al. Specific Alu elements
with unusual phenotypes where no genotype can be involved in a significant percentage of copy number variations of
the STK11 gene in patients with Peutz-Jeghers syndrome. Fam Can-
found.
cer 2015;14:455-61.
24. Zbuk KM, Eng C. Hamartomatous polyposis syndromes. Nat Clin Pract
Gastroenterol Hepatol 2007;4:492-502.
REFERENCES 25. Idrees MT, Ulbright TM, Oliva E, et al. The World Health Organization
2016 classification of testicular non-germ cell tumours: a review and
1. Guyatt GH, Oxman AD, Kunz R, et al. What is “quality of evidence” and update from the International Society of Urological Pathology Testis
why is it important to clinicians? BMJ 2008;336:995-8. Consultation Panel. Histopathology 2017;70:513-21.
2. Lu KH, Wood ME, Daniels M, et al. American Society of Clinical 26. Giardiello FM, Brensinger JD, Tersmette AC, et al. Very high risk of
Oncology Expert Statement: collection and use of a cancer family his- cancer in familial Peutz-Jeghers syndrome. Gastroenterology
tory for oncology providers. J Clin Oncol 2014;32:833-40. 2000;119:1447-53.

1044 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

27. Resta N, Pierannunzio D, Lenato GM, et al. Cancer risk associated with 49. Daly MB, Pilarski R, Yurgelun MB, et al. NCCN Guidelines Insights: Ge-
STK11/LKB1 germline mutations in Peutz-Jeghers syndrome netic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic,
patients: results of an Italian multicenter study. Dig Liver Dis Version 1.2020. J Natl Compr Canc Netw 2020;18:380-91.
2013;45:606-11. 50. Manahan ER, Kuerer HM, Sebastian M, et al. Consensus Guidelines on
28. Hearle N, Schumacher V, Menko FH, et al. Frequency and spectrum of Genetic` Testing for Hereditary Breast Cancer from the American So-
cancers in the Peutz-Jeghers syndrome. Clin Cancer Res 2006;12: ciety of Breast Surgeons. Ann Surg Oncol 2019;26:3025-31.
3209-15. 51. Korsse SE, Harinck F, van Lier MG, et al. Pancreatic cancer risk in
29. van Lier MG, Westerman AM, Wagner A, et al. High cancer risk and Peutz-Jeghers syndrome patients: a large cohort study and implica-
increased mortality in patients with Peutz-Jeghers syndrome. Gut tions for surveillance. J Med Genet 2013;50:59-64.
2011;60:141-7. 52. Chen KT. Female genital tract tumors in Peutz-Jeghers syndrome. Hu-
30. Lim W, Olschwang S, Keller JJ, et al. Relative frequency and man Pathol 1986;17:858-61.
morphology of cancers in STK11 mutation carriers. Gastroenterology 53. Canto MI, Harinck F, Hruban RH, et al. International Cancer of the
2004;126:1788-94. Pancreas Screening (CAPS) Consortium summit on the management
31. Mehenni H, Resta N, Park JG, et al. Cancer risks in LKB1 germline mu- of patients with increased risk for familial pancreatic cancer. Gut
tation carriers. Gut 2006;55 984–590. 2013;62:339-47.
32. Chen HY, Jin XW, Li BR, et al. Cancer risk in patients with Peutz- 54. Canto MI, Almario JA, Schulick RD, et al. Risk of neoplastic progression
Jeghers syndrome: a retrospective cohort study of 336 cases. Tumour in individuals at high risk for pancreatic cancer undergoing long-term
Biol 2017;39(6) 1010428317705131. surveillance. Gastroenterology 2018;155:740-51.e2.
33. Syngal S, Brand RE, Church JM, et al. ACG clinical guideline: genetic 55. Goggins M, Overbeek KA, Brand R, et al. Management of patients
testing and management of hereditary gastrointestinal cancer syn- with increased risk for familial pancreatic cancer: updated recom-
dromes. Am J Gastroenterol 2015;110:223-62; quiz 263. mendations from the International Cancer of the Pancreas Screening
34. Gupta S, Provenzale D, Regenbogen SE, et al. NCCN Guidelines In- (CAPS) Consortium. Gut 2020;69:7-17.
sights: Genetic/Familial High-Risk Assessment: Colorectal, Version 56. McGowan L, Young RH, Scully RE. Peutz-Jeghers syndrome with "ad-
3.2017. J Natl Compr Cancer Netw 2017;15:1465-75. enoma malignum" of the cervix. A report of two cases. Gynecol Oncol
35. Giardiello FM, Trimbath JD. Peutz-Jeghers syndrome and manage- 1980;10:125-33.
ment recommendations. Clin Gastroenterol Hepatol 2006;4:408-15. 57. Crispo A, Brennan P, Jockel KH, et al. The cumulative risk of lung can-
36. Utsunomiya J, Gocho H, Miyanaga T, et al. Peutz-Jeghers syndrome: cer among current, ex- and never-smokers in European men. Br J
its natural course and management. Johns Hopkins Med J Cancer 2004;91:1280-6.
1975;136:71-82. 58. Pinsky PF, Zhu CS, Kramer BS. Lung cancer risk by years since quitting
37. Hinds R, Philp C, Hyer W, et al. Complications of childhood Peutz- in 30þ pack year smokers. J Med Screen 2015;22:151-7.
Jeghers syndrome: implications for pediatric screening. J Pediatr Gas- 59. Detterbeck FC, Mazzone PJ, Naidich DP, et al. Screening for lung can-
troenterol Nutr 2004;39:219-20. cer: diagnosis and management of lung cancer, 3rd ed: American Col-
38. Fallon SC, Lopez ME, Zhang W, et al. Risk factors for surgery in pedi- lege of Chest Physicians evidence-based clinical practice guidelines.
atric intussusception in the era of pneumatic reduction. J Pediatr Surg Chest 2013;143(Suppl):e78S-92.
2013;48:1032-6. 60. Wender R, Fontham ET, Barrera E Jr, et al. American Cancer Society
39. Gastineau S, Viala J, Caldari D, et al. Contribution of capsule endos- lung cancer screening guidelines. CA Cancer J Clin 2013;63:107-17.
copy to Peutz-Jeghers syndrome management in children. Dig Liver 61. de Brabander J, Eskens F, Korsse SE, et al. Chemoprevention in pa-
Dis 2012;44:839-43. tients with Peutz-Jeghers syndrome: lessons learned. Oncologist
40. Bizzarri B, Borrelli O, de'Angelis N, et al. Management of duodenal- 2018;23:399.e33.
jejunal polyps in children with peutz-jeghers syndrome with single- 62. Udd L, Katajisto P, Rossi DJ, et al. Suppression of Peutz-Jeghers polyp-
balloon enteroscopy. J Pediatr Gastroenterol Nutr 2014;59:49-53. osis by inhibition of cyclooxygenase-2. Gastroenterology 2004;127:
41. Latchford AR, Neale K, Phillips RK, et al. Peutz-Jeghers syndrome: 1030-7.
intriguing suggestion of gastrointestinal cancer prevention from sur- 63. Chow E, Macrae F. A review of juvenile polyposis syndrome.
veillance. Dis Colon Rectum 2011;54:1547-51. J Gastroenterol Hepatol 2005;20:1634-40.
42. Tomlinson IP, Houlston RS. Peutz-Jeghers syndrome. J Med Genet 64. Schreibman IR, Baker M, Amos C, et al. The hamartomatous polyposis
1997;34:1007-11. syndromes: a clinical and molecular review. Am J Gastroenterol
43. Goverde A, Korsse SE, Wagner A, et al. Small-bowel surveillance in pa- 2005;100:476-90.
tients with Peutz-Jeghers syndrome: comparing magnetic resonance 65. Latchford AR, Neale K, Phillips RK, et al. Juvenile polyposis syndrome:
enteroclysis and double balloon enteroscopy. J Clin Gastroenterol a study of genotype, phenotype, and long-term outcome. Dis Colon
2017;51:e27-33. Rectum 2012;55:1038-43.
44. McGrath DR, Spigelman AD. Preventive measures in Peutz-Jeghers 66. Heald B, Rigelsky C, Moran R, et al. Prevalence of thoracic aortopathy
syndrome. Fam Cancer 2001;1:121-5. in patients with juvenile polyposis syndrome-hereditary hemorrhag-
45. Ohmiya N, Taguchi A, Shirai K, et al. Endoscopic resection of Peutz- ic telangiectasia due to SMAD4. Am J Med Genet A 2015;167A:
Jeghers polyps throughout the small intestine at double-balloon 1758-62.
enteroscopy without laparotomy. Gastrointest Endosc 2005;61: 67. Aretz S, Stienen D, Uhlhaas S, et al. High proportion of large
140-7. genomic deletions and a genotype phenotype update in 80 unre-
46. Oncel M, Remzi FH, Church JM, et al. Benefits of ’clean sweep’ in lated families with juvenile polyposis syndrome. J Med Genet
Peutz-Jeghers patients. Colorectal Dis 2004;6:332-5. 2007;44:702-9.
47. van Lier MG, Mathus-Vliegen EM, Wagner A, et al. High cumulative 68. Aytac E, Sulu B, Heald B, et al. Genotype-defined cancer risk in juve-
risk of intussusception in patients with Peutz-Jeghers syndrome: nile polyposis syndrome. Br J Surg 2015;102:114-8.
time to update surveillance guidelines? Am J Gastroenterol 69. Sweet K, Willis J, Zhou XP, et al. Molecular classification of patients
2011;106:940-5. with unexplained hamartomatous and hyperplastic polyposis. JAMA
48. Hamdi Y, Soucy P, Kuchenbaeker KB, et al. Association of breast cancer 2005;294:2465-73.
risk in BRCA1 and BRCA2 mutation carriers with genetic variants 70. Ngeow J, Heald B, Rybicki LA, et al. Prevalence of germline PTEN,
showing differential allelic expression: identification of a modifier of BMPR1A, SMAD4, STK11, and ENG mutations in patients with
breast cancer risk at locus 11q22.3. Breast Cancer Res Treat 2017;161: moderate-load colorectal polyps. Gastroenterology 2013;144:1402-9;
117-34. 1409.e1–e5.

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1045


Gastrointestinal hamartomatous polyposis syndromes

71. Woodford-Richens K, Bevan S, Churchman M, et al. Analysis of genetic 93. Faughnan ME, Mager JJ, Hetts SW, et al. Second international guide-
and phenotypic heterogeneity in juvenile polyposis. Gut 2000;46: lines for the diagnosis and management of hereditary hemorrhagic
656-60. telangiectasia. Ann Intern Med 2020;173:989-1001.
72. Alberici P, Gaspar C, Franken P, et al. Smad4 haploinsufficiency: a 94. Campos FG, Figueiredo MN, Martinez CA. Colorectal cancer risk in ha-
matter of dosage. Pathogenetics 2008;1:2. martomatous polyposis syndromes. World J Gastrointest Surg 2015;7:
73. Hahn JN, Falck VG, Jirik FR. Smad4 deficiency in T cells leads to the 25-32.
Th17-associated development of premalignant gastroduodenal le- 95. Pilarski R, Burt R, Kohlman W, et al. Cowden syndrome and the PTEN
sions in mice. J Clin Invest 2011;121:4030-42. hamartoma tumor syndrome: systematic review and revised diag-
74. Kim BG, Li C, Qiao W, et al. Smad4 signalling in T cells is required for nostic criteria. J Natl Cancer Inst 2013;105:1607-16.
suppression of gastrointestinal cancer. Nature 2006;441:1015-9. 96. Chalhoub N, Baker SJ. PTEN and the PI3-kinase pathway in cancer.
75. Woodford-Richens K, Williamson J, Bevan S, et al. Allelic loss at Annu Rev Pathol 2009;4:127-50.
SMAD4 in polyps from juvenile polyposis patients and use of fluores- 97. Liaw D, Marsh DJ, Li J, et al. Germline mutations of the PTEN gene in
cence in situ hybridization to demonstrate clonal origin of the epithe- Cowden disease, an inherited breast and thyroid cancer syndrome.
lium. Cancer Res 2000;60:2477-82. Nat Genet 1997;16:64-7.
76. Blatter RH, Plasilova M, Wenzel F, et al. Somatic alterations in juvenile 98. Marsh DJ, Coulon V, Lunetta KL, et al. Mutation spectrum and
polyps from BMPR1A and SMAD4 mutation carriers. Genes Chromo- genotype-phenotype analyses in Cowden disease and Bannayan-
somes Cancer 2015;54:575-82. Zonana syndrome, two hamartoma syndromes with germline PTEN
77. O'Malley M, LaGuardia L, Kalady MF, et al. The prevalence of heredi- mutation. Hum Mol Genet 1998;7:507-15.
tary hemorrhagic telangiectasia in juvenile polyposis syndrome. Dis 99. Lee YR, Yehia L, Kishikawa T, et al. WWP1 Gain-of-function inactiva-
Colon Rectum 2012;55:886-92. tion of PTEN in cancer predisposition. N Engl J Med 2020;382:2103-16.
78. Gallione CJ, Richards JA, Letteboer TG, et al. SMAD4 mutations found 100. Tan MH, Mester JL, Ngeow J, et al. Lifetime cancer risks in individuals
in unselected HHT patients. J Med Genet 2006;43:793-7. with germline PTEN mutations. Clin Cancer Res 2012;18:400-7.
79. Faughnan ME, Palda VA, Garcia-Tsao G, et al. International guidelines 101. Bubien V, Bonnet F, Brouste V, et al. High cumulative risks of cancer in
for the diagnosis and management of hereditary haemorrhagic telan- patients with PTEN hamartoma tumour syndrome. J Med Genet
giectasia. J Med Genet 2011;48:73-87. 2013;50:255-63.
80. Oliveira PH, Cunha C, Almeida S, et al. Juvenile polyposis of infancy in 102. Riegert-Johnson DL, Gleeson FC, Roberts M, et al. Cancer and
a child with deletion of BMPR1A and PTEN genes: surgical approach. Lhermitte-Duclos disease are common in Cowden syndrome patients.
J Pediatr Surg 2013;48:e33-7. Heredit Cancer Clin Pract 2010;8(1):6.
81. Tsuchiya KD, Wiesner G, Cassidy SB, et al. Deletion 10q23.2-q23.33 in 103. Heald B, Mester J, Rybicki L, et al. Frequent gastrointestinal polyps
a patient with gastrointestinal juvenile polyposis and other features and colorectal adenocarcinomas in a prospective series of PTEN mu-
of a Cowden-like syndrome. Genes Chromosomes Cancer 1998;21: tation carriers. Gastroenterology 2010;139:1927-33.
113-8. 104. Nieuwenhuis MH, Kets CM, Murphy-Ryan M, et al. Cancer risk and
82. Delnatte C, Sanlaville D, Mougenot JF, et al. Contiguous gene deletion genotype-phenotype correlations in PTEN hamartoma tumor syn-
within chromosome arm 10q is associated with juvenile polyposis of drome. Fam Cancer 2014;13:57-63.
infancy, reflecting cooperation between the BMPR1A and PTEN 105. Nieuwenhuis MH, Kets CM, Murphy-Ryan M, et al. Is colorectal surveil-
tumor-suppressor genes. Am J Hum Genet 2006;78:1066-74. lance indicated in patients with PTEN mutations? Colorectal Dis
83. Menko FH, Kneepkens CM, de Leeuw N, et al. Variable phenotypes 2012;14:e562-6.
associated with 10q23 microdeletions involving the PTEN and 106. Stanich PP, Owens VL, Sweetser S, et al. Colonic polyposis and
BMPR1A genes. Clin Genet 2008;74:145-54. neoplasia in Cowden syndrome. Mayo Clin Proc 2011;86:489-92.
84. Fox VL, Perros S, Jiang H, et al. Juvenile polyps: recurrence in patients 107. Arch EM, Goodman BK, Van Wesep RA, et al. Deletion of PTEN in a
with multiple and solitary polyps. Clin Gastroenterol Hepatol 2010;8: patient with Bannayan-Riley-Ruvalcaba syndrome suggests allelism
795-9. with Cowden disease. Am J Med Genet 1997;71:489-93.
85. Wu TT, Rezai B, Rashid A, et al. Genetic alterations and epithelial 108. Hendriks YM, Verhallen JT, van der Smagt JJ, et al. Bannayan-Riley-Ru-
dysplasia in juvenile polyposis syndrome and sporadic juvenile valcaba syndrome: further delineation of the phenotype and man-
polyps. Am J Pathol 1997;150:939-47. agement of PTEN mutation-positive cases. Fam Cancer 2003;2:79-85.
86. Brosens LA, van Hattem A, Hylind LM, et al. Risk of colorectal cancer in 109. Carethers JM, Furnari FB, Zigman AF, et al. Absence of PTEN/MMAC1
juvenile polyposis. Gut 2007;56:965-7. germ-line mutations in sporadic Bannayan-Riley-Ruvalcaba syn-
87. Gonzalez RS, Adsay V, Graham RP, et al. Massive gastric juvenile-type drome. Cancer Res 1998;58:2724-6.
polyposis: a clinicopathological analysis of 22 cases. Histopathology 110. Alimi A, Weeth-Feinstein LA, Stettner A, et al. Overlap of juvenile pol-
2017;70:918-28. yposis syndrome and Cowden syndrome due to de novo chromo-
88. Friedl W, Uhlhaas S, Schulmann K, et al. Juvenile polyposis: massive some 10 deletion involving BMPR1A and PTEN: implications for
gastric polyposis is more common in MADH4 mutation carriers than treatment and surveillance. Am J. Med Genet A 2015;167:1305-8.
in BMPR1A mutation carriers. Hum Genet 2002;111:108-11. 111. Hiljadnikova Bajro M, Sukarova-Angelovska E, Adelaide J, et al. A new
89. Howe JR, Mitros FA, Summers RW. The risk of gastrointestinal case with 10q23 interstitial deletion encompassing both PTEN and
carcinoma in familial juvenile polyposis. Ann Surg Oncol 1998;5: BMPR1A narrows the genetic region deleted in juvenile polyposis
751-6. syndrome. J Appl Genet 2013;54:43-7.
90. Wirtzfeld DA, Petrelli NJ, Rodriguez-Bigas MA. Hamartomatous polyp- 112. Shaco-Levy R, Jasperson KW, Martin K, et al. Gastrointestinal polyposis
osis syndromes: molecular genetics, neoplastic risk, and surveillance in cowden syndrome. J Clin Gastroenterol 2017;51:e60-7.
recommendations. Ann Surg Oncol 2001;8:319-27. 113. Borowsky J, Setia N, Rosty C, et al. Spectrum of gastrointestinal tract
91. MacFarland SP, Ebrahimzadeh JE, Zelley K, et al. Phenotypic differ- pathology in a multicenter cohort of 43 Cowden syndrome patients.
ences in juvenile polyposis syndrome with or without a disease- Mod Pathol 2019;32:1814-22.
causing SMAD4/BMPR1A variant. Cancer Prev Res (Phila) 2021;14: 114. Ni Y, He X, Chen J, et al. Germline SDHx variants modify breast and thy-
215-22. roid cancer risks in Cowden and Cowden-like syndrome via FAD/NAD-
92. Cohen S. Management of JPS in children and adolescents: a positon dependant destabilization of p53. Hum Mol Genet 2012;21:300-10.
paper from the ESPGHAN Polyposis Working Group. J Pediatr Gastro- 115. Kato M, Mizuki A, Hayashi T, et al. Cowden’s disease diagnosed
enterol Nutr 2019;68:453-62. through mucocutaneous lesions and gastrointestinal polyposis with

1046 GASTROINTESTINAL ENDOSCOPY Volume 95, No. 6 : 2022 www.giejournal.org


Gastrointestinal hamartomatous polyposis syndromes

recurrent hematochezia, unrevealed by initial diagnosis. Intern Med and Mallinckrodt Pharmaceuticals. Brian C. Jacobson: Motus, GI, Dark
2000;39:559-63. Canyon LLC, and Remedy Partners. Aasma Shaukat: Iterative Scopes, Free-
116. Stanich PP, Pilarski R, Rock J, et al. Colonic manifestations of PTEN ha- nome Holdings Inc. Swati G. Patel: Olympus America, Freenome Holdings
martoma tumor syndrome: case series and systematic review. World J Inc, ERBE USA. Sapna Syngal: Myriad Genetics, Inc, DC Health, Inc, and Glaxo-
Gastroenterol 2014;20:1833-8. Smith Kline, Inc. Douglas Robertson: Covidien, Freenome Holdings, Inc, and
117. Plesec T, Brown K, Allen C, et al. Clinicopathological features of a Amadix. The remaining authors disclose no conflicts.
kindred with SCG5-GREM1-associated hereditary mixed polyposis
syndrome. Hum Pathol 2017;60:75-81. This article is being published jointly in Gastrointestinal Endoscopy, Gastro-
118. Whitelaw SC, Murday VA, Tomlinson IP, et al. Clinical and molecular enterology, and The American Journal of Gastroenterology. The article is
features of the hereditary mixed polyposis syndrome. Gastroenter- identical except for minor stylistic and spelling differences in keeping
ology 1997;112:327-34. with each journal’s style. Citations from any of the 3 journals can be
119. Jaeger E, Leedham S, Lewis A, et al. Hereditary mixed polyposis syn- used when citing this article.
drome is caused by a 40-kb upstream duplication that leads to *Authors share co-first authorship.
increased and ectopic expression of the BMP antagonist GREM1.
Nat Genet 2012;44:699-703. Copyright ª 2022 by the American Society for Gastrointestinal Endoscopy,
120. Laitman Y, Jaeger E, Katz L, et al. GREM1 germline mutation screening the American Gastroenterological Association, and the American College of
in Ashkenazi Jewish patients with familial colorectal cancer. Genet Gastroenterology
Res 2015;97:e11. 0016-5107/$36.00
121. Lewis A, Freeman-Mills L, de la Calle-Mustienes E, et al. A polymorphic https://doi.org/10.1016/j.gie.2022.02.044
enhancer near GREM1 influences bowel cancer risk through differen-
Current affiliations: Division of Gastroenterology, University of California-
tial CDX2 and TCF7L2 binding. Cell Rep 2014;8:983-90.
San Diego School of Medicine, San Diego, California (1), Divisions of
122. Lieberman S, Walsh T, Schechter M, et al. Features of patients with Gastroenterology and Clinical Cancer Genomics, Center for Precision Med-
hereditary mixed polyposis syndrome caused by duplication of icine, City of Hope National Medical Center, Duarte, California (2), The Zane
GREM1 and implications for screening and surveillance. Gastroenter- Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontar-
ology 2017;152:1876-80.e1. io, Canada (3), Division of Gastroenterology, Hepatology and Nutrition,
123. Shackelford DB, Shaw RJ. The LKB1-AMPK pathway: metabolism and Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
growth control in tumour suppression. Nat Rev Cancer 2009;9:563-75. (4), Division of Gastroenterology, Department of Medicine, The Johns Hop-
kins University School of Medicine, Baltimore, Maryland (5), Veterans Af-
fairs Medical Center, White River Junction, Vermont (6), Geisel School of
Medicine at Dartmouth, Hanover, New Hampshire (7), University of Con-
Abbreviations used in this paper: AVM, arteriovenous malformation; BMP, necticut, Farmington, Connecticut (8), Department of Gastroenterology,
bone morphogenetic protein; BRRS, Bannayan-Riley-Ruvalcaba Hepatology and Nutrition, Cleveland Clinic, Cleveland, Ohio (9), Veterans
syndrome; CI, confidence interval; CL, confidence limits; CR, cumulative Affairs Puget Sound Health Care System, Seattle, Washington (10), Univer-
risk; CRC, colorectal cancer; CS, Cowden’s syndrome; EGD, sity of Washington School of Medicine, Seattle, Washington (11), Division
esophagogastroduodenoscopy; GRADE, Grading of Recommendations of Gastroenterology and Hepatology, New York University Langone
Assessment, Development and Evaluation; HHT, hereditary hemorrhagic Health, New York, New York (12), Veterans Affairs Medical Center, San Die-
telangiectasia; HMPS, hereditary mixed polyposis syndrome; JPS, juvenile go, California (13), University of California San Diego, La Jolla, California
polyposis syndrome; MRE, magnetic resonance enterography; MRI, (14), Veterans Affairs San Diego Healthcare System, San Diego, California
magnetic resonance imaging; PHTS, PTEN hamartoma tumor (15), Division of Gastroenterology, Massachusetts General Hospital, Boston,
syndrome; PJS, Peutz-Jeghers syndrome; RR, relative risk; SIR, Massachusetts (16), University of Colorado School of Medicine, Rocky
standardized incidence ratio; USMSTF, U.S. Multi-Society Task Force on Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
Colorectal Cancer; VCE, video capsule endoscopy. (17), Minneapolis Veterans Affairs Health Care System, Minneapolis, Minne-
sota (18), University of Minnesota, Minneapolis, Minnesota (19), Brigham
CONFLICTS OF INTEREST: The authors disclose no conflicts of interest relative and Women’s Hospital, Boston Massachusetts (20), Dana-Farber Cancer
to the current work since 2016. These authors disclose the following industry Institute, Boston Massachusetts (21), Harvard Medical School, Boston Mas-
relationships (consulting, research, reimbursement) without conflict of interest sachusetts (22).
relevant to the current work since 2016: C. Richard Boland: Ambry Genetics.
Gregory Idos: Myriad Genetics, Inc. Carol Burke: Salix Pharmaceuticals, Ferring Correspondence to: C. Richard Boland, MD, University of California San
Pharmaceuticals, Aries Pharmaceuticals, Pfizer, Cancer Prevention Pharma- Diego, 9444 Medical Center Drive, East Campus Office Building, Room 2-
ceuticals, Janssen Pharmaceuticals, SLA Pharma AG, and Freenome Holdings, 065, La Jolla, California 92037. E-mail: crboland@health.ucsd.edu.
Inc. Samir Gupta: Freenome Holdings, Inc, Guardant Health, Inc, CellMax, Inc,

www.giejournal.org Volume 95, No. 6 : 2022 GASTROINTESTINAL ENDOSCOPY 1047


Chinese Journal of Plastic and Reconstructive Surgery 3 (2021) 147–149

Contents lists available at ScienceDirect

Chinese Journal of Plastic and Reconstructive Surgery


journal homepage: www.keaipublishing.com/en/journals/chinese-
journal-of-plastic-and-reconstructive-surgery

A case of labial lentigines in Peutz-Jeghers syndrome treated using a


Q-switched alexandrite laser
Shuai Yang, Hong Xu, Ying Zhang, Changbo Tao, Peisheng Jin *
Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China

A R T I C L E I N F O A B S T R A C T

Keywords: Peutz-Jeghers syndrome (PJS) is a polygenic autosomal dominant disease characterized by multiple gastroin-
Labial lentigines testinal polyps and pigmentation of the mucosa and skin. While there are a few reports regarding successful
Peutz-jeghers syndrome treatment of intestinal polyps in PJS, there is little research regarding treatment of mucocutaneous melanosis.
Q-switched alexandrite laser
This study investigated the many advantages of using a Q-switched alexandrite laser to treat mucocutaneous
Treatment
melanosis. In this case, a 19-year-old male with PJS presented with labial lentigines and received two Q-switched
alexandrite laser treatments in 2018. Subsequently, the efficacy of the treatment was evaluated. The result of the
evaluation was that, after the two laser treatments, the labial lentigines were successfully removed, and there
were no complications.

1. Introduction eventually they extended over the entire lip (Fig. 1). The patient had not
received prior treatment for labial lentigines. Digital photographs were
Peutz-Jeghers syndrome (PJS) is a polygenic autosomal dominant taken before each treatment and at each subsequent visit to evaluate
disease characterized by multiple gastrointestinal polyps, in addition to treatment efficacy.
pigmentation of the mucosa and skin.1–5 In patients with PJS, irregularly The subject provided informed consent prior to treatment. The
pigmented macules of varying sizes and colors can be found on the treatment area was identified as the labial lentigines. The face was
vermillion border of the lip, palate, perioral skin, buccal mucosa, and cleaned, disinfected with 75% alcohol, and dried. A Q-switched alexan-
tongue. Common treatments, such as cryosurgery, surgical excision, drite laser with a 755-nm wavelength and a 50-ns pulse duration setting
electrodesiccation, dermabrasion, and carbon dioxide or argon laser was used. All treatments were performed using a 2-mm spot size and
ablation, commonly result in incomplete removal of the macules, scar- fluence between 4 and 9 J/cm.2 No topical anesthesia was needed before
ring, or changes in normal pigmentation. Although there are a few re- the operation, although topical antibiotic ointment was applied to the
ports regarding successful treatment of intestinal polyps in patients with treated site to prevent possible infection after the operation. The patient
PJS,6,7 there is little research regarding treatment of the associated was advised to be vigilant about sun protection, and a follow-up was
mucocutaneous melanosis.8–10 This study reports a case of labial lenti- performed to evaluate the treatment efficacy and complications.
gines in a patient with PJS that was treated using a Q-switched alexan- The labial lentigines immediately disappeared or faded after the first
drite laser. laser irradiation. Their surfaces became whitish, and grayish patches
were seen at the treated site, accompanied by edema. Crust formed
2. Case presentation within 1–2 days and sloughed off after one week. The treatment interval
was 3 months, and the first treatment effect was evaluated prior to the
A 19-year-old male with PJS presented to the plastic surgery outpa- second treatment (Fig. 2). The second treatment used the same protocol
tient clinic at the Affiliated Hospital of Xuzhou Medical University with as the first, and long-term follow-up was conducted after the final
labial lentigines. The disease had been detected in the patient around the treatment. Fig. 3 shows a photograph taken 1 year after the final treat-
age of 5 years. Several black-pigmented macules begin to appear on the ment. The labial lentigines were removed successfully without recur-
lips, and the size of the pigmented macules gradually increased, until rence, scar formation, or other complications.

* Corresponding author. Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Huai-hai West Road, Xuzhou 221002, Jiangsu, China.
E-mail address: 100000401006@xzhmu.edu.cn (P. Jin).

https://doi.org/10.1016/j.cjprs.2021.09.006
Received 1 September 2021; Accepted 8 September 2021
Available online 20 October 2021
2096-6911/© 2021 China Medical Cosmetology Press Co. Ltd. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co. Ltd. This is an open access
article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
S. Yang et al. Chinese Journal of Plastic and Reconstructive Surgery 3 (2021) 147–149

Fig. 1. Before treatment, black-pigmented macules extended over the entire lip.

successfully without recurrence, scar formation, or other complications.


In clinical practice, there are also reports of successful cases of applying
Q-switched Nd-YAG laser treatment and Q-switched ruby laser treat-
ment, which provide additional options for the treatment of mucocuta-
neous melanosis in PJS.

4. Conclusion

Our study supports the use of a Q-switched alexandrite laser for safe
and effective treatment of labial lentigines in PJS.

Fig. 2. On follow-up after the first treatment, the patient’s labial lentigines were
Ethics Declarations
reduced in size and lightened in color.
Ethics approval and consent to participate

The need for ethical approval was waived as this is a case report.

Consent for publication

The patient gave written informed consent to publish the data con-
tained within this study.

Competing interest

The authors declare that they have no competing interests.

Authors’ contributions

Yang S: Data curation, Writing-Original draft. Tao C: Investigation. Xu


H: Resources. Zhang Y: Investigation. Jin P: Supervision.

References
Fig. 3. At a follow-up assessment one year after the last treatment, the labial
1. Tacheci I, Kopacova M, Bures J. Peutz-Jeghers syndrome. Curr Opin Gastroenterol.
lentigines were removed successfully without recurrence, scar formation, or
2021;37(3):245–254. https://doi.org/10.1097/MOG.0000000000000718.
other complications. The patient satisfaction was excellent. 2. Yamamoto M, Iwamoto K, Suzuki R, et al. Laparoscopic-assisted disinvagination and
polypectomy for multiple intussusceptions induced by small intestinal polyps in
patients with Peutz-Jeghers syndrome: a case report. World J Surg Oncol. 2021;19(1):
3. Discussion 22. https://doi.org/10.1186/s12957-021-02133-5.
3. He X, Mao Y, Ben Y, et al. Clinical characteristics and basic research development of
Peutz-Jeghers syndrome. Chin Med Sci J. 2001;16(1):49–51.
Pigmentation of mucosa and skin, along with multiple gastrointes- 4. Huang ZH, Song Z, Zhang P, et al. Clinical features, endoscopic polypectomy and
tinal polyps, is characteristic of PJS. Mucocutaneous melanosis in PJS STK11 gene mutation in a nine-month-old Peutz-Jeghers syndrome Chinese infant.
World J Gastroenterol. 2016;22(11):3261–3267. https://doi.org/10.3748/
seriously affects the physical and mental health of patients,1–5 but its
wjg.v22.i11.3261.
treatment still perplexes the majority of clinicians. Previous treatments, 5. Torroni F, Romeo E, Rea F, et al. Conservative approach in Peutz-Jeghers syndrome:
such as surgical excision, electrodesiccation, cryosurgery, dermabrasion, single-balloon enteroscopy and small bowel polypectomy. World J Gastrointest
Endosc. 2014;6(7):318–323. https://doi.org/10.4253/wjge.v6.i7.318.
and carbon dioxide or argon laser ablation, commonly result in incom-
6. Kirakosyan E, Lokhmatov M. High-tech diagnostic methods and enteroscopic
plete removal of the polyps, scarring, or changes in normal pigmenta- treatment of children with Peutz-Jeghers syndrome. Eur J Pediatr Surg. 2020;30(6):
tion.6,7,11,12 In this case, we treated labial lentignes with a Q-switched 529–535. https://doi.org/10.1055/s-0039-3400286.
alexandrite laser and achieved a satisfactory therapeutic outcome. 7. Suwa T, Imai K, Hotta K, et al. Underwater ischemic polypectomy for multiple small
bowel polyps in a patient with Peutz-Jeghers syndrome. Am J Gastroenterol. 2021;
Long-term follow-up suggests that the labial lentigines were removed 116(3):452. https://doi.org/10.14309/ajg.0000000000001162.

148
S. Yang Chinese Journal of Plastic and Reconstructive Surgery 3 (2021) 147–149

8. Xi Z, Hui Q, Zhong L. Q-switched alexandrite laser treatment of oral labial lentigines 11. Kopacova M, Tacheci I, Rejchrt S, et al. Peutz-Jeghers syndrome: diagnostic and
in Chinese subjects with Peutz-Jeghers syndrome. Dermatol Surg. 2009;35(7): therapeutic approach. World J Gastroenterol. 2009;15(43):5397–5408. https://
1084–1088. https://doi.org/10.1111/j.1524-4725.2009.01192.x. doi.org/10.3748/wjg.15.5397.
9. Nair V. Laugier-Hunziker syndrome: complete clearance of mucosal lentigines with a 12. Kato S, Takeyama J, Tanita Y, et al. Ruby laser therapy for labial lentigines in Peutz-
single session of Q-Switched Nd:YAG laser. J Cosmet Laser Ther. 2019;21(6):343–345. Jeghers syndrome. Eur J Pediatr. 1998;157(8):622–624. https://doi.org/10.1007/
https://doi.org/10.1080/14764172.2019.1660791. s004310050898.
10. DePadova-Elder SM, Milgraum SS. Q-switched ruby laser treatment of labial
lentigines in Peutz-Jeghers syndrome. J Dermatol Surg Oncol. 1994;20(12):830–832.
https://doi.org/10.1111/j.1524-4725.1994.tb03714.x.

Editorial Announcement—Electronic Manuscript Submission

We are delighted to announce that on August 3, 2021, the online submission and peer review tracking system, Editorial Manager, was successfully
implemented for the Chinese Journal of Plastic and Reconstructive Surgery (CJPRS). From September 1, 2021, we have made it compulsory for authors to
submit all their manuscripts through this system only.
To submit a manuscript, please proceed to the Editorial Manager website at https://www.editorialmanager.com/cjprs. New instructions for authors
can be accessed at the journal homepage: http://www.keaipublishing.com/en/journals/chinese-journal-of-plastic-and-reconstructive-surgery/.
We will spare no effort to make the move as seamless as possible, and any queries can be directed to the Editorial Office via email to cjprs_2020@ho
tmail.com.

149
Gastroenterology 2022;162:2063–2085

CLINICAL PRACTICE GUIDELINES


Diagnosis and Management of Cancer Risk in the
Gastrointestinal Hamartomatous Polyposis Syndromes:
Recommendations From the US Multi-Society Task Force
on Colorectal Cancer
C. Richard Boland,1,* Gregory E. Idos,2,* Carol Durno,3,4 Francis M. Giardiello,5
Joseph C. Anderson,6,7,8 Carol A. Burke,9 Jason A. Dominitz,10,11 Seth Gross,12
Samir Gupta,13,14,15 Brian C. Jacobson,16 Swati G. Patel,17 Aasma Shaukat,18,19
Sapna Syngal,20,21,22 and Douglas J. Robertson6,7
1
Division of Gastroenterology, University of California-San Diego School of Medicine, San Diego, California; 2Divisions of
Gastroenterology and Clinical Cancer Genomics, Center for Precision Medicine, City of Hope National Medical Center, Duarte,
California; 3The Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; 4Division of
Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada;
5
Division of Gastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland;
6
Veterans Affairs Medical Center, White River Junction, Vermont; 7Geisel School of Medicine at Dartmouth, Hanover, New
Hampshire; 8University of Connecticut, Farmington, Connecticut; 9Department of Gastroenterology, Hepatology and Nutrition,
Cleveland Clinic, Cleveland, Ohio; 10Veterans Affairs Puget Sound Health Care System, Seattle, Washington; 11University of
Washington School of Medicine, Seattle, Washington; 12Division of Gastroenterology and Hepatology, New York University
Langone Health, New York, New York; 13Veterans Affairs Medical Center, San Diego, California; 14University of California San
Diego, La Jolla, California; 15Veterans Affairs San Diego Healthcare System, San Diego, California; 16Division of
Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; 17University of Colorado School of Medicine,
Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado; 18Minneapolis Veterans Affairs Health Care
System, Minneapolis, Minnesota; 19University of Minnesota, Minneapolis, Minnesota; 20Brigham and Women’s Hospital, Boston,
Massachusetts; 21Dana-Farber Cancer Institute, Boston, Massachusetts; and 22Harvard Medical School, Boston, Massachusetts

The gastrointestinal hamartomatous polyposis syndromes are gastrointestinal and colonic polyposis, colon cancer, and other
rare, autosomal dominant disorders associated with an extraintestinal manifestations and cancers. Due to the relative
increased risk of benign and malignant intestinal and extra- rarity of the hamartomatous polyposis syndromes, recom-
intestinal tumors. They include Peutz-Jeghers syndrome, ju- mendations for management are based on few studies. This U.S
venile polyposis syndrome, the PTEN hamartoma tumor Multi-Society Task Force on Colorectal Cancer consensus
syndrome (including Cowden’s syndrome and Bannayan-Riley- statement summarizes the clinical features, assesses the
Ruvalcaba syndrome), and hereditary mixed polyposis syn-
drome. Diagnoses are based on clinical criteria and, in some
cases, confirmed by demonstrating the presence of a germline *Authors share co-first authorship.
pathogenic variant. The best understood hamartomatous pol-
yposis syndrome is Peutz-Jeghers syndrome, caused by germ- Abbreviations used in this paper: AVM, arteriovenous malformation; BMP,
bone morphogenetic protein; BRRS, Bannayan-Riley-Ruvalcaba syn-
line pathogenic variants in the STK11 gene. The management is drome; CI, confidence interval; CL, confidence limits; CR, cumulative risk;
focused on prevention of bleeding and mechanical obstruction CRC, colorectal cancer; CS, Cowden’s syndrome; EGD, esophagogas-
troduodenoscopy; GRADE, Grading of Recommendations Assessment,
of the small bowel by polyps and surveillance of organs at Development and Evaluation; HHT, hereditary hemorrhagic telangiectasia;
CLINICAL PRACTICE GUIDELINES

increased risk for cancer. Juvenile polyposis syndrome is HMPS, hereditary mixed polyposis syndrome; JPS, juvenile polyposis
caused by a germline pathogenic variant in either the SMAD4 or syndrome; MRE, magnetic resonance enterography; MRI, magnetic
resonance imaging; PHTS, PTEN hamartoma tumor syndrome; PJS,
BMPR1A genes, with differing clinical courses. Patients with Peutz-Jeghers syndrome; RR, relative risk; SIR, standardized incidence
SMAD4 pathogenic variants may have massive gastric polypo- ratio; USMSTF, US Multi-Society Task Force on Colorectal Cancer; VCE,
sis, which can result in gastrointestinal bleeding and/or video capsule endoscopy.
protein-losing gastropathy. Patients with SMAD4 mutations This article is being published jointly in Gastroenterology, Gastrointestinal
usually have the simultaneous occurrence of hereditary hem- Endoscopy, and The American Journal of Gastroenterology. The article is
orrhagic telangiectasia (juvenile polyposis syndrome– identical except for minor stylistic and spelling differences in keeping with
each journal’s style. Citations from any of the 3 journals can be used when
hereditary hemorrhagic telangiectasia overlap syndrome) citing this article.
that can result in epistaxis, gastrointestinal bleeding from
Most current article
mucocutaneous telangiectasias, and arteriovenous malforma-
tions. Germline pathogenic variants in the PTEN gene cause © 2022 by the American Gastroenterological Association,
overlapping clinical phenotypes (known as the PTEN hamar- the American Society for Gastrointestinal Endoscopy, and
the American College of Gastroenterology
toma tumor syndromes), including Cowden’s syndrome and 0016-5085/$36.00
related disorders that are associated with an increased risk of https://doi.org/10.1053/j.gastro.2022.02.021
2064 Boland et al Gastroenterology Vol. 162, No. 7

current literature, and provides guidance for diagnosis, Methods


assessment, and management of patients with the hamar- A computer-aided PubMed search was performed from 2000
tomatous polyposis syndromes, with a focus on endoscopic to 2018, with additional back searches as required, and consisted
management. of the following search terms: hamartoma, hamartomatous polyp,
hamartoma syndrome, Peutz-Jeghers syndrome, juvenile polyp,
juvenile polyposis, Cowden’s syndrome, Cowden’s disease, PTEN-

T he gastrointestinal hamartomatous polyposis syn-


dromes are rare, autosomal dominant disorders
associated with an increased risk of benign and malignant
hamartoma, Bannayan-Riley-Ruvalcaba syndrome, hyperplastic
polyposis, serrated polyposis, and hereditary mixed polyposis
syndrome. Only English-language articles were reviewed. Pub-
intestinal and extraintestinal tumors. Nevertheless, there lished articles were selected on the basis of relevance to the
has been tremendous progress in recent years, both in un- diagnosis or clinical management of these diseases. Emphasis
derstanding the underlying genetics that underpin these was placed on the risk for gastrointestinal cancer in these dis-
disorders and in elucidating the biology of associated pre- orders to gain consensus on rational and reasonable strategies
malignant and malignant conditions. Emerging data in for management once these diseases are diagnosed in a family.
affected populations focus increasingly on those with The document was approved by the governing boards of each of
defined cancer susceptibility germline pathogenic variants the sponsoring gastroenterology societies.
leading to less heterogeneity in terms of quantifying cancer The USMSTF approach to an adapted use of Grading of
risk. Recommendations Assessment, Development and Evaluation
The US Multi-Society Task Force on Colorectal Cancer (GRADE) has been described previously.1 In brief, the GRADE
(USMSTF) is a group of colorectal cancer (CRC) content process categorizes the quality of the evidence as high, mod-
experts appointed by the American College of Gastroenter- erate, low, or very low on the basis of the strength of under-
lying studies, and that categorization can be adjusted on the
ology, American Gastroenterological Association, and
basis of study limitations. For example, randomized trials begin
American Society for Gastrointestinal Endoscopy, supple-
as high-quality evidence and observational studies as low-
mented at times by other experts to complement existing
quality evidence, but their quality may be adjusted up or
expertise. In this USMSTF Consensus Statement, the
down on the basis of specific study factors. Although the
gastrointestinal hamartomatous polyposis syndromes were GRADE process entails a formal meta-analysis to assess the
chosen because of recent progress in understanding these quality of evidence for each recommendation, the USMSTF
diseases. The following entities reviewed are: Peutz-Jeghers employs a modified, qualitative approach for this assessment.
syndrome (PJS), juvenile polyposis syndrome (JPS), PTEN The GRADE process separates evaluation of the quality of the
hamartoma tumor syndrome (PHTS, including Cowden’s evidence to support a recommendation from the strength of
syndrome [CS] and Bannayan-Riley-Ruvalcaba syndrome that recommendation. This is done in recognition of the fact
[BRRS]), and hereditary mixed polyposis syndrome (HMPS). that, although the quality of the evidence can influence the
Germline alterations are known to cause each of these dis- strength of the recommendation, other factors can influence a
orders, but the diagnosis can also be made on the basis of recommendation, such as adverse effects, patient preferences,
clinical criteria. values, and cost. Generally, strong recommendations mean that
Although there are essentially no long-term prospective most informed patients would choose the recommended man-
controlled studies of comparative effectiveness of manage- agement. Weak recommendations mean that patients’ choices
ment strategies for these syndromes, there have been will vary according to their values and preferences, and clini-
consensus statements by expert panels that made manage- cians should ensure that patient care is in keeping with their
ment recommendations for these disorders. The goal of this values and preferences. When the quality of the evidence to
USMSTF Statement was to review the literature focusing on support a recommendation is low or very low, or if there is a
the most recent data, synthesize both the data and the sug- close balance between desirable and undesirable consequences,
gested approaches to diagnosis and management by other then usually only a weak recommendation would be warranted.
Weaker recommendations are indicated by phrases such as “we
expert groups, and present the consensus recommendations
suggest,” and stronger recommendations are typically stated as
of the USMSTF (Table 1). Review of summary tables, con-
“we recommend.”
ference calls, and revisions of iterative drafts, including
However, the relative infrequency of, and absence of
CLINICAL PRACTICE GUIDELINES

recommendation statements, were used to reach consensus,


controlled prospective trials of the interventions (eg, to prevent
at which time documents were forwarded to Governing cancer) in, these syndromes leave all of the recommendations
Boards for approval. As our USMSTF is a group of individuals without a robust basis of underlying evidence. Thus, all of the
with expertise in gastroenterology and gastrointestinal ma- interventional recommendations fall (at best) into the “low
lignancies, we have reserved our management recommen- quality of evidence” GRADE category, indicating that the true
dations to these areas and defer to other expert groups’ effect of the interventions may be markedly different than
recommendations for other cancers (which are reviewed estimated at this time, and that further research is likely to
here). This document therefore recommends clinical ap- impact or change our confidence in the effects. As such, this
proaches to diagnosing and managing these conditions that review is intended to establish a starting point for future
affect children and adults, focuses on cancer risk, and pro- research into the care of patients with the hamartomatous
vides insights into future research opportunities. polyposis syndromes.
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2065

Table 1.Questions and Recommendations of Best Practice

Which individuals with hamartomatous polyps should be referred for genetic evaluation?
We recommend patients with any of the following undergo a genetic evaluation: 2 or more lifetime hamartomatous polyps, a family history of
hamartomatous polyps, or a cancer associated with a hamartomatous polyposis syndrome in first or second-degree relatives. Genetic
testing (if indicated) should be performed using a multigene panel test. (Strong recommendation, low quality of evidence)
Peutz-Jeghers syndrome
Who should undergo a genetic evaluation for Peutz-Jeghers syndrome?
We recommend genetic evaluation for any individual with the following: 1) 2 or more histologically confirmed Peutz-Jeghers polyps, 2) any
number of Peutz-Jeghers polyps in an individual who has a family history of Peutz-Jeghers syndrome in a first-degree relative, 3)
characteristic mucocutaneous pigmentation in a person with a family history of Peutz-Jeghers syndrome, 4) any number of Peutz-
Jeghers polyps in a person with the characteristic mucocutaneous pigmentation of Peutz-Jeghers syndrome. (Strong
recommendation, low quality of evidence)
Which organs should undergo surveillance when caring for a patient with Peutz-Jeghers syndrome?
Patients with Peutz-Jeghers syndrome are at increased risk for cancer in multiple organs including cancer of the breast, small bowel,
colon, stomach, pancreas, ovaries, testes, and lungs.
Given this risk, we recommend a multidisciplinary approach to cancer surveillance in these organs (Strong recommendation, low
quality of evidence)
How and when should small bowel surveillance be performed in Peutz-Jeghers syndrome?
We recommend that baseline small bowel surveillance using video capsule endoscopy or magnetic resonance enterography be performed
between ages 8-10 years or earlier if the patient is symptomatic. If no polyps are found at the initial examination, surveillance should
resume at age 18. Because of the risk of small bowel intussusception, small bowel surveillance in adulthood is recommended to
continue throughout life every 2-3 years. (Strong recommendation, low quality of evidence)
What is the recommended approach to endoscopic surveillance of the colon, stomach, and duodenum in Peutz-Jeghers syndrome?
We suggest a baseline upper gastrointestinal endoscopy between the ages of 8 and 10 years, which could be performed at the time of
capsule placement for small bowel surveillance or if polyps are identified on magnetic resonance enterography. Although the initiation
age for colonoscopy remains uncertain, we also suggest initiation of colonoscopy at same time as esophagogastroduodenoscopy. In
those in whom characteristic polyps are detected, both colonoscopy and esophagogastroduodenoscopy should be repeated every 2–
3 years. In those in whom there are no Peutz-Jeghers polyps at baseline, surveillance is repeated at age 18 years, or sooner should
symptoms arise, and then every 3 years. (Weak recommendation, very low quality of evidence)
What size polyps found on small bowel imaging in Peutz-Jeghers syndrome should be removed?
We recommend polypectomy of small bowel polyps that are symptomatic or 10 mm to prevent intussusception and other complications,
such as bleeding.(Strong recommendation, low quality of evidence)
What is the recommended pancreatic cancer surveillance in Peutz-Jeghers syndrome?
We suggest annual pancreatic cancer surveillance with either magnetic resonance cholangiopancreatography or endoscopic ultrasound
starting at age 35 years. (Weak recommendation, low quality of evidence)
Juvenile Polyposis Syndrome
Who should undergo a genetic evaluation for juvenile polyposis syndrome?
We recommend genetic evaluation for any individual with 1) 5 or more juvenile polyps of the colon or rectum; or 2) 2 or more juvenile
polyps in other parts of the gastrointestinal tract; or (3) any number of juvenile polyps and 1 or more first-degree relatives with juvenile
polyposis syndrome. (Strong recommendation, low quality of evidence)
Which organs should undergo surveillance when caring for a patient with juvenile polyposis syndrome?
Juvenile polyposis syndrome patients are at increased risk for cancer in multiple organs including cancer of the colon and stomach.
Given this risk, we recommend patients with juvenile polyposis syndrome undergo surveillance of the colon and stomach. (Strong
recommendation, low quality of evidence)
At what age should colonoscopic and upper endoscopic surveillance begin in individuals identified with juvenile polyposis syndrome?
We suggest initiating colonoscopic and upper endoscopic surveillance at age 12–15 years, or earlier if symptomatic. Surveillance should
be repeated every 1–3 years depending on polyp burden. (Weak recommendation, low quality of evidence)
Which patients with juvenile polyposis syndrome should undergo screening for hereditary hemorrhagic telangiectasia?
We suggest patients with SMAD4 pathogenic variants be clinically evaluated for HHT at the time of the diagnosis, including screening for
and appropriate management of cerebral and pulmonary AVMs. (Weak recommendation, low quality of evidence)
CLINICAL PRACTICE GUIDELINES

PTEN hamartoma tumor syndrome


Which gastrointestinal findings should prompt a genetic evaluation for PTEN hamartoma tumor syndrome?
We recommend individuals with multiple gastrointestinal hamartomas or ganglioneuromas undergo genetic evaluation for Cowden’s
syndrome and related conditions. (Strong recommendation, low quality of evidence)
Which organs should undergo surveillance for cancer when caring for a patient with PTEN hamartoma tumor syndrome?
In PTEN hamartoma tumor syndrome, patients are at increased risk for cancer in multiple organs, including cancer of the breast, thyroid,
kidney, uterus, colon, and skin.
Given this risk, we recommend a multi-disciplinary approach to cancer surveillance in these organs. (Strong recommendation, low
quality of evidence)
What is the recommended colonoscopic surveillance in individuals identified with PTEN hamartoma tumor syndrome?
We suggest colonoscopy surveillance to begin at age 35 years (or 10 years younger than age of any relative with colorectal cancer),
repeated at intervals no greater than 5 years, depending on polyp burden. (Weak recommendation, low quality of evidence)

NOTE. Specific circumstances may merit modification of the recommendations. In cases where very-early-onset cancers
develop, the above statements may be modified to start surveillance 10 years earlier than the youngest cancer diagnosis in the
family.
2066 Boland et al Gastroenterology Vol. 162, No. 7

Cancer Family History Assessment and the American College of Medical Genetics and Genomics and
National Society of Genetic Counselors are available for
Referral for Genetic Testing in details regarding the elements and process of genetic
Gastroenterology Practice counseling.3 Although traditional models of genetic evalua-
tion and testing included a certified genetic counselor,
alternative models exist and are emerging that include
Question: Which individuals with hamartomatous polyps provisions for pretest counseling to be provided by physi-
should be referred for genetic evaluation? cians and other health care providers in order to deal with
Recommendation: We recommend patients with any of
the increasing demand for genetic testing. If a patient is
the following undergo a genetic evaluation: 2 or more
lifetime hamartomatous polyps, a family history of found to be a carrier of a germline pathogenic variant, or the
hamartomatous polyps, or a cancer associated with a results are ambiguous due to the finding of a variant of
hamartomatous polyposis syndrome in first- or second- uncertain significance, the help of a genetics provider for
degree relatives. Genetic testing (if indicated) should be post-test counseling and education is recommended. In the
performed using a multigene panel test. (Strong current era, the vast majority of genetic testing for inherited
recommendation, low quality of evidence) cancer risk predisposition is performed using a multigene
panel testing approach.4 Some patients or families may elect
Hereditary cancer syndromes account for approximately to decline genetic testing due to concerns about risk to
5%–10% of new cancer diagnoses and many of the cancers confidentiality and insurance; in these cases, surveillance
that arise in families with undiagnosed hereditary cancer may still be indicated in the presence of a concerning clin-
syndromes are preventable. The identification of individuals ical and/or family history.
with a hereditary gastrointestinal cancer syndrome requires If a germline pathogenic variant is identified, other
a thorough evaluation of the patient’s personal and family family members should be offered testing for clarification of
history of cancer. The collection and assessment of family their own risk. This testing may facilitate initiation of
cancer history is a valuable tool for cancer interception and screening for associated cancers before symptomatic man-
prevention and can be critical in the identification of genetic ifestations occur and reduce the morbidity and mortality
susceptibility. An accurate family history is one that collects associated with the syndrome. For example, early small
the following information: 1) type of cancer, 2) age at bowel surveillance may find a polyp that could be removed
diagnosis of each primary cancer, 3) lineage (maternal or from a child with a pathogenic variant in STK11 before
paternal), 4) ethnicity (people of some ethnicities, such as leading to intussusception. It is important to recognize that
those with Ashkenazi Jewish ancestry, are at greater risk for genetic testing may not identify pathogenic variants in every
certain cancers), and 5) results of any previous cancer- family suspected of a hereditary syndrome. However, there
related genetic testing.2 may be clinical features in the family history that suggest a
Features of a patient’s personal history and clinical familial predisposition to cancer and suggest more intensive
characteristics may suggest an inherited susceptibility to surveillance recommendations. Referral to Centers of
cancer. Although it is not rare to identify individuals with an Excellence might be particularly helpful when genetic
isolated hamartomatous polyp (particularly an isolated ju- testing results are ambiguous in the setting of suspicious
venile polyp), other features may prompt further evaluation features and prophylactic surgery is being considered.
for an underlying hereditary syndrome. Features associated Lastly, in the era of multigene panel testing, there may be a
with the hamartomatous polyposis syndromes are outlined scenario in which a germline variant is found incidentally
in detail in this document and include early age at cancer associated with an unsuspected syndrome. In these cases,
onset, multiple cancers in close relatives, unusual numbers patients may be eligible for cancer screening and surveil-
of hamartomatous polyps, or associated dermatologic find- lance as outlined. However, phenotype and cancer risk
ings. Genetic evaluation may include genetic counseling compared with patients with classic familial features are not
and/or genetic testing. established and are areas of active research. Enlisting the
Genetic counseling is a key component to hereditary assistance of a genetic specialist may be particularly helpful
cancer risk assessment. The purpose of genetic counseling is in interpreting ambiguous results and providing manage-
CLINICAL PRACTICE GUIDELINES

to educate individuals about the genetic and biologic factors ment recommendations in these cases.
that are related to a patient’s cancer diagnosis or risk of When children are identified with a hamartomatous
disease. Counseling helps an individual understand the polyposis syndrome, their transition of care to adulthood for
relevant genetic, medical, and psychosocial information to cancer surveillance is a unique aspect that bears consider-
make informed decisions about their health care. This in- ation. It is imperative to transition adolescents with life-long
cludes reviewing and expanding the following: family his- medical conditions from child-centered to adult-centered
tory information, elements of genetic testing, tailored cancer care. Preparation for this transition takes place throughout
risks associated with a pathogenic variant, impact on med- childhood and adolescence to achieve independent health
ical management, reproductive issues and options, confi- management in adulthood. Steps required are individualized
dentiality of results, risks with genetic discrimination, based on the developmental needs of the patient. Inherited
potential significance of test results for other family mem- conditions involve generational factors, as multiple family
bers, and other pertinent topics. Practice guidelines from members may be affected. Health care providers can assist
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2067

in the transition of care by coordinating screening and anemia is the most common presentation, followed by
surveillance to ensure patients receive recommended care.5 abdominal pain, diarrhea, and intussusception. The clinical
management in early life is initially focused on preventing
complications of small bowel polyposis–related obstruction
Gastrointestinal Hamartomatous and bleeding and, in adulthood, the focus is primarily on
Polyposis Syndromes management of cancer risk.
The hamartomatous polyposis syndromes are rare en-
tities with an estimated prevalence of 1/100,000–
200,000,6,7 but this has not been measured directly in any
Diagnosis
The diagnostic clinical features of PJS include the pres-
population. The term hamartoma implies a non-neoplastic
ence of 2 or more histologically confirmed PJ polyps; any
tumor with a markedly distorted architecture composed of
number of PJ polyps in an individual who has a family his-
an abnormal mixture of cells and tissue normally present in
tory of PJS in a first-degree relative; characteristic muco-
that particular area. The diagnosis is based on the presence
cutaneous pigmentation in a person with a family history of
of a pathogenic germline variant or meeting clinical criteria
PJS; or any number of PJ polyps in a person with the
for the syndrome. The hamartomatous polyposis syndromes
characteristic mucocutaneous pigmentation of PJS.11
are distinct from Lynch syndrome and the adenomatous
polyposis syndromes, based on the presence of hamartomas
(Figures 1 and 2). Certain hamartomatous polyps of the gut Genetics
have a unique histopathological appearance, such as those In 1997, a genetic locus for PJS was mapped to chro-
associated with PJS, PHTS, JPS, and HMPS.8 Hamartomas are mosome 19p13.3,12 which in 1998 led to the cloning of the
not typically characterized by dysplasia, but some evidence STK11 (serine/threonine kinase) gene, which encodes the
suggests the existence of a hamartoma–carcinoma pathway LKB1 (liver kinase B1) protein, and linkage to PJS.13 STK11
in some of these polyps. functions like a tumor suppressor gene, regulates cell
growth via adenosine monophosphate–activated protein
kinase,14 and negatively regulates mTOR signaling.15
Peutz-Jeghers Syndrome PJS is inherited in an autosomal dominant fashion with
an inactivating germline pathogenic variant inherited from
the affected parent. It was initially assumed that the polyps
Question: Who should undergo a genetic evaluation for
Peutz-Jeghers syndrome? occurred after the loss of the second, wild-type, allele
Recommendation: We recommend genetic evaluation inherited from the unaffected parent in a somatic tissue
for any individual with the following: 1) 2 or more according to the classic “two-hit” model of Knudson.16
histologically confirmed Peutz-Jeghers polyps, 2) any However, loss of the wild-type allele (ie, the second hit) is
number of Peutz-Jeghers polyps in an individual who not an obligatory feature of PJ polyps.17 Moreover, recent
has a family history of Peutz-Jeghers syndrome in a data in mice indicate that the presence of a single inacti-
first-degree relative, 3) characteristic mucocutaneous
vating germline pathogenic variant (ie, haploinsufficiency),
pigmentation in a person with a family history of Peutz-
Jeghers syndrome, and 4) any number of Peutz-Jeghers as occurs in individuals with PJS, promotes the development
polyps in a person with the characteristic of gastrointestinal polyposis, and that loss of the wild-type
mucocutaneous pigmentation of Peutz-Jeghers allele is not necessary for the formation of the polyps in
syndrome. (Strong recommendation, low quality of these mice. Furthermore, conditional knockout of the STK11
evidence) gene targeted to gastrointestinal smooth muscle cells yields
the same polyposis phenotype in mice.18 Evidence suggests
that LKB1 deficiency in either T cells19 or mesenchymal
cells18,19 leads to immune cell proliferation in the stroma.
Clinical Features Stromal deficiency of LKB1 leads to tumor formation (in
PJS was the first hamartomatous polyposis syndrome mice) via the interleukin-11–JAK/STAT3 (Janus kinase/
described, by Peutz in Holland in 1921 and by Jeghers, signal transducer and activator of transcription 3) pathway,
CLINICAL PRACTICE GUIDELINES

McKusick, and Katz in the United States in 1949.9 The and administration of the JAK1/2 inhibitor ruxolitinib
clinical recognition of PJS was facilitated by the character- dramatically reduces polyposis in mice.20 The phenomenon
istic mucocutaneous freckling around the mouth and mul- of stromal-driven epithelial polyp development may be
tiple cerebriform-appearing polyps due to smooth muscle referred to as a “landscaper” genetic effect.21 These dis-
bands coursing through the polyp (Figures 1A and 2A–C, coveries raise the possibility of the development of novel
and Table 2). Hamartomatous polyps vary in size and may preventive pharmacological interventions for patients.
have a characteristic histologic structure, which makes it Genotype–phenotype relationships reveal that missense
possible to distinguish the PJ polyp. PJ polyps are typically alterations in STK11 are associated with later onset of
composed of branching bands of smooth muscle covered by symptoms than other sequence variations.11,22 Some of the
hyperplastic glandular mucosa.10 PJS polyps may develop in germline pathogenic variants in STK11 are Alu-mediated
the stomach, small intestine, and colon. Rectal bleeding with deletions and inversions.23 Deletions and inversions are
2068 Boland et al Gastroenterology Vol. 162, No. 7
CLINICAL PRACTICE GUIDELINES

Figure 1. Histologic characteristics of polyps. hamartomatous polyps. Images courtesy of Drs Aaron Pollett and Thomas
Plesec. (A) Peutz-Jeghers polyp: pathognomonic broad bands of mucosal smooth muscle seen throughout the lesion. (B)
Juvenile polyp: characteristic chronic inflammatory infiltrate and cystic dilatation. (C) Adenomatous polyp: characteristic
hypercellularity with glandular crowding, enlarged nuclei, increased mitotic activity and reduced goblet cells. By definition, all
tubular adenomas show epithelial dysplasia. (D) Ganglioneuroma: benign neuroectodermal tumor composed of ganglion and
Schwann cells.

likely to completely inactivate a gene, whereas a missense genetic diagnostic platforms must include strategies to
pathogenic variant may have an intermediate effect on detect these types of germline variations; at least some of
protein function. Approximately 15% of STK11 pathogenic the “missing” germline pathogenic variants may involve
variants in PJS involve large genetic deletions.24 Therefore, obscure rearrangements of the STK11 gene.
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2069

CLINICAL PRACTICE GUIDELINES

Figure 2. Endoscopic images of hamartomatous polyposis syndromes. Endoscopic photos provided courtesy of Swati Patel,
MD, Gregory Idos, MD, and Carol Burke, MD. (A) Peutz-Jeghers small bowel polyps. (B) Peutz-Jeghers gastric polyps. (C)
Peutz-Jeghers colon polyps. (D) Juvenile polyposis gastric polyps. (E) Juvenile polyposis colon polyp. (F) Cowden syndrome
associated–esophageal glycogenic acanthosis. (G) Cowden syndrome small bowel polyps. (H) Cowden syndrome colon
polyps. (I) Cowden syndrome gastric polyps.
2070 Boland et al Gastroenterology Vol. 162, No. 7

Table 2.Clinical Features of Gastrointestinal Hamartomatous Polyposis Syndromes

“Commercially available
Syndrome gene testing” Polyps Clinical features

PJS STK11 Peutz-Jeghers polyps Childhood: Labial pigmentation; gastrointestinal


(pathologically bleeding and intussusception
characteristic) Adults: Increased risk for multiple cancers
JPS SMAD4 or BMPR1A Juvenile (inflammatory) Childhood: Gastrointestinal bleeding, auto-
polyps; juvenile polyps and amputation of polyps; anemia
inflammatory polyps are Adults: CRC and gastric cancer
pathologically indistinct
CS PTEN (inactivation) Hyperplastic polyps; juvenile- Childhood: none
WWP1 (gain-of-function) like polyps; Adults: multiple cancer risks
ganglioneuromas; lipomas;
hamartomas;adenomas
BRRS PTEN Same as for CS Developmental delay, hemangiomas, lipomas,
gastrointestinal polyps
HMPS GREM1 (duplication Pathologically mixed with Increased risk of colonic polyposis and CRC
upstream of promoter) features of adenoma,
hyperplastic polyps,
inflammatory polyps

Cancer Risk age-adjusted cancer risks were noted for the small intestine
(RR, 520; 95% CL, 220 and 1306; cumulative risk [CR],
13%), stomach (RR, 213; 95% CL, 96 and 368; CR, 29%),
pancreas (RR, 132; 95% CL, 44 and 261; CR, 36%), colon
Question: Which organs should undergo surveillance
when caring for a patient with Peutz-Jeghers syndrome? (RR, 84; 95% CL, 47 and 137; CR, 39%), esophagus (RR, 57;
Recommendation: Patients with Peutz-Jeghers 95% CL, 2.5 and 557; CR, 0.5%), ovary (RR, 27; 95% CL, 7.3
syndrome are at increased risk for cancer in multiple and 68; CR, 21%), lung (RR, 17.0; 95% CL, 5.4 and 39; CR,
organs, including cancer of the breast, small bowel, 15%), uterus (RR, 16.0; 95% CL, 1.9 and 56; CR, 9%), testes
colon, stomach, pancreas, ovaries, testes, and lungs. (RR, 4.5; 95% CL, 0.12 and 25; CR, 9%), and breast (RR,
Given this risk, we recommend a multidisciplinary 15.2; 95% CL, 7.6 and 27; CR, 54%).26 Mean age at the time
approach to cancer surveillance in these organs. (Strong
of cancer diagnosis was 42.9 years. The absolute risk of
recommendation, low quality of evidence)
developing any cancer between the ages of 15 and 64 was
estimated to be 93%.
Very high lifetime risks of cancer occur in multiple or- The risk of cancer in PJS was revisited in 2 follow-up
gans in patients with PJS, inside and outside the gut. Intra- studies that included some who were members of the
tubular large-cell hyalinizing Sertoli cell neoplasms, ovarian original cohort.28,30 The lattermost report included some of
sex cord tumors, and adenoma malignum of the uterine the original 210 cases and expanded to 419 cases, in which
cervix, although not common, are linked to PJS25 (Table 3). 297 had confirmed germline pathogenic variants in
The lifetime risk of colon, stomach, and small bowel STK11.28 The cumulative incidences of cancer by decade
cancer has been estimated at 12%–39%, 29%, and 13% from age 20 to 70 years were 2%, 5%, 17%, 31%, 60%, and
respectively6,26–28 (Table 3). Most CRCs occur after the mid- 85% respectively, confirming the initial estimates and tu-
CLINICAL PRACTICE GUIDELINES

20s (range, 27–71 years, median age 46 years), but these mor spectrum—predominantly the gastrointestinal tract
malignancies have been reported in teenage years as well.26 and breast. Cancer risks were the same in those with a
Mean age at diagnosis of gastric adenocarcinoma ranges clinical diagnosis but no detectable germline pathogenic
between 30 and 40 years and of small intestinal cancer variant in the STK11 gene.
between 37 and 42 years.26,27,29 Several collaborative studies from Europe and the United
In a meta-analysis of 210 cases reported in 6 publica- States again found similar risks for cancer.29,31 A systematic
tions (retrospective cohort studies with kindred-based review of 20 cohort studies looking at 1644 patients with PJS
ascertainment from the United States, United Kingdom, confirmed at least 1 cancer in >90% of patients with PJS at a
and The Netherlands) on PJS that were based on clinical and mean age of 42 years.29 CRC was the most commonly diag-
histologic criteria with varying types of ascertainment, the nosed tumor, followed by cancers of the breast, small intes-
relative risk (RR) for any cancer was 15.2 (95% confidence tine, stomach, lung, pancreas, cervix, ovary, bile ducts, and
limits [CL], 2 and 19) compared with a variety of time testicles. A multicenter study from Italy of 119 STK11 path-
period–specific US-based registries. Significantly increased ogenic variant carriers reported an overall RR for cancer of
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2071

Table 3.Risk of Cancer in Hamartomatous Polyposis Syndromes

Site General population risk,a % Syndrome risk, % Mean age at diagnosis, y Reference

PJS
Colorectal 4.3 39 42–46 26, 29
Stomach <1 29 30–40 26, 29
Small bowel <1 13 37–42 26, 29
Breast 12.9 32–54 37–59 26, 28, 29
Ovarian (mostly SCTAT) 1.2 21 28 26
Cervix (adenoma malignum) <1 10–23 34–40 26
Uterus 3.1 9 43 26, 29
Pancreas 1.7 11–36 41–52 26, 28, 29, 32
Testicular (Sertoli cell tumor) <1 9 6-9 26, 29
Lung 6.3 7–17 47 26, 28, 29
JPS
Colon 4.3 39 44 86
Stomach <1 5–21 54 65, 67, 89
CS
Breast 12.9 25–85 38–46 100, 101, 102
Thyroid 1.3 3–38 31–38 100, 101, 102
Uterus 3.1 5–28 25 95, 100, 101, 102
Kidney (renal cell) 1.7 15–34 40 97, 100, 104
Colon 4.3 9–18 35 100–103, 106
Melanoma 2.3 6 3b 100, 101
HMPS
Colon 4.3 Increased — 119–121

SCTAT, sex cord tumors with annular tubules.


a
National Cancer Institute. Surveillance, Epidemiology, and End Results Cancer Statistics Review 1975–2017. Lifetime risk (%)
of being diagnosed with cancer by site, 2017.
b
Youngest age of onset.

22.0 in women (in part because of additional risks for cer- Gastrointestinal Polyposis and Cancer
vical cancers) and 8.6 in men, compared with an Italian-based Gastric, small bowel, and colorectal polyposis occur in
general population registry, and a cumulative risk of cancer 88%–100% of patients, with the majority appearing in the
reaching 89% by age 65 years.27 A more recent study from small bowel (60%–90%) and colon (50%–64%).36 Polyps
China confirmed elevated cancer risks (albeit somewhat can vary in number (1–100) and size (0.1–3 cm in diameter)
lower than the American and European data), early age of and age of onset of symptoms may vary. Polyp growth be-
onset, and a similar tumor spectrum.32 gins in childhood by age 10 years (33%), with most expe-
riencing symptoms such as bleeding, abdominal pain,
Surveillance of Affected Individuals intussusception, or obstruction (68%) by age 18 years.37 In
The effectiveness of cancer surveillance in PJS has not affected or at-risk individuals, early surveillance with
been evaluated in controlled studies. Consequently, sur- esophagogastroduodenoscopy (EGD), colonoscopy, and
veillance recommendations have been developed by small bowel imaging with video capsule endoscopy (VCE)
consensus groups and expert opinion analyzing cancer risks and/or magnetic resonance enterography (MRE) is recom-
and published organ-specific surveillance protocols mended to begin at age 8 years.11,33–35
CLINICAL PRACTICE GUIDELINES

(Tables 3 and 4).11,33–35 Intussusception is rare in children younger than 5 years,


and the precise risk of intussusception between 5 and 18
Question: How and when should small bowel surveillance years of age is unknown. Retrospective registry data report
be performed in Peutz-Jeghers syndrome? that 23 of 34 adults with PJS (68%) had undergone lapa-
Recommendation: We recommend that baseline small rotomy by the age of 18 years, 70% of which were per-
bowel surveillance using video capsule endoscopy or formed as an emergency. By the age of 10 years, 30% had
magnetic resonance enterography be performed
required a laparotomy.37 In a single-institution study of 379
between ages 8 and 10 years or earlier if the patient is
symptomatic. If no polyps are found at the initial pediatric patients who underwent pneumatic reduction for
examination, surveillance should resume at age 18 intussusception (from all causes), one-quarter required
years. Because of the risk of small bowel operative management.38 There is a paucity of studies that
intussusception, small bowel surveillance in adulthood is assess modalities to evaluate the small bowel in patients
recommended to continue throughout life every 2–3 with PJS. Retrospective data comparing VCE to small bowel
years. (Strong recommendation, low quality of evidence) barium studies have reported VCE as a useful diagnostic tool
CLINICAL PRACTICE GUIDELINES

2072 Boland et al
Table 4.Surveillance Guidelines in Hamartomatous Polyposis Syndromes

ACG 2015 NCCN 2020 ESPGHAN 2019 USMSTF 2020

Age of Age of Age of Age of Evidence


Examination initiation, y Interval, y initiation, y Interval, y initiation, y Interval, y initiation, y Interval, y grade

JPS
Colonoscopy 12–15 1–3 15 2–3 12–15 — 12–15 1–3 Low
Upper endoscopy 12–15 1–3 15 2–3 Late teensa — 12–15 1–3 Low
Screen for vascular lesions 6 mo — 6 mo — At diagnosis — 6 mob — —
PJS
Colonoscopy 8, 18c 3 Late teens 2–3 8 3 8–10, 18c 2–3 Very low
Upper endoscopy 8, 18 c 3 Late teens 2–3 8 3 8–10, 18c 2–3 Very low
VCE 8, 18 c 3 w8–10d 2–3 8 3 8, 18c 2–3 Low
CT or MRE of small bowel — — w8–10d 2–3 — — — — —
MRI/MRCP or EUS of pancreas 30 1–2 w30–35e 1–2 — — 35 1 Low
MRI and/or mammogram 25 1 w25 1 — — — — —
Physical examination Birth to teenage 1 w10 1 — — — — —
Pelvic examination and Pap smear 25f 1 w18–20 1 — — — — —
Testicular examination Birth to teenage 1 w10 1 — — — — —
PHTS
Colonoscopy 15 2 35 5 — — 35 5 Low
Upper endoscopy 15 2–3 — — — — — — —
Thyroid examination and US Adolescence 1 7 1 — — — — —
MRI and/or mammogram 30–35 1 30-35 1 — — — — —
Endometrial sampling 30–35 1 — 1–2 — — — — —
Urinalysis or renal US 18 1 40 1–2 — — — — —
Skin examination w18 1 At diagnosis 1 — — — — —

ACG, American College of Gastroenterology; CT, computed tomography; ESPGHN, European Society for Paediatric Gastroenterology Hepatology and Nutrition; EUS,
endoscopic ultrasound; MRCP, magnetic resonance cholangiopancreatography; NCCN, National Comprehensive Cancer Network.
a
Upper gastrointestingal screening in JPS SMAD4 carriers is indicated in asymptomatic patients starting in late teens. For non-SMAD4 JPS patients, upper endoscopic

Gastroenterology Vol. 162, No. 7


screening is only indicated if the patient has relevant symptoms or anemia not explained by colonic polyps.
b
SMAD4 mutation carriers should be clinically evaluated for HHT at the time of the diagnosis, including screening for, and appropriate management of, cerebral and
pulmonary AVMs.
c
First procedure at 8 years of age; if polyps present, repeat every 3 years; if no polyps, restart at 18 years of age and every 3 years.
d
Baseline at age 8–10 years of age with follow-up interval based on findings but at least by age 18 years of age, then every 2–3 years, although this may be individualized by
at least age 18 years, then every 2–3 years, although this may be individualized, or with symptoms).
e
Based on clinical judgment, early initiation age may be considered, such as 10 years younger than the earliest age of onset in the family.
f
ACG 2015 Guidelines recommend transvaginal ultrasound as part of surveillance beginning at age 25 years.
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2073

in PJS. A retrospective study from France included 27 chil- during EGD, and the remaining polyps were removed by
dren who underwent at least 1 VCE.39 The authors found means of enteroscopy or laparotomy of the small bowel.41 A
VCE was a useful diagnostic tool, however, findings at VCE substantial proportion of patients required intervention for
may not predict future bowel obstructions. Although VCE removal of large polyps and the authors concluded that
may not predict future bowel obstructions, it has a greater surveillance reduced polyp burden and likelihood of polyp-
sensitivity in detecting small bowel polyps compared with related complications, and provided cancer surveillance.
MRE and provides patients with another alternative if they Therefore, we recommend polypectomy for polyps in the
are unable to undergo MRE. Ten children with PJS with stomach and colorectum that are >0.5 cm in size on endo-
polyps >15 mm identified by VCE and MRE underwent scopic surveillance and an attempt to remove all polyps if
single-balloon enteroscopy.40 In this small study, single- endoscopically feasible.33,35,42,43
balloon enteroscopy was effective for treating small bowel In the small bowel, balloon enteroscopy and MRE have
polyps. Further larger, multicenter studies are warranted similar diagnostic yields for lesions 15 mm, but endoscopy
to accurately determine the safety of therapeutic single- permits polyp removal.43 Consequently, removal, preferably
balloon enteroscopy in children.7 Based on the pub- by enteroscopy, of small intestinal polyps that are symp-
lished data, VCE or MRE is recommended between ages 8 tomatic or rapidly growing, or asymptomatic polyps that are
and 10 years in asymptomatic patients. Investigations >1.0–1.5 cm in size, has been recommended.33,35,37,42,44,45
should commence earlier if patients have symptoms. If no Surgery is often needed when small bowel intussusception
polyps are found on the baseline investigation, repeat occurs. Some authorities recommend an attempt to clear the
small bowel evaluation may commence at age 18 years. If small intestine of polyps during laparotomy by means of
polyps are found, further investigation and surveillance intraoperative endoscopy with polypectomy or, for larger
should be individualized based on polyp size and location. polyps, by means of enterotomy. This “clean sweep”
Because of the risk of small bowel intussusception, small approach appears to decrease the need for recurrent small
bowel surveillance in adulthood continues throughout life bowel surgery.46 Thus, the early management focus is on
every 2–3 years. If polyps are present in the colon or large hamartomatous polyps and their tendency to obstruct
stomach, EGD or colonoscopy is continued as necessary the gut or bleed. As children grow older and transition into
every 2–3 years. adulthood, the focus shifts to managing the cancer risk.

Question: What is the recommended approach to Question: What size polyps found on small bowel
endoscopic surveillance of the stomach, duodenum, and imaging in Peutz-Jeghers syndrome should be removed?
colon in Peutz-Jeghers syndrome? Recommendation: We recommend polypectomy of
Recommendation: We suggest a baseline upper small bowel polyps that are symptomatic or 10 mm to
gastrointestinal endoscopy between the ages of 8 and prevent intussusception and other complications, such
10 years, which could be performed at the time of as bleeding. (Strong recommendation, low quality of
capsule placement for small bowel surveillance or if evidence)
polyps are identified on magnetic resonance
enterography. Although the initiation age for
colonoscopy remains uncertain, we also suggest
initiation of colonoscopy at the same time as Breast Cancer
esophagogastroduodenoscopy. In those in whom
characteristic polyps are detected, both colonoscopy Invasive ductal adenocarcinoma poses the greatest risk
and esophagogastroduodenoscopy should be repeated of early malignancy to patients with PJS (24%–54% lifetime
every 2–3 years. In those in whom there are no Peutz- risk in women) and often presents at a young age (range,
Jeghers polyps at baseline, surveillance is repeated at 19–61 years in 1 study).26,47 The risk of breast cancer in
age 18 years, or sooner should symptoms arise, and women with PJS is within the same range as women affected
then every 3 years. (Weak recommendation, very low by BRCA-1 or BRCA-2 pathogenic variants (40%–85% life-
quality of evidence)
time risk for breast cancer).26,48 Therefore, consensus
opinion surveillance recommendations by groups that
include breast cancer experts49,50 include monthly breast
CLINICAL PRACTICE GUIDELINES

self-examination starting at age 18 years, biannual clinical


Management of Polyposis breast examination starting at age 25 years, annual breast
Although the malignant potential of PJ polyps is un- magnetic resonance imaging (MRI) from ages 25–29 years,
known and the evidence of benefit from gastrointestinal and mammography with consideration of tomosynthesis (3-
surveillance is not robust, endoscopic removal of polyps is dimensional mammography) alternating every 6 months
recommended to prevent polyp-related complications and with breast MRI with contrast from ages 30 to 75 years.6,11
to reduce the risk of cancer. In a study of long-term out- The option of prophylactic mastectomy might be discussed
comes of gastrointestinal procedures from a PJ polyposis on a case-by-case basis, factoring in family history. In pa-
registry, investigators tracked the results of 776 procedures tients with PJS, referral to a breast cancer specialist for
among 63 patients with PJS at a median age of 20 years management of breast cancer surveillance is reasonable,
(range, 3–59 years). A total of 2461 polypectomies were and a multidisciplinary approach including a breast surgeon
performed; more than 1000 polyps were removed during is recommended when prophylactic mastectomy is being
colonoscopy and more than 400 polyps were removed considered.
2074 Boland et al Gastroenterology Vol. 162, No. 7

suspicion is required for diagnosis.56 Recommendations for


Question: What is the recommended pancreatic cancer gynecological surveillance are pelvic examination with Pap
surveillance in Peutz-Jeghers syndrome? smear and transvaginal ultrasound annually starting at age
Recommendation: We suggest annual pancreatic cancer 25 years.33,35
surveillance with either magnetic resonance
cholangiopancreatography or endoscopic ultrasound
starting at age 35 years. (Weak recommendation, low
Testicles
quality of evidence) The estimated risk of testicular cancer in male patients
with PJS is 9%, mean age at diagnosis is 9 years (range, 3–
20 years).6,26,27 These tumors present as testicular masses.
The tumors are Sertoli cell tumors that can cause gyneco-
Pancreas Cancer mastia and other signs of hyperestrogenism and occasion-
Pancreatic cancer is the third most frequently occurring ally virilization and/or accelerated height growth.26
malignancy in patients with PJS. The lifetime risk of Accelerated height velocity can be challenging to detect as
pancreatic ductal adenocarcinoma is between 11% and adolescents have “growth spurts” as part of normal matu-
36%, presenting, on average, at ages 41–52 years, with 95% ration and development. Expert opinion recommends
of cases occurring after age 24 years.6,26,28,51,52 PJS is annual history and physical examination (including self-
associated with the highest relative risk of all pancreatic examination) with observation for feminizing changes and
cancer syndromes, with the exception of hereditary examination of the testicles6,34,35; based on the range of age
pancreatitis (25%–40% lifetime risk).43 Consequently, a at diagnosis of this tumor, examination should start from
high-risk pancreatic surveillance protocol is recommended. birth. Ultrasound of the testicles every 2 years from birth to
The consensus recommendations of the International Can- age 12 years has been suggested.44
cer of the Pancreas Screening Consortium53 recommend
pancreas MRI/magnetic resonance cholangiopancreatog- Lung
raphy and/or endoscopic ultrasound every 1–2 years The lifetime risk of lung cancer in patients with PJS has
starting from age 40 years, for which evidence suggests that been estimated between 7% and 17%,6,26–28,30 compared
these cancers can be found in earlier stages.54 The National with 0.2%–0.6% in nonsmokers in the general population.57
Comprehensive Cancer Network Guidelines recommend The cumulative risk of lung cancer in PJS surpasses 5% by
initiating pancreatic cancer surveillance between the ages of age 55 years.6 Of note, lung cancer risk in patients with PJS
30 and 35 years. Due to reports of pancreatic cancer di- has not been calculated with adjustment for smoking status.
agnoses in patients with PJS before the age of 40 years,26 the The RRs of lung cancer in patients with PJS compared with
USMSTF suggests initiating annual surveillance at age 35 nonsmokers are similar to people with a more than 30 pack-
years with MRI/magnetic resonance cholangiopancreatog- year history of smoking who have quit for 10–15 years
raphy and/or endoscopic ultrasound. Ideally, these exami- (hazard ratio, 14.8).58 Currently, the American College of
nations would alternate on an annual basis, as they are Chest Physicians, American Society of Clinical Oncology, and
complementary. In addition, routine fasting glucose and American Cancer Society recommend low-dose computed
hemoglobin A1c at initiation of screening is recommended tomography annually for individuals with this level of cu-
by the Cancer of the Pancreas Screening Consortium. At the mulative risk for lung cancer from ages 55 to 74 years.59,60
current time, the USMSTF awaits definitive data before There are no data to show benefit of lung cancer surveil-
making a recommendation regarding fasting glucose and lance in patients with PJS. Lung cancer surveillance with
hemoglobin A1c. Recent updates to the Cancer of the annual low-dose computed tomography, as performed in
Pancreas Screening Guidelines detail surveillance protocols smokers at high risk for lung cancer, may be considered in
and management recommendations based on imaging and patients with PJS. Smoking cessation counseling in patients
endoscopic findings.55 with PJS is advisable to mitigate risk.33,34

Gynecological Cancers Chemoprevention


CLINICAL PRACTICE GUIDELINES

The lifetime risks for ovarian, uterine, and cervical can- Currently, there are no known chemopreventive agents
cer are estimated at 21%, 9%, and 10%–23%, respectively, in clinical practice that slow or prevent the development of
with mean age at presentation of 28–35 years, 43 years, and intestinal polyps and cancers in PJS. Pathogenic variants in
34–40 years, respectively.6,26,27 Of note, almost all ovarian the STK11 gene decrease inhibition of mTOR leading to the
tumors in patients with PJS are sex cord tumors with development of intestinal polyps. A trial examining the oral
annular tubules, and rarely cystadenomas or granulosa cell selective mTOR inhibitor, everolimus, was stopped prema-
tumors. Sex cord tumors with annular tubule neoplasms are turely because of poor patient accrual. Only 2 patients were
a heterogeneous group of benign or malignant neoplasms, enrolled, 1 with pancreatic cancer that progressed and
but the tumors rarely have lymph node metastasis.26 Also, another patient who withdrew from the protocol because of
an unusual percentage of cervical cancers in patients with severe complications from the medication.61
PJS are adenoma malignum, a rare, well-differentiated, cer- In a murine model of PJS, treatment with celecoxib, a
vical adenocarcinoma that is associated with a poor prog- COX2 inhibitor, resulted in a >50% reduction in polyp
nosis and difficult to diagnose on Pap smear. A high level of burden. When used in patients with PJS with diffuse
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2075

polyposis in the body of the stomach (tens to hundreds), 2 Juvenile polyps are most often solitary and are not
of 6 had a significant reduction in polyp number, as syndromic, occurring sporadically in infants and children. A
assessed by 5 independent evaluators, after administering typical history associated with a solitary juvenile polyp is
celecoxib (200 mg twice per day for 6 months).62 the asymptomatic passing of a polyp into an infant’s diaper.
Juvenile polyps appear endoscopically to have a smooth red
surface, may be sessile or pedunculated, and pathologically
Summary are characterized by cystic dilatation of mucus-filled glands
PJS is associated with a very high cumulative lifetime suspended in an inflamed stroma (Figures 1B and 2D and
risk of cancer of multiple organs, including, but not limited E). These lesions may also be called retention polyps or
to, the gastrointestinal tract. Intensive surveillance is rec- inflammatory polyps because of their microscopic
ommended to prevent and manage complications of polyp- appearance.
osis and identify cancer at an early stage. The development
of polyposis and the cancer risks may be a reflection of the
effects of haploinsufficiency of the LKB1 protein, for which Diagnosis
there are possible medical therapies to be explored. It is The diagnosis of JPS is made based on clinical criteria or
unclear whether the basic mechanisms responsible for identification of a germline pathogenic variant in SMAD4 or
hamartoma formation in younger life are the same as those BMPR1A. The clinical diagnosis of JPS is made when a per-
that create the risks for cancer later in life. Simulation son has any 1 of the following: 1) 5 or more juvenile polyps
models and clinical trials are needed to optimize endoscopic of the colon or rectum; 2) any number of juvenile polyps in
surveillance frequencies and the use of imaging modalities parts of the gastrointestinal tract other than the colon; or 3)
in adults. In addition, collaborative multicenter consortia any number of juvenile polyps and 1 or more first-degree
may help facilitate chemoprevention trials in the future. relatives with JPS.65

Genetics
Juvenile Polyposis Syndrome JPS is an autosomal dominant disorder with approxi-
mately 75% of cases inherited from a parent and 25%
representing de novo pathogenic variants.64 Approximately
Question: Who should undergo a genetic evaluation for 60% of patients with JPS have a pathogenic variant in the
juvenile polyposis syndrome?
BMPR1A or SMAD4 gene.67 Rarely (1 in 1,000,000) in-
Recommendation: We recommend genetic evaluation
for any individual with 1) 5 or more juvenile polyps of dividuals develop features of JPS and PHTS, known as ju-
the colon or rectum; 2) 2 or more juvenile polyps in venile polyposis of infancy, due to a large deletion
other parts of the gastrointestinal tract; or 3) any encompassing both the BMPR1A and PTEN genes.
number of juvenile polyps and 1 or more first-degree In a study from the Cleveland Clinic of 35 patients with
relatives with juvenile polyposis syndrome. (Strong JPS, germline pathogenic variants in SMAD4 and BMPR1A
recommendation, low quality of evidence) were associated with similar colonic polyposis phenotypes,
but SMAD4 pathogenic variant carriers were more likely to
have more gastric polyps, and an 11% risk of gastrointes-
tinal cancer.68 However, no gastric cancers were reported in
Clinical Features patients with BMPR1A pathogenic variants in 8 patients
JPS is an autosomal dominant inherited condition in followed for a mean of 11 years.
which multiple juvenile polyps are found in the colorectum However, germline pathogenic variants in other genes
(98% of affected patients) (Figure 1B), stomach (14%), may cause a hamartomatous polyp phenotype. In a study of
jejunum and ileum (7%), and duodenum (7%).63–65 The 49 patients referred to the Cleveland Clinic for unexplained
incidence of JPS is between 1 in 100,000 and 1 in 160,000 hamartomatous or hyperplastic polyps, germline pathogenic
individuals.65 The polyps in JPS vary in size from small variants were found in multiple genes, including endoglin
sessile nodules to pedunculated lesions that are 3 cm in (ENG, a gene associated with HHT), STK11 (the PJS gene),
CLINICAL PRACTICE GUIDELINES

diameter. Histologically, the typical juvenile polyp has a SMAD4, BMPR1A, and PTEN.69 In a later study from this
distinctive cystic architecture, dilated mucus-filled glands, a group of 603 patients with a “moderate load of gastroin-
prominent lamina propria, and Paneth cells enmeshed testinal polyps” with at least 1 confirmed to be a hamartoma
within a dense infiltration of inflammatory cells. In patients or hyperplastic polyp, 13% were found to have a germline
with germline pathogenic variants in SMAD4, additional pathogenic variant in at least 1 of the genes listed above and
features of gastric polyposis, gastric cancer, and a JPS– 20% of the cohort had a personal history of CRC.70
hereditary hemorrhagic telangiectasia (HHT) overlap syn- As discussed in the context of PJS, it has been tradi-
drome are common. The management of JPS is based on tionally thought that a germline pathogenic variant associ-
decreasing the risk of bleeding and gastric and colorectal ated with a hereditary colon cancer syndrome did not
cancer through polypectomy. Patients with JPS-HHT overlap change the biology of the normal tissues and a second (so-
syndrome should have lifelong HHT surveillance. One study matic) hit was required for a tumor to form. In fact, allelic
also demonstrated that as many as 38% may have thoracic loss of the SMAD4 locus was found in the epithelial
aorta abnormalities.66 component of juvenile polyps of patients with JPS (together
2076 Boland et al Gastroenterology Vol. 162, No. 7

with a germline pathogenic variant in SMAD4) using in situ Gastrointestinal Polyposis and Cancer
hybridization, but not in the inflammatory or stromal cells. In a retrospective chart review study of 257 children
This was compatible with the Knudson 2-hit model.71 with juvenile polyps at a single-referral center, patients
However, another group has reported that SMAD4 hap- presented at a median age of 5.6 years, and at colonoscopy
loinsufficiency (ie, a lower dose of the gene product due to 39% had multiple polyps.84 Among 192 patients who un-
the specific type of germline pathogenic variant) causes the derwent complete colonoscopy at initial diagnosis, 117
JPS phenotype in humans, supported by data from mice.72 (60.9%) had a single polyp and 75 (39.1%) had multiple
However, just as in PJS, being haploinsufficient for SMAD4 polyps. These lesions recurred in 21 of 47 patients (44.7%)
(ie, just the germline pathogenic variant), is associated with after an initial eradication, including 3 of 18 presenting with
partially diminished transforming growth factor–b a single polyp, and neoplasia was found in 3.9% of lesions.
signaling, at least in mice,72 as this alters proliferation in T Patients with JPS often have a variable presentation of polyp
cells, which contributes to the development of polyps and distribution, which may occur throughout the colon and/or
cancer.73,74 Specific deletion of SMAD4 in mouse T cells stomach and the cancer risk is attributable to the presence
leads to up-regulation of the Th17-inflammatory pathway in of dysplasia in the polyps. One study of 78 juvenile polyps
the stroma, and the growth of large polyps in the gastro- from 12 patients with JPS and 34 patients with sporadic
intestinal tracts of mice.73,74 This raises the question of juvenile polyps reported that dysplasia was present in 31%
whether there is abnormal regulation of immune cells in JPS, of the polyps from patients with JPS but in none of the
and may explain the inflammatory appearance of juvenile polyps from patients with sporadic juvenile polyps.85
polyps independent of abnormal biology in the epi- Dysplasia in polyps from patients with JPS was associated
thelium—another landscaper mechanism, as discussed with somatic variants in the APC gene.85 In a longitudinal
above for PJS. These observations may have important study from St Mark’s Hospital of 44 patients with JPS from
clinical implications for future attempts to halt the appear- 30 kindreds, a total of 787 polyps (juvenile and adenoma-
ance or growth of the inflammatory polyps. However, allelic tous) were resected, and 65 of 787 (8.3%) contained mild/
loss (ie, a somatic variants as the second hit) occurs in at moderate architectural dysplasia, and 20 additional polyps
least some polyps of JPS patients.75,76 (2.5%) were adenomatous.65
Patients with JPS are at increased risk for cancer prin-
cipally in the stomach and colon (Table 3). In a small cohort
Juvenile Polyposis Syndrome–Hereditary of patients with JPS (n ¼ 84) relative to age-, sex-, and race-
Hemorrhagic Telangiectasia Overlap Syndrome matched controls, the RR of CRC was estimated to be 34
HHT occurs in approximately 15%–81% of patients with (95% CL, 14.4 and 65.7), with a cumulative lifetime risk of
a germline SMAD4 pathogenic variant.77,78 The clinical fea- CRC reaching 38.7%.86 The CRCs were diagnosed at a mean
tures of HHT, such as epistaxis, obscure gastrointestinal age of 43.9 years. Interestingly, no other gastrointestinal
bleeding, digital clubbing, visceral arteriovenous malfor- cancers were noted in this cohort, but the numbers were
mations (AVMs), and mucocutaneous telangiectasias should small and the duration of the study was short. Individuals
be sought in SMAD4 patients. International guidelines with gastric polyposis, usually in association with patho-
(outlined below) recommend that surveillance and treat- genic variants in SMAD4 are also at risk for gastric cancer,
ment for HHT complications are necessary for all SMAD4 with the lifetime risk estimated to be at least 30% and
carriers.79 median age of diagnosis is 58 years; this has not been re-
ported in association with BMPR1A pathogenic var-
iants.67,87–91 Estimates of gastrointestinal cancer risk range
Juvenile Polyposis of Infancy from 11%68 to 55%,89 but none of the studies are pro-
Juvenile polyposis of infancy is a severe form of juvenile spective long-term studies (which would lead to un-
polyposis. This disease presents in the first 2 years of life derestimates), and all are prone to referral-based
with diarrhea, abdominal pain, rectal bleeding, refractory ascertainment bias (overestimates).
anemia, hypoalbuminemia, and enteropathy. Case reports
indicate that a large deletion in the long arm of chromosome Question: At what age should colonoscopic and upper
CLINICAL PRACTICE GUIDELINES

endoscopic surveillance begin in individuals identified


10 (10q23), encompassing the PTEN and BMPR1A genes, is
with juvenile polyposis syndrome?
associated with the development of the disease.69,80–83 Recommendation: We suggest initiating colonoscopic
and upper endoscopic surveillance at age 12–15 years,
Question: Which organs should undergo surveillance or earlier if symptomatic. Surveillance should be
when caring for a patient with juvenile polyposis repeated every 1–3 years depending on polyp burden.
syndrome? (Weak recommendation, low quality of evidence)
Recommendation: Patients with juvenile polyposis
syndrome are at increased risk for cancer in multiple
organs, including cancer of the colon and stomach.
Given this risk, we recommend patients with juvenile
polyposis syndrome undergo surveillance of the colon Management of Polyposis
and stomach. (Strong recommendation, low quality of
The goal of surveillance in JPS is to mitigate symptoms
evidence)
related to the disorder and decrease the risk of
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2077

complications from the manifestations, including cancer. Management of Juvenile Polyposis Syndrome–
Colonoscopy should be first performed at age 12–15 years Hereditary Hemorrhagic Telangiectasia
and repeated every 1–3 years, depending on polyp burden As recommended by HHT Foundation International, pa-
found, with removal of all polyps when feasible or at least tients with SMAD4 pathogenic variants should be screened
all polyps 5 mm. Upper endoscopy should be first per- for vascular findings associated with HHT.79,93 Children
formed at age 12–15 years and repeated every 1–3 years with possible or confirmed HHT should be screened for
depending on polyp burden found, with removal of polyps brain AVMs at the time of diagnosis and undergo at least 1
5 mm. Due to the possible presence of intestinal telangi- follow-up MRI at puberty because brain AVM development
ectasias, an annual history and physical examination and appears to correlate with times of growth. Lung AVM
complete blood counts to monitor for rectal bleeding and/or screening and surveillance is recommended at diagnosis
anemia should begin at age 12–15 years in patients with a and then every 3–5 years with pulse oximetry testing and
germline SMAD4 pathogenic variant. consideration of transthoracic contrast echocardiogram. In
Endoscopic polypectomy is recommended for colorectal adulthood, surveillance should include annual hemoglobin
polyposis management. Surgery with colectomy and ileor- or hematocrit for all patients older than 35 years. Trans-
ectal anastomosis is recommend for patients with CRC, thoracic contrast echocardiogram as a screen for pulmonary
endoscopically unmanageable colon polyp burden and AVMs should be performed at the time of diagnosis, within 5
uncontrolled anemia from colonic bleeding.33 In some years preceding planned pregnancy, after pregnancy, and
cases, proctocolectomy is necessary for rectal cancer or otherwise every 5–10 years. Physicians should consider
advanced polyp burden of the rectum. Colectomy in pa- referring these patients to HHT Centers of Excellence for
tients with JPS should be reserved for patients with polyp this evaluation. Brain MRI with and without contrast should
burdens that cannot be managed by polypectomy, persis- be performed at birth or at the time of diagnosis to screen
tent blood loss leading to severe anemia or hypo- for cerebral vascular malformations.
albuminemia, or cancer. A decision to proceed to colectomy
should be reviewed with gastroenterologists and surgeons
with expertise in caring for individuals with hereditary
Chemoprevention
polyposis syndromes. No known effective chemoprevention strategies exist for
The risk of gastric cancer is a concern in patients with the development of polyposis in patients with JPS.
JPS with SMAD4 pathogenic variants and gastric polyposis.
There is a paucity of published data evaluating the stomach Summary
and small bowel in pediatric patients with JPS. Those with BMPR1A and SMAD4 gene alterations are responsible for
upper gastrointestinal symptoms, or with anemia not JPS, yet only 60% of patients will have a pathogenic alter-
explained by colonic polyps, should undergo evaluation ation identified. The clinical overlap syndromes include JPS-
with upper endoscopy.92 In pediatric patients without HHT in patients with a SMAD4 pathogenic variant and ju-
SMAD4 pathogenic variants, according to the existing data, venile polyposis of infancy in patients with a combined
gastroscopy is not indicated unless the child has symp- BMPR1A and PTEN deletion. The symptoms of JPS are
toms.92 In asymptomatic patients with SMAD4 pathogenic usually related to bleeding from colorectal polyposis or in
variants, it is prudent to assess the upper tract between the SMAD4-related JPS to gastric polyposis or manifestations of
ages of 12 and 15 years. At the time of this publication, it is HHT. Cancer risk in JPS is elevated, mainly in the colon and
uncertain as to whether BMPR1A pathogenic variants are stomach (39%–68%), and is largely associated with SMAD4
associated with gastric cancer risk, and that pending new pathogenic variants.94 Excess risk of nongastrointestinal
evidence, upper endoscopy surveillance is suggested at cancer is not reported in JPS.
intervals similar to those recommended for SMAD4 carriers.
In adults, partial or complete gastrectomy is indicated in
patients with gastric cancer, high-grade dysplasia, inability PTEN-Hamartoma Tumor Syndrome
to adequately survey or endoscopically control polyposis, Clinical Features
persistent anemia or gastrointestinal bleeding from gastric
CLINICAL PRACTICE GUIDELINES

polyposis or angioectasia, symptoms of gastric outlet


obstruction, or protein-losing gastropathy.63,64
Question: Which gastrointestinal findings should prompt
a genetic evaluation for PTEN hamartoma tumor
syndrome?
Question: Which patients with juvenile polyposis Recommendation: We recommend individuals with
syndrome should undergo screening for hereditary multiple gastrointestinal hamartomas or
hemorrhagic telangiectasia? ganglioneuromas undergo genetic evaluation for
Recommendation: We suggest patients with SMAD4 Cowden’s syndrome and related conditions. (Strong
pathogenic variants be clinically evaluated for hereditary recommendation, low quality of evidence)
hemorrhagic telangiectasia at the time of the diagnosis,
including screening for and appropriate management of
cerebral and pulmonary arteriovenous malformations. PHTS includes a variety of phenotypic variations known
(Weak recommendation, low quality of evidence)
as CS, BRRS, and Proteus syndrome. The clinical diagnosis of
2078 Boland et al Gastroenterology Vol. 162, No. 7

the PHTS is made in patients meeting the revised diagnostic Consortium reported in 2012 on 368 individuals with
criteria, which include the presence of hamartomas of the germline pathogenic variants in PTEN. Elevated standard-
skin and gastrointestinal tract (see Figures 1D and 2F–H), ized incidence ratios (SIRs) were found for carcinomas of
mucocutaneous lesions, macrocephaly, and an increased the breast (SIR, 25.4; 95% CL, 19.8 and 32.0), thyroid (SIR,
risk of benign and malignant lesions of the breast, thyroid, 51.1; 95% CL, 38.1 and 67.1), endometrium (SIR, 42.9; 95%
and endometrium.95 CL, 28.1 and 62.8), colorectum (SIR, 10.3; 95% CL, 5.6 and
17.4), kidney (SIR, 30.6; 95% CL, 17.8 and 49.4), and mel-
Diagnosis anoma (SIR, 8.5; 95% CL, 4.1 and 15.6). This led to cumu-
The genetic diagnosis of PHTS is established with a lative lifetime risks for cancer of the breast at 85.2% (95%
germline pathogenic variant in the phosphatase and tensin CL, 71.4% and 99.1%), thyroid 35.2% (95% CL, 19.7% and
homolog (PTEN) gene. The clinical criteria for the diagnosis 50.7%), endometrium 28.2% (95% CL, 17.1% and 39.3%),
of CS is complex, and can be found at the National colorectum 9.0% (95% CL, 3.8% and 14.1%), kidney 33.6%
Comprehensive Cancer Network website.49 The discussion (95% CL, 10.4% and 56.9%), and melanoma 6% (95% CL,
for this article will focus on the manifestations and man- 1.6% and 9.4%).100
agement of patients with a confirmed diagnosis of PHTS. A multicenter study from France estimated cancer risks
in 154 patients with a germline pathogenic variant in
PTEN.101 SIRs for female breast cancer were 39.1 (95%
Genetics confidence interval [CI], 24.8–58.6), thyroid cancer 43.2
(95% CI, 19.7–82.1) in women and 199.5 (95% CI, 106.39–
342.03) in men, melanoma 28.3 (96% CI, 7.6–35.4) in
Question: Which organs should undergo surveillance for women and 39.4 (95% CI, 10.6–100.9) in men, and endo-
cancer when caring for a patient with PTEN hamartoma metrial cancer 48.7 (95% CI, 9.8–142.3). Estimated cumu-
tumor syndrome? lative lifetime risks by age 70 years were 85% for any
Recommendation: In PTEN hamartoma tumor cancer, 77% for female breast cancer, and 38% for thyroid
syndrome, patients are at increased risk for cancer in cancer. Median age for developing cancer was 36 years.
multiple organs, including cancer of the breast, thyroid,
kidney, uterus, colon, and skin. Although 85% were reported to have colonic polyps of any
Given this risk, we recommend a multidisciplinary variety, risks for gastrointestinal cancer were not reported
approach to cancer surveillance in these organs. (Strong to be elevated in this series. However, a review of 211 pa-
recommendation, low quality evidence) tients with CS from the Mayo Clinic, including a review of
the literature, reported a 16% lifetime risk for CRC. In this
study, only 46% had a proven germline pathogenic variant
The PTEN gene encodes a dual-function phosphatase in PTEN, and the cumulative risk for any cancer by age 70
that negatively regulates the growth-promoting activity of years was 89%.102
the phosphatidylinositol 3-kinase pathway. The PHTS In a study of 2548 patients who met “relaxed” Interna-
family of syndromes are caused by inactivating pathogenic tional Cowden’s Consortium criteria for CS with 5 or more
variants in PTEN, making it a classic tumor suppressor gastrointestinal polyps in which at least 1 was considered
gene.96 Germline pathogenic variants in the PTEN gene hyperplastic or hamartomatous, germline pathogenic vari-
lead to heterogeneous phenotypes called PHTS, which in- ants in PTEN were found in 127 (5%).103 At endoscopy,
cludes CS, BRRS, and some cases of the PTEN-related gastrointestinal polyps were found throughout the gut, and
Proteus syndrome (not considered further here).97,98 one-half were considered “hyperplastic.” In this group, 13%
Germline pathogenic variants in PTEN are associated of those who underwent colonoscopy developed CRC (7.1%
with other systemic nonpolyposis phenotypes, including of the entire series of patients), all before age 50 years (the
abnormalities of the central nervous system and skeleton, youngest was 35 years old), with a SIR for CRC of 224.1
but these will not be discussed here in detail. Patients with (95% CI, 109.3–411.3).
CS are also at risk for dysplastic cerebellar gangliocytoma A multicenter study from 9 countries of 180 carriers of
(Lhermitte-Duclos disease), but the specific genetic basis of germline pathogenic variants in PTEN estimated the cumu-
CLINICAL PRACTICE GUIDELINES

this and the other protean manifestations of germline lative risk of any cancer or Lhermitte-Duclos disease by age
pathogenic variants in PTEN are not yet understood. 60 years was 56% for men and 87% for women.104
Interestingly, gain-of-function germline pathogenic vari- Increased risk was reported for cancers of the breast, thy-
ants in the WWP1 gene, an E3 ubiquitin ligase commonly roid, endometrium, skin, kidneys, colorectum, and lungs. An
up-regulated in cancers, inhibits the activity of PTEN, earlier report from this group on 156 patients reported that
causes a CS-like syndrome (oligopolyposis and cancer- benign gastrointestinal polyps were found in 31% of pa-
prone phenotype), and provides some insight into what tients at a mean age of 38 years (range, 18–62 years) and
may be responsible for PHTS in the absence of germline most were considered “hamartomas.”105 The cumulative
variants in PTEN.99 risk of developing polyps was 70% by age 60 years. The
cumulative risk of developing CRC was 18% by age 60 years
Cancer Risk (occurring between ages 53 and 62 years), suggesting that
Cancer risks for patients with germline pathogenic var- surveillance colonoscopy might not be necessary in early
iants in PTEN are very high. The International Cowden adult life.
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2079

Gastrointestinal Polyps and Polyposis Surveillance of Affected Individuals


Gastrointestinal polyps are frequently found in patients Surveillance recommendations are provided in Table 4.
with germline pathogenic variants in PTEN, with variable The assessment at diagnosis of CS/PHTS should include a
prevalence. Estimates suggest that 35%–93% of patients complete (and especially dermatologic and neurologic)
with CS have gastrointestinal polyps, but in some reports clinical examination, mammography and breast MRI, thyroid
the germline basis of the disease was not known and a ultrasound, transvaginal ultrasound, upper gastrointestinal
uniform interpretation of nonadenomatous polyps had not endoscopy, colonoscopy, and renal ultrasound. Although
been established.103,106 The wide range of estimated polyp there are no data regarding risk-reduction surgery in
prevalence is probably a reflection of the heterogeneity of women with CS, the option for risk-reducing mastectomy
the disease and challenges in making definitive diagnoses and hysterectomy should be discussed on a case-by-case
outside of germline tests. Gastrointestinal polyps include basis.
hyperplastic polyps, inflammatory polyps, ganglioneuromas, The risk of CRC in patients with CS is estimated to be up
lipomas, adenomas, and the nonspecific term hamartomas. to 9%–18% lifetime risk with mean age at diagnosis of 44
A prospective study of 127 PTEN pathogenic variant years, but ranging from 35 to 49 years.100,102,103,105,114,115
carriers from 2548 subjects in an International Cowden Consequently, expert opinion recommends colonoscopy
Consortium study in which 69 underwent endoscopic starting at age 35 years unless symptomatic or if a close
studies found that 64 (93%) had gastrointestinal polyps.103 relative has had colon cancer before age 40 years, then start
Of that group, one-half had hyperplastic polyps, but all of 10 years before the earliest known colon cancer in the
the above-listed polyps (plus adenomas) were also found. family and repeat every 5 years or more frequently if the
The number of polyps ranged from 1 to “innumerable,” and patient is symptomatic or polyps are found. This recom-
were distributed throughout the gut. mendation differs from a recent American College of
BRRS is caused by germline pathogenic variants in the Gastroenterology Guideline,32 which recommends initiating
PTEN gene.98,107 It is a pediatric condition associated with colonoscopy at age 15 years. Recent evidence suggesting
macrocephaly, developmental delay, gastrointestinal later onset of significant colon cancer risk informed our
hamartomatous polyps, and pigmented macules on the toes recommendations.
and glans penis. It has also been called the Bannayan- Colectomy is rarely required in patients with CS and is
Zonana or Ruvalcaba-Riley-Smith syndrome. Inexplicably, reserved for patients with CRC or in whom surveillance and
members of the same family may have features of either clearing of premalignant lesions is not endoscopically
BRRS or CS,108 and some cases of BRRS do not have feasible.
detectable pathogenic variants in PTEN.109 Confusing over-
lap between CS and JPS can occur when, as mentioned
above, a chromosomal deletion occurs on chromosome Breast Cancer
10q22.3-q24.1. This leads to the loss of both PTEN and The management of breast cancer risk begins at age 25
BMPR1A28; a situation that can also look like BRRS, also years with clinical breast examination every 6–12 months;
known as juvenile polyposis of infancy (see section on annual mammography and breast MRI surveillance starting
JPS).110,111 between ages 30 and 35 years or 5–10 years before the
In a smaller study of 19 patients with CS (in which only earliest known breast cancer in the family.49 In patients with
12 were shown to have germline pathogenic variants in PHTS, referral to a breast cancer specialist for management of
PTEN), pan-colonic polyps were found in 58%, pan- breast cancer surveillance is reasonable and a multidisci-
gastrointestinal polyps in 42%, and the pathological inter- plinary approach, including a breast surgeon, is recom-
pretation of the polyps included inflammatory polyps in mended when prophylactic mastectomy is being considered.
95%, but also adenomas, lipomas, and ganglioneuromas.112
Moreover, there was more than 1 pathological variety in
79% of patients, indicating the clinical heterogeneity in this Endometrial Cancer
entity. Esophageal glycogenic acanthosis is also found in The management of endometrial cancer risk begins be-
PHTS, which is a benign condition and there is no reported tween the ages of 30 and 35 years. Endometrial cancer can
CLINICAL PRACTICE GUIDELINES

increased risk for esophageal cancer.113 often be detected early on the basis of symptoms, and women
should be educated regarding seeking medical attention on
the basis of symptoms, including abnormal uterine bleeding
Question: What is the recommended colonoscopic or postmenopausal bleeding. Endometrial biopsy is sensitive
surveillance in individuals identified with PTEN and specific for endometrial cancer and surveillance via bi-
hamartoma tumor syndrome? opsy every 1–2 years can be considered.33,49
Recommendation: We suggest colonoscopy
surveillance to begin at age 35 years (or 10 years
younger than age of any relative with colorectal cancer), Thyroid Cancer
repeated at intervals no greater than 5 years, depending An annual thyroid ultrasound should be performed,
on polyp burden. (Weak recommendation, low quality of
evidence)
beginning at the time of PHTS diagnosis (including in
childhood).33,49
2080 Boland et al Gastroenterology Vol. 162, No. 7

Renal Cancer studies can be used to help clarify the benefits and risks of
The recommended management of kidney cancer risk is various interventions and surveillance programs. The
an annual renal ultrasound and/or renal MRI starting at age management of these diseases changes dramatically when
40 years if there is a family history of renal cancer, or every the patient matures from childhood to adulthood—which is
2 years if not.33,49 where almost all of the cancer risk lies. Important knowl-
edge gaps are listed in Table 5 and expanded upon below.
Melanoma
The management of melanoma risk includes an annual Peutz-Jeghers Syndrome
clinical skin examination beginning at age 18 years.33 As indicated above, STK11/LKB1 pathogenic variants
result in up-regulation of inflammatory cytokines and pro-
Summary motion of overgrowth of both stromal and normal gastro-
Germline pathogenic variants in PTEN are associated intestinal epithelium, driving polyp formation (Table 5). Up-
with variety of gastrointestinal hamartomatous polyps and a regulation of cytokines is associated with hyperactivation of
markedly increased risk of cancer, but largely in non- JAK-STAT, which contributes to inflammation and cancer.
gastrointestinal organs. There is a moderate increase in the Inhibition of JAK-STAT significantly reduced polyp growth
risk of CRC, but it may be that the age of risk is such that in mice. Consequently, the JAK inhibitor ruxolitinib (already
colonoscopic surveillance can be withheld until age 35 years clinically approved for myeloproliferative disease) may have
or later or 10 years younger than the age of the youngest therapeutic potential in patients with PJS.21 Also, LKB1 ac-
relative with CRC. However, the reports of CRC risk have tivity inhibits the activation of adenosine monophosphate–
been variable,95,116 and no surveillance recommendation dependent protein kinase. Loss of LKB1 activity results in
has been rigorously evaluated. The principal organs at risk adenosine monophosphate–dependent protein kinase acti-
for cancer include the breast, thyroid, endometrium, and vation with up-regulation of mTORC1 signaling contributing
colorectum. to growth of PJS polyposis. Targeting of adenosine
monophosphate–dependent protein kinase activation is
currently being investigated.123
Hereditary Mixed Polyposis Syndrome
The Genetic Basis of the Disease and Cancer
Juvenile Polyposis Syndrome
Risk Of note is the mouse model of juvenile polyposis created
HMPS is a rare autosomal dominant disease reported in by a conditional knockout of the SMAD4 gene only in T-cell
only a few families. It is characterized by attenuated colonic populations.73 These animals spontaneously developed
polyposis. The polyps include adenomas, hyperplastic, and a massive polyps within the gastroduodenal region. The
particular polyp with an admixture of variable histologies epithelium in the polyps contained increased expression of
including adenomatous, hyperplastic, juvenile, and mixed pro-inflammatory cytokines, particularly interleukin-11, a
polyps.117 In the original HMPS kindred described by cytokine known to promote gastric epithelial cell survival
Whitelaw et al,118 13 members were diagnosed with CRC and hyperplasia with evidence of increased TH17 cell ac-
and 23 developed multiple polyps of several different his- tivity. This suggests possible therapeutic targets for the
tologic types (adenomatous, hyperplastic, and juvenile). future chemoprevention of juvenile polyposis.
HMPS has been associated with large duplications of the
promoter region or entire GREM1 gene.119–121 These un-
usual pathogenic variants increase the expression of the
gene product, which then inhibits the bone morphogenetic Summary and Conclusions
protein (BMP) pathway.119 This promoter duplication was Among the gastrointestinal hamartomatous polyposis
found in 0.7% of Ashkenazi Jews in Israel who met clinical syndromes, PJS is the best understood. The polyps are
criteria for Lynch syndrome.120 The largest series reported readily identified as PJ polyps pathologically, and only 1
4 families with 16 affected members; the onset of polyposis known gene (STK11) is associated with this entity. The
CLINICAL PRACTICE GUIDELINES

starts in the late 20s, which is when colonoscopic surveil- phenotype is clinically distinct. Not all patients (or families)
lance should begin.122 There are not enough data to know have detectable germline pathogenic variants in STK11, so
the optimal surveillance intervals or whether extraintestinal additional genes in this pathway are possibly contributors
neoplasia is a risk. The underlying genetic basis of the ma- to this entity. The polyps may evolve through an expansion
jority of HMPS families is unknown. of elements of the gut stroma due to haploinsufficiency of
STK11. In children, the main risks are for gastrointestinal
obstruction and bleeding. Later in adult life, a very high risk
Future Research Considerations of intestinal and extraintestinal cancers exists. The major
The hamartoma syndromes are rare and it is difficult for organs at risk for cancer (in order of decreasing relative
single institutions or even collaborative centers to accu- risk) include the small intestine, stomach, pancreas, colon,
mulate enough cases and follow them prospectively long esophagus, ovary, lung, uterus, and breast, with estimated
enough to develop robust conclusions about cancer risk. lifetime risks for any cancer reaching >90%. The ovaries
Modeling, simulation, and collaborative multicenter clinical and testes are also at risk for rare variant tumors. The
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2081

Table 5.Areas Requiring Further Investigation in Hamartomatous Polyposis Syndromes

1. Discover the germline variants in families with clinically recognizable syndromes who do not have germline mutations in the genes known
to be associated with these syndromes.
2. Understand the effects of haplo-insufficiency of STK11 in PJS and the SMAD4 in JPS on the development of polyposis.
3. Identify safe and effective pharmacological interventions for the inhibition of polyp formation children and adults with the hamartomatous
polyposis syndromes.
4. Determine whether pharmacological intervention can mitigate cancer risk in PJS and JPS in adults (independent of the polyposis risk).
5. Develop simulation models in adults with a hamartomatous polyposis syndrome to determine the optimal endoscopic frequency.
6. Design and implement studies to determine the best imaging modalities and surveillance intervals in adults with a hamartomatous pol-
yposis syndrome.
7. Determine whether the hamartomatous phenotype in pediatric patients predicts the cancer risk in adulthood.
8. Perform outcomes studies evaluating the transition of care in pediatric polyposis patients entering adult life, including outcomes such as
compliance with surveillance.
9. Determine difference in phenotype and cancer risk in individuals who meet clinical criteria, but do not have an identifiable pathogenic
variant compared to those with a germline pathogenic variant in a hamartomatous polyposis syndrome.
10. Determine the lifetime cancer risks in patients with a pathogenic variant, but without clinical manifestations (those identified incidentally on
multigene panel testing).
11. Quantify the frequency of mosaicism as a cause of the hamartomatous polyposis syndromes.
12. Determine the optimal modality of small bowel evaluation in individuals with PJS.

screening and surveillance strategies change dramatically patients and families with unusual phenotypes where no
when the children reach adulthood. genotype can be found.
JPS has principal risks for obstruction and bleeding in
the pediatric ages, but the management in adults shifts to
cancer risks in the stomach and colon. Importantly, unlike
References
PJS, the gastrointestinal cancers tend to arise within the 1. Guyatt GH, Oxman AD, Kunz R, et al. What is "quality of
juvenile polyps, suggesting that polyp removal might pre- evidence" and why is it important to clinicians? BMJ
2008;336:995–998.
vent cancer. Cancer risk is linked to germline pathogenic
variants in the SMAD4 gene rather than the BMPR1A gene. 2. Lu KH, Wood ME, Daniels M, et al. American Society of
Clinical Oncology Expert Statement: collection and use
The polyps are histologically inflammatory in nature, and
of a cancer family history for oncology providers. J Clin
their growth may be driven by haploinsufficiency of SMAD4
Oncol 2014;32:833–840.
in the immune cells of the polyp stroma. The management
3. Hampel H, Bennett RL, Buchanan A, et al. A practice
focuses on the polyps in children and cancers later in life,
guideline from the American College of Medical Genetics
when endoscopic approaches are the mainstay of
and Genomics and the National Society of Genetic
surveillance.
Counselors: referral indications for cancer predisposition
The hamartoma syndromes associated with germline assessment. Genet Med 2015;17:70–87.
pathogenic variants in PTEN raise a completely different
4. Heald B, Hampel H, Church J, et al. Collaborative Group
clinical challenge. Although nearly all patients with PHTS of the Americas on Inherited Gastrointestinal Cancer
have a variety of different hamartomatous gastrointestinal Position statement on multigene panel testing for pa-
polyps, studies suggest an increased risk of cancer in this tients with colorectal cancer and/or polyposis. Fam
setting.100,103 The extracolonic cancer risks are greatest for Cancer 2020;19:223–239.
the breast, thyroid, melanoma, and endometrium. The ab- 5. Attard TM, Burke CA, Hyer W, et al. ACG clinical report
solute risk for CRC appears to be increased, ranging from and recommendations on transition of care in children
9% to 18% during a lifetime, and the rest of the gastroin- and adolescents with hereditary polyposis syndromes.
CLINICAL PRACTICE GUIDELINES

testinal tract does not have an established increase in cancer Am J Gastroenterol 2021;116:638–646.
risk. The age for beginning surveillance for CRC remains to 6. van Lier MG, Wagner A, Mathus-Vliegen EM, et al. High
be determined, but the early onset of CRC provides some cancer risk in Peutz-Jeghers syndrome: a systematic
suggestions. Be aware that the PTEN gene and BMPR1A are review and surveillance recommendations. Am J Gas-
located near one another on chromosome 10q, and that troenterol 2010;105:1258–1264; author reply 1265.
large-scale chromosomal deletions could adversely affect 7. Latchford AR, Phillips RK. Gastrointestinal polyps and
both genes, complicating the clinical picture. Long-term cancer in Peutz-Jeghers syndrome: clinical aspects.
prospective studies of mutation carriers are still needed to Fam Cancer 2011;10:455–461.
further clarify the risk of cancer and the role of surveillance 8. Shaco-Levy R, Jasperson KW, Martin K, et al. Morpho-
in these syndromes. With increases in genetic testing and logic characterization of hamartomatous gastrointestinal
evaluation, future studies will be conducted with more polyps in Cowden syndrome, Peutz-Jeghers syndrome,
robust cohorts of genetically characterized, less heteroge- and juvenile polyposis syndrome. Hum Pathol 2016;
neous populations. However, there is also a need to study 49:39–48.
2082 Boland et al Gastroenterology Vol. 162, No. 7

9. Menko FH. LKB1/ STK11, Peutz-Jeghers syndrome and 28. Hearle N, Schumacher V, Menko FH, et al. Frequency
cancer. Fam Cancer 2011;10:413–414. and spectrum of cancers in the Peutz-Jeghers syn-
10. Jass JR. Colorectal polyposes: from phenotype to drome. Clin Cancer Res 2006;12:3209–3215.
diagnosis. Pathol Res Pract 2008;204:431–447. 29. van Lier MG, Westerman AM, Wagner A, et al. High
11. Beggs AD, Latchford AR, Vasen HF, et al. Peutz-Jeghers cancer risk and increased mortality in patients with
syndrome: a systematic review and recommendations Peutz-Jeghers syndrome. Gut 2011;60:141–147.
for management. Gut 2010;59:975–986. 30. Lim W, Olschwang S, Keller JJ, et al. Relative frequency
12. Amos CI, Bali D, Thiel TJ, et al. Fine mapping of a ge- and morphology of cancers in STK11 mutation carriers.
netic locus for Peutz-Jeghers syndrome on chromosome Gastroenterology 2004;126:1788–1794.
19p. Cancer Res 1997;57:3653–3656. 31. Mehenni H, Resta N, Park JG, et al. Cancer risks in LKB1
13. Hemminki A, Markie D, Tomlinson I, et al. A serine/ germline mutation carriers. Gut 2006;55. 984–590.
threonine kinase gene defective in Peutz-Jeghers syn- 32. Chen HY, Jin XW, Li BR, et al. Cancer risk in patients
drome. Nature 1998;391:184–187. with Peutz-Jeghers syndrome: a retrospective cohort
14. Hardie DG. The LKB1-AMPK pathway-friend or foe in study of 336 cases. Tumour Biol 2017;39(6):
cancer? Cancer Cell 2013;23:131–132. 1010428317705131.
15. Shaw RJ, Bardeesy N, Manning BD, et al. The LKB1 33. Syngal S, Brand RE, Church JM, et al. ACG clinical
tumor suppressor negatively regulates mTOR signaling. guideline: genetic testing and management of hereditary
Cancer Cell 2004;6:91–99. gastrointestinal cancer syndromes. Am J Gastroenterol
16. Knudson AG Jr. Mutation and cancer: statistical study of 2015;110:223–262; quiz 263.
retinoblastoma. Proc Natl Acad Sci U S A 1971; 34. Gupta S, Provenzale D, Regenbogen SE, et al. NCCN
68:820–823. Guidelines Insights: Genetic/Familial High-Risk Assess-
17. Hernan I, Roig I, Martin B, et al. De novo germline mu- ment: Colorectal, Version 3.2017. J Natl Compr Cancer
tation in the serine-threonine kinase STK11/LKB1 gene Netw 2017;15:1465–1475.
associated with Peutz-Jeghers syndrome. Clin Genet 35. Giardiello FM, Trimbath JD. Peutz-Jeghers syndrome
2004;66:58–62. and management recommendations. Clin Gastroenterol
18. Katajisto P, Vaahtomeri K, Ekman N, et al. LKB1 Hepatol 2006;4:408–415.
signaling in mesenchymal cells required for suppression 36. Utsunomiya J, Gocho H, Miyanaga T, et al. Peutz-
of gastrointestinal polyposis. Nat Genet 2008; Jeghers syndrome: its natural course and management.
40:455–459. Johns Hopkins Med J 1975;136:71–82.
19. Poffenberger MC, Metcalfe-Roach A, Aguilar E, et al. 37. Hinds R, Philp C, Hyer W, et al. Complications of child-
LKB1 deficiency in T cells promotes the development of hood Peutz-Jeghers syndrome: implications for pediatric
gastrointestinal polyposis. Science 2018;361:406–411. screening. J Pediatr Gastroenterol Nutr 2004;
20. Ollila S, Domenech-Moreno E, Laajanen K, et al. Stromal 39:219–220.
Lkb1 deficiency leads to gastrointestinal tumorigenesis 38. Fallon SC, Lopez ME, Zhang W, et al. Risk factors for
involving the IL-11-JAK/STAT3 pathway. J Clin Invest surgery in pediatric intussusception in the era of pneu-
2018;128:402–414. matic reduction. J Pediatr Surg 2013;48:1032–1036.
21. Hollstein PE, Shaw RJ. Inflamed T cells and stroma drive 39. Gastineau S, Viala J, Caldari D, et al. Contribution of
gut tumors. Science 2018;361:332–333. capsule endoscopy to Peutz-Jeghers syndrome man-
22. Amos CI, Keitheri-Cheteri MB, Sabripour M, et al. Ge- agement in children. Dig Liver Dis 2012;44:839–843.
notype-phenotype correlations in Peutz-Jeghers syn- 40. Bizzarri B, Borrelli O, de’Angelis N, et al. Management of
drome. J Med Genet 2004;41:327–333. duodenal-jejunal polyps in children with peutz-jeghers
23. Borun P, De Rosa M, Nedoszytko B, et al. Specific Alu syndrome with single-balloon enteroscopy. J Pediatr
elements involved in a significant percentage of copy Gastroenterol Nutr 2014;59:49–53.
number variations of the STK11 gene in patients with 41. Latchford AR, Neale K, Phillips RK, et al. Peutz-Jeghers
Peutz-Jeghers syndrome. Fam Cancer 2015; syndrome: intriguing suggestion of gastrointestinal
14:455–461. cancer prevention from surveillance. Dis Colon Rectum
24. Zbuk KM, Eng C. Hamartomatous polyposis syndromes. 2011;54:1547–1551.
CLINICAL PRACTICE GUIDELINES

Nat Clin Pract Gastroenterol Hepatol 2007;4:492–502. 42. Tomlinson IP, Houlston RS. Peutz-Jeghers syndrome.
25. Idrees MT, Ulbright TM, Oliva E, et al. The World Health J Med Genet 1997;34:1007–1011.
Organization 2016 classification of testicular non-germ 43. Goverde A, Korsse SE, Wagner A, et al. Small-bowel
cell tumours: a review and update from the Interna- surveillance in patients with Peutz-Jeghers syndrome:
tional Society of Urological Pathology Testis Consulta- comparing magnetic resonance enteroclysis and double
tion Panel. Histopathology 2017;70:513–521. balloon enteroscopy. J Clin Gastroenterol 2017;
26. Giardiello FM, Brensinger JD, Tersmette AC, et al. Very 51:e27–e33.
high risk of cancer in familial Peutz-Jeghers syndrome. 44. McGrath DR, Spigelman AD. Preventive measures in
Gastroenterology 2000;119:1447–1453. Peutz-Jeghers syndrome. Fam Cancer 2001;1:121–125.
27. Resta N, Pierannunzio D, Lenato GM, et al. Cancer risk 45. Ohmiya N, Taguchi A, Shirai K, et al. Endoscopic
associated with STK11/LKB1 germline mutations in resection of Peutz-Jeghers polyps throughout the small
Peutz-Jeghers syndrome patients: results of an Italian intestine at double-balloon enteroscopy without lapa-
multicenter study. Dig Liver Dis 2013;45:606–611. rotomy. Gastrointest Endosc 2005;61:140–147.
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2083

46. Oncel M, Remzi FH, Church JM, et al. Benefits of ’clean 62. Udd L, Katajisto P, Rossi DJ, et al. Suppression of
sweep’ in Peutz-Jeghers patients. Colorectal Dis 2004; Peutz-Jeghers polyposis by inhibition of cyclo-
6:332–335. oxygenase-2. Gastroenterology 2004;127:1030–1037.
47. van Lier MG, Mathus-Vliegen EM, Wagner A, et al. High 63. Chow E, Macrae F. A review of juvenile polyposis syn-
cumulative risk of intussusception in patients with Peutz- drome. J Gastroenterol Hepatol 2005;20:1634–1640.
Jeghers syndrome: time to update surveillance guide- 64. Schreibman IR, Baker M, Amos C, et al. The hamar-
lines? Am J Gastroenterol 2011;106:940–945. tomatous polyposis syndromes: a clinical and molecular
48. Hamdi Y, Soucy P, Kuchenbaeker KB, et al. Association review. Am J Gastroenterol 2005;100:476–490.
of breast cancer risk in BRCA1 and BRCA2 mutation 65. Latchford AR, Neale K, Phillips RK, et al. Juvenile pol-
carriers with genetic variants showing differential allelic yposis syndrome: a study of genotype, phenotype, and
expression: identification of a modifier of breast cancer long-term outcome. Dis Colon Rectum 2012;
risk at locus 11q22.3. Breast Cancer Res Treat 2017; 55:1038–1043.
161:117–134. 66. Heald B, Rigelsky C, Moran R, et al. Prevalence of
49. Daly MB, Pilarski R, Yurgelun MB, et al. NCCN Guide- thoracic aortopathy in patients with juvenile polyposis
lines Insights: Genetic/Familial High-Risk Assessment: syndrome-hereditary hemorrhagic telangiectasia due to
Breast, Ovarian, and Pancreatic, Version 1.2020. J Natl SMAD4. Am J Med Genet A 2015;167A:1758–1762.
Compr Canc Netw 2020;18:380–391. 67. Aretz S, Stienen D, Uhlhaas S, et al. High proportion of
50. Manahan ER, Kuerer HM, Sebastian M, et al. Consensus large genomic deletions and a genotype phenotype
Guidelines on Genetic` Testing for Hereditary Breast update in 80 unrelated families with juvenile polyposis
Cancer from the American Society of Breast Surgeons. syndrome. J Med Genet 2007;44:702–709.
Ann Surg Oncol 2019;26:3025–3031. 68. Aytac E, Sulu B, Heald B, et al. Genotype-defined cancer
51. Korsse SE, Harinck F, van Lier MG, et al. Pancreatic risk in juvenile polyposis syndrome. Br J Surg 2015;
cancer risk in Peutz-Jeghers syndrome patients: a large 102:114–118.
cohort study and implications for surveillance. J Med 69. Sweet K, Willis J, Zhou XP, et al. Molecular classification
Genet 2013;50:59–64. of patients with unexplained hamartomatous and hy-
52. Chen KT. Female genital tract tumors in Peutz-Jeghers perplastic polyposis. JAMA 2005;294:2465–2473.
syndrome. Human Pathol 1986;17:858–861. 70. Ngeow J, Heald B, Rybicki LA, et al. Prevalence of
53. Canto MI, Harinck F, Hruban RH, et al. International germline PTEN, BMPR1A, SMAD4, STK11, and ENG
Cancer of the Pancreas Screening (CAPS) Consortium mutations in patients with moderate-load colorectal
summit on the management of patients with increased polyps. Gastroenterology 2013;144:1402–1409; 1409.
risk for familial pancreatic cancer. Gut 2013;62:339–347. e1–e5.
54. Canto MI, Almario JA, Schulick RD, et al. Risk of 71. Woodford-Richens K, Bevan S, Churchman M, et al.
neoplastic progression in individuals at high risk for Analysis of genetic and phenotypic heterogeneity in ju-
pancreatic cancer undergoing long-term surveillance. venile polyposis. Gut 2000;46:656–660.
Gastroenterology 2018;155:740–751.e2. 72. Alberici P, Gaspar C, Franken P, et al. Smad4 hap-
55. Goggins M, Overbeek KA, Brand R, et al. Management loinsufficiency: a matter of dosage. Pathogenetics 2008;
of patients with increased risk for familial pancreatic 1(1):2.
cancer: updated recommendations from the Interna- 73. Hahn JN, Falck VG, Jirik FR. Smad4 deficiency in T cells
tional Cancer of the Pancreas Screening (CAPS) Con- leads to the Th17-associated development of premalig-
sortium. Gut 2020;69:7–17. nant gastroduodenal lesions in mice. J Clin Invest 2011;
56. McGowan L, Young RH, Scully RE. Peutz-Jeghers syn- 121:4030–4042.
drome with "adenoma malignum" of the cervix. A report 74. Kim BG, Li C, Qiao W, et al. Smad4 signalling in T cells is
of two cases. Gynecol Oncol 1980;10:125–133. required for suppression of gastrointestinal cancer. Na-
57. Crispo A, Brennan P, Jockel KH, et al. The cumulative risk ture 2006;441:1015–1019.
of lung cancer among current, ex- and never-smokers in 75. Woodford-Richens K, Williamson J, Bevan S, et al. Allelic
European men. Br J Cancer 2004;91:1280–1286. loss at SMAD4 in polyps from juvenile polyposis patients
58. Pinsky PF, Zhu CS, Kramer BS. Lung cancer risk by and use of fluorescence in situ hybridization to demon-
CLINICAL PRACTICE GUIDELINES

years since quitting in 30þ pack year smokers. J Med strate clonal origin of the epithelium. Cancer Res 2000;
Screen 2015;22:151–157. 60:2477–2482.
59. Detterbeck FC, Mazzone PJ, Naidich DP, et al. 76. Blatter RH, Plasilova M, Wenzel F, et al. Somatic alter-
Screening for lung cancer: diagnosis and management ations in juvenile polyps from BMPR1A and SMAD4
of lung cancer, 3rd ed: American College of Chest mutation carriers. Genes Chromosomes Cancer 2015;
Physicians evidence-based clinical practice guidelines. 54:575–582.
Chest 2013;143(Suppl):e78S–e92. 77. O’Malley M, LaGuardia L, Kalady MF, et al. The preva-
60. Wender R, Fontham ET, Barrera E Jr, et al. American lence of hereditary hemorrhagic telangiectasia in juvenile
Cancer Society lung cancer screening guidelines. CA polyposis syndrome. Dis Colon Rectum 2012;
Cancer J Clin 2013;63:107–117. 55:886–892.
61. de Brabander J, Eskens F, Korsse SE, et al. Chemo- 78. Gallione CJ, Richards JA, Letteboer TG, et al. SMAD4
prevention in patients with Peutz-Jeghers syndrome: mutations found in unselected HHT patients. J Med
lessons learned. Oncologist 2018;23:399.e33. Genet 2006;43:793–797.
2084 Boland et al Gastroenterology Vol. 162, No. 7

79. Faughnan ME, Palda VA, Garcia-Tsao G, et al. Interna- review and revised diagnostic criteria. J Natl Cancer Inst
tional guidelines for the diagnosis and management of 2013;105:1607–1616.
hereditary haemorrhagic telangiectasia. J Med Genet 96. Chalhoub N, Baker SJ. PTEN and the PI3-kinase
2011;48:73–87. pathway in cancer. Annu Rev Pathol 2009;4:127–150.
80. Oliveira PH, Cunha C, Almeida S, et al. Juvenile polyp- 97. Liaw D, Marsh DJ, Li J, et al. Germline mutations of the
osis of infancy in a child with deletion of BMPR1A and PTEN gene in Cowden disease, an inherited breast and
PTEN genes: surgical approach. J Pediatr Surg 2013; thyroid cancer syndrome. Nat Genet 1997;16:64–67.
48:e33–e37. 98. Marsh DJ, Coulon V, Lunetta KL, et al. Mutation spec-
81. Tsuchiya KD, Wiesner G, Cassidy SB, et al. Deletion trum and genotype-phenotype analyses in Cowden
10q23.2-q23.33 in a patient with gastrointestinal juvenile disease and Bannayan-Zonana syndrome, two hamar-
polyposis and other features of a Cowden-like syn- toma syndromes with germline PTEN mutation. Hum Mol
drome. Genes Chromosomes Cancer 1998;21:113–118. Genet 1998;7:507–515.
82. Delnatte C, Sanlaville D, Mougenot JF, et al. Contiguous 99. Lee YR, Yehia L, Kishikawa T, et al. WWP1 Gain-of-
gene deletion within chromosome arm 10q is associated function inactivation of PTEN in cancer predisposition.
with juvenile polyposis of infancy, reflecting cooperation N Engl J Med 2020;382:2103–2116.
between the BMPR1A and PTEN tumor-suppressor 100. Tan MH, Mester JL, Ngeow J, et al. Lifetime cancer risks
genes. Am J Hum Genet 2006;78:1066–1074. in individuals with germline PTEN mutations. Clin Cancer
83. Menko FH, Kneepkens CM, de Leeuw N, et al. Variable Res 2012;18:400–407.
phenotypes associated with 10q23 microdeletions 101. Bubien V, Bonnet F, Brouste V, et al. High cumulative
involving the PTEN and BMPR1A genes. Clin Genet risks of cancer in patients with PTEN hamartoma tumour
2008;74:145–154. syndrome. J Med Genet 2013;50:255–263.
84. Fox VL, Perros S, Jiang H, et al. Juvenile polyps: 102. Riegert-Johnson DL, Gleeson FC, Roberts M, et al. Can-
recurrence in patients with multiple and solitary polyps. cer and Lhermitte-Duclos disease are common in Cowden
Clin Gastroenterol Hepatol 2010;8:795–799. syndrome patients. Heredit Cancer Clin Pract 2010;8(1):6.
85. Wu TT, Rezai B, Rashid A, et al. Genetic alterations and 103. Heald B, Mester J, Rybicki L, et al. Frequent gastroin-
epithelial dysplasia in juvenile polyposis syndrome and testinal polyps and colorectal adenocarcinomas in a
sporadic juvenile polyps. Am J Pathol 1997;150:939–947. prospective series of PTEN mutation carriers. Gastro-
86. Brosens LA, van Hattem A, Hylind LM, et al. Risk of enterology 2010;139:1927–1933.
colorectal cancer in juvenile polyposis. Gut 2007; 104. Nieuwenhuis MH, Kets CM, Murphy-Ryan M, et al.
56:965–967. Cancer risk and genotype-phenotype correlations in
87. Gonzalez RS, Adsay V, Graham RP, et al. Massive PTEN hamartoma tumor syndrome. Fam Cancer 2014;
gastric juvenile-type polyposis: a clinicopathological 13:57–63.
analysis of 22 cases. Histopathology 2017;70:918–928. 105. Nieuwenhuis MH, Kets CM, Murphy-Ryan M, et al. Is
88. Friedl W, Uhlhaas S, Schulmann K, et al. Juvenile pol- colorectal surveillance indicated in patients with PTEN
yposis: massive gastric polyposis is more common in mutations? Colorectal Dis 2012;14:e562–e566.
MADH4 mutation carriers than in BMPR1A mutation 106. Stanich PP, Owens VL, Sweetser S, et al. Colonic pol-
carriers. Hum Genet 2002;111:108–111. yposis and neoplasia in Cowden syndrome. Mayo Clin
89. Howe JR, Mitros FA, Summers RW. The risk of gastro- Proc 2011;86:489–492.
intestinal carcinoma in familial juvenile polyposis. Ann 107. Arch EM, Goodman BK, Van Wesep RA, et al. Deletion of
Surg Oncol 1998;5:751–756. PTEN in a patient with Bannayan-Riley-Ruvalcaba syn-
90. Wirtzfeld DA, Petrelli NJ, Rodriguez-Bigas MA. Hamar- drome suggests allelism with Cowden disease. Am J
tomatous polyposis syndromes: molecular genetics, Med Genet 1997;71:489–493.
neoplastic risk, and surveillance recommendations. Ann 108. Hendriks YM, Verhallen JT, van der Smagt JJ, et al.
Surg Oncol 2001;8:319–327. Bannayan-Riley-Ruvalcaba syndrome: further delinea-
91. MacFarland SP, Ebrahimzadeh JE, Zelley K, et al. tion of the phenotype and management of PTEN
Phenotypic differences in juvenile polyposis syndrome mutation-positive cases. Fam Cancer 2003;2:79–85.
with or without a disease-causing SMAD4/BMPR1A 109. Carethers JM, Furnari FB, Zigman AF, et al. Absence of
CLINICAL PRACTICE GUIDELINES

variant. Cancer Prev Res (Phila) 2021;14:215–222. PTEN/MMAC1 germ-line mutations in sporadic
92. Cohen S. Management of JPS in children and adolescents: Bannayan-Riley-Ruvalcaba syndrome. Cancer Res
a positon paper from the ESPGHAN Polyposis Working 1998;58:2724–2726.
Group. J Pediatr Gastroenterol Nutr 2019;68:453–462. 110. Alimi A, Weeth-Feinstein LA, Stettner A, et al. Overlap of
93. Faughnan ME, Mager JJ, Hetts SW, et al. Second in- juvenile polyposis syndrome and Cowden syndrome due
ternational guidelines for the diagnosis and management to de novo chromosome 10 deletion involving BMPR1A
of hereditary hemorrhagic telangiectasia. Ann Intern Med and PTEN: implications for treatment and surveillance.
2020;173:989–1001. Am J. Med Genet A 2015;167:1305–1308.
94. Campos FG, Figueiredo MN, Martinez CA. Colorectal 111. Hiljadnikova Bajro M, Sukarova-Angelovska E,
cancer risk in hamartomatous polyposis syndromes. Adelaide J, et al. A new case with 10q23 interstitial
World J Gastrointest Surg 2015;7:25–32. deletion encompassing both PTEN and BMPR1A nar-
95. Pilarski R, Burt R, Kohlman W, et al. Cowden syndrome rows the genetic region deleted in juvenile polyposis
and the PTEN hamartoma tumor syndrome: systematic syndrome. J Appl Genet 2013;54:43–47.
June 2022 Gastrointestinal Hamartomatous Polyposis Syndromes 2085

112. Shaco-Levy R, Jasperson KW, Martin K, et al. Gastro- 120. Laitman Y, Jaeger E, Katz L, et al. GREM1 germline
intestinal polyposis in cowden syndrome. J Clin Gas- mutation screening in Ashkenazi Jewish patients with
troenterol 2017;51:e60–e67. familial colorectal cancer. Genet Res 2015;97:e11.
113. Borowsky J, Setia N, Rosty C, et al. Spectrum of 121. Lewis A, Freeman-Mills L, de la Calle-Mustienes E, et al.
gastrointestinal tract pathology in a multicenter cohort of A polymorphic enhancer near GREM1 influences bowel
43 Cowden syndrome patients. Mod Pathol 2019; cancer risk through differential CDX2 and TCF7L2
32:1814–1822. binding. Cell Rep 2014;8:983–990.
114. Ni Y, He X, Chen J, et al. Germline SDHx variants modify 122. Lieberman S, Walsh T, Schechter M, et al. Features of
breast and thyroid cancer risks in Cowden and Cowden- patients with hereditary mixed polyposis syndrome
like syndrome via FAD/NAD-dependant destabilization caused by duplication of GREM1 and implications for
of p53. Hum Mol Genet 2012;21:300–310. screening and surveillance. Gastroenterology 2017;
115. Kato M, Mizuki A, Hayashi T, et al. Cowden’s disease 152:1876–1880.e1.
diagnosed through mucocutaneous lesions and gastro- 123. Shackelford DB, Shaw RJ. The LKB1-AMPK pathway:
intestinal polyposis with recurrent hematochezia, unre- metabolism and growth control in tumour suppression.
vealed by initial diagnosis. Intern Med 2000;39:559–563. Nat Rev Cancer 2009;9:563–575.
116. Stanich PP, Pilarski R, Rock J, et al. Colonic manifes-
tations of PTEN hamartoma tumor syndrome: case se-
Correspondence
ries and systematic review. World J Gastroenterol 2014; Address correspondence to: C. Richard Boland, MD, University of California
20:1833–1838. San Diego, 9444 Medical Center Drive, East Campus Office Building, Room
2-065, La Jolla, California 92037. e-mail: crboland@health.ucsd.edu
117. Plesec T, Brown K, Allen C, et al. Clinicopathological
features of a kindred with SCG5-GREM1-associated Conflicts of interest
hereditary mixed polyposis syndrome. Hum Pathol The authors disclose no conflicts of interest relative to the current work since
2016. These authors disclose the following industry relationships (consulting,
2017;60:75–81. research, reimbursement) without conflict of interest relevant to the current
118. Whitelaw SC, Murday VA, Tomlinson IP, et al. Clinical work since 2016: C. Richard Boland: Ambry Genetics. Gregory Idos: Myriad
Genetics, Inc. Carol Burke: Salix Pharmaceuticals, Ferring Pharmaceuticals,
and molecular features of the hereditary mixed polyposis Aries Pharmaceuticals, Pfizer, Cancer Prevention Pharmaceuticals, Janssen
syndrome. Gastroenterology 1997;112:327–334. Pharmaceuticals, SLA Pharma AG, and Freenome Holdings, Inc. Samir
Gupta: Freenome Holdings, Inc, Guardant Health, Inc, CellMax, Inc, and
119. Jaeger E, Leedham S, Lewis A, et al. Hereditary mixed Mallinckrodt Pharmaceuticals. Brian C. Jacobson: Motus, GI, Dark Canyon
polyposis syndrome is caused by a 40-kb upstream LLC, and Remedy Partners. Aasma Shaukat: Iterative Scopes, Freenome
duplication that leads to increased and ectopic expres- Holdings Inc. Swati G. Patel: Olympus America, Freenome Holdings Inc,
ERBE USA. Sapna Syngal: Myriad Genetics, Inc, DC Health, Inc, and
sion of the BMP antagonist GREM1. Nat Genet 2012; GlaxoSmith Kline, Inc. Douglas Robertson: Covidien, Freenome Holdings,
44:699–703. Inc, and Amadix. The remaining authors disclose no conflicts.

CLINICAL PRACTICE GUIDELINES


CASE REPORT – OPEN ACCESS
International Journal of Surgery Case Reports 79 (2021) 286–290

Contents lists available at ScienceDirect

International Journal of Surgery Case Reports


journal homepage: www.casereports.com

Peutz-Jeghers syndrome in a woman presenting as intussusception: A


case report
Erwin Syarifuddin a , Rina Masadah b , Ronald Erasio Lusikooy a , Warsinggih a,∗ ,
Julianus Aboyaman Uwuratuw a , Muhammad Faruk c
a
Division of Digestive, Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
b
Department of Pathology Anatomy, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
c
Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia

a r t i c l e i n f o a b s t r a c t

Article history: INTRODUCTION: Peutz-Jeghers syndrome (PJS) is an uncommon autosomal dominant syndrome with a
Received 16 December 2020 variable to high penetrance that leads to the development of polyps within the gastrointestinal mucosa.
Received in revised form 14 January 2021 Here we report a case of an adult female suffering jejunoileal intussusception due to PJS.
Accepted 14 January 2021
PRESENTATION OF CASE: A 30-year-old woman came to an emergency department with a small bowel
Available online 18 January 2021
obstruction caused by intussusception. The patient underwent an emergency exploratory laparotomy. An
intussusception at the level of 60 cm from the ligamentum treitz was revealed, and the intussusception
Keywords:
small bowel segment was not viable; we decided to perform segmental jejunoileal resection with the
Peutz-Jeghers syndrome
Intussusception
Bishop-Koop procedure, and the specimen histopathology of the segmental jejunoileal resection showed
Hamartoma a typical hamartomatous polyp features. Two month later, diagnostic endoscopy showed multiple polyps
Polyps (between 5 and 15 mm) in the large bowel. The polyps were removed with endoscopic polypectomy
Case report and examined histopathologically, showing characteristics of PJS. Further detailed family history was
obtained, and similar skin lesions were detected on our patient’s child (since birth). Although endoscopy
screening identified multiple polyps in the child’s ileum and large bowel, he was not suffering from
abdominal symptoms.
CONCLUSION: In patients with intussusception at a young age, PJS can be caused by the presence of a
hamartoma polyp as a trigger for intussusception. If there are multiple polyps found in the gastrointestinal
mucosa and other pathognomonic signs are found, such as hyperdense macular lesions on the lip and
buccal mucosa, such cases should be confirmed as PJS.
© 2021 The Author(s). Published by Elsevier Ltd on behalf of IJS Publishing Group Ltd. This is an open
access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction (GI) tract develops hamartomatous polyps, which occur most com-
monly in the small bowel, colon, and rectum (≥ 90% of cases), and
Peutz-Jeghers syndrome (PJS) is an uncommon autosomal dom- less commonly in the stomach or urinary tract [7]. Hamartomas can
inant syndrome with a variable to high penetrance that leads to range from 5 to 50 mm in diameter (median size, 35 mm). Hamar-
the development of polyps within the colon and rectum, and only tomas are associated with intussusception, bleeding, anemia, and
a small number of cases have been reported in the literature [1]. obstruction [8].
PJS affects around 1 in 8300 to 200,000 births [2]. PJS was first Here, we report the uncommon case of a female suffering
reported by Peutz in 1921 [3,4], and subsequently, detailed case PJS jejunoileal intussusception due to PJS, in line with the updated
by Jeghers, McKusick, and Katz in 1949 [5]. This syndrome consists consensus-based surgical case report (SCARE) guidelines [9].
of the association of gastrointestinal polyps mucocutaneous pig-
mentation and a familial incidence [6]. In PJS, the gastrointestinal
2. Case presentation

A 30-year-old woman came to an emergency department with


∗ Corresponding author at: Warsinggih Division of Digestive, Department of the chief complaint of intermittent episodes of colicky abdomi-
Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Jalan Perintis nal pain with tarry stool 4 weeks before admission. From the vital
Kemerdekaan KM 11, Makassar, 90245, Indonesia. signs, the patient was tachycardic and hypotensive. On physical
E-mail addresses: erwinsyarifuddin@yahoo.com (E. Syarifuddin),
r.masadah@med.unhas.ac.id (R. Masadah), ronaldlusikooy@gmail.com
examination, she had an of pigmentation in the oral cavity (Fig. 1).
(R.E. Lusikooy), kbd.warsinggih@gmail.com ( Warsinggih), The abdomen was less distended, with increased bowel sound,
boyuwuratuw@gmail.com (J.A. Uwuratuw), faroex8283@gmail.com (M. Faruk). tenderness, and hypertimpanic on percussion. The digital rectal

https://doi.org/10.1016/j.ijscr.2021.01.053
2210-2612/© 2021 The Author(s). Published by Elsevier Ltd on behalf of IJS Publishing Group Ltd. This is an open access article under the CC BY-NC-ND license (http://
creativecommons.org/licenses/by-nc-nd/4.0/).
CASE REPORT – OPEN ACCESS
E. Syarifuddin et al. International Journal of Surgery Case Reports 79 (2021) 286–290

with mucocutaneous pigmentation, especially of the vermilion bor-


der of the lips. PJS is an autosomal dominant disease caused by a
germline mutation in the STK11 (LKB1) gene [8]. The estimation of
the population prevalence of PJS differs between studies, ranging
from 1 in 8300 to 1 in 280,000 individuals. The probable prevalence
is around 1 in 100,000 people [2].
The diagnosis of PJS can be made in patients with hamartoma-
tous polyp(s) with at least two of the following clinical criteria
also present: labial melanin deposits, a family history of the syn-
drome, and small bowel polyposis. The syndrome appears equally
in males and females and is found in all racial groups [6]. The clin-
ical manifestation of PJS is characterized by asymptomatic periods
interspersed with complications, such as abdominal pain, intus-
susception often leading to intestinal obstruction, polyp extrusion
through the rectum, and bleeding, which is often occult [7,10,11].
Small bowel obstruction is the presenting complaint in half of
the cases, and relaparotomy due to polyp-induced complications
occurs commonly and might do so at quite short intervals [10].
In addition to polyposis, previous studies have reported an
increased risk of GI and extra-GI malignancies in PJS patients, com-
pared with that of the general population [7]. The PJS is associated
with an increased risk of gastrointestinal and non-gastrointestinal
malignancies. A meta-analysis involving six studies and 210
patients showed a cumulative risk of 93% from 15 to 64 years for all
types of malignancies. Thus, the relative risk of an individual with
SPJ to present neoplasia in any region, compared with the general
population, is up to 15 times higher [11,12]. The most frequent
neoplasm in patients with PJS is the colonic tumor (57%), followed
by breast (45%), pancreas (36%), stomach (29%), ovary (21%), small
Fig. 1. Mucocutaneous pigmentation on the buccal mucosa and lips.
intestine (13%), and uterus (10%) tumors [11,13]. Among gastroin-
testinal cancers, increased cancer risk was indicated for the colon,
examination revealed normal limits. Blood investigation showed stomach, small intestine, and pancreas. Also, female PJS patients
anemia (hemoglobin level, 7.6 g/dl). An abdominal X-ray showed a are at greater risk of gastrointestinal and gynecological cancers,
small bowel obstruction. ovarian cancer, cervical cancer, uterine cancer, and breast cancer.
The patient underwent an emergency exploratory laparotomy. Meanwhile, the high risk of pulmonary cancer, renal cancer, pro-
We (the digestive surgeon and residents) detected a jejunoileal static cancer, bone cancer, and leukemia has also been reported.
intussusception caused by multiple polyps at ∼60 cm from the lig- There is wide variability in cancer risk estimates, as reviewed in a
amentum treitz (Fig. 2A and B), and the milking procedure was recent meta-analysis study [14].
successfully performed, but a 40-cm segment of the jejunoileal Until recently, PJS was considered a benign condition and was
intussusception was not viable. Therefore, we decided to perform treated conservatively. Repeated laparotomies were considered
segmental resection and, through this, found multiple polyps in risky and ineffective because of the continuous growth of the
the borderline resection of the mucosa proximal jejunum and dis- polyps. Patients were, therefore, treated surgically only when acute
tal ileum. We decided to extend the resection until we no longer bowel obstruction occurred [10].
found polyps. We decided to perform the Bishop-Koop procedure More recently, the management approach has changed, becom-
(Fig. 3A). The specimen was sent for histopathological examination ing more aggressive. When the diagnosis of PJS is histologically
and described as a typical hamartomatous polyp (Fig. 3B). She was confirmed, we suggest that all polyps should be removed with the
given an intravenous broad-spectrum antibiotic, and analgesics aim of preventing intussusception with gangrene requiring resec-
drugs were administered for 5 days. tion, which might lead to short-bowel syndrome [10].
Postoperatively, the patient had satisfactory progress, tolerat- Most publications recommend polypectomy for polyps in the
ing oral feeding, and was discharged on the seventh postoperative stomach or colon that are greater than 1 cm in size noted dur-
day. Two month later, we performed diagnostic endoscopy; there ing endoscopic surveillance. Surgery has been recommended for
were multiple polyps in the large bowel measuring 5–15 mm, symptomatic or rapidly growing small intestinal polyps or asymp-
without any features of intussusceptions (Fig. 4). This polyp was tomatic polyps greater than 1–1.5 cm in size. Some experts suggest
removed with endoscopic polypectomy. Histopathological exam- a clean sweep. This can be facilitated by concomitant interopera-
ination revealed a polyp characteristic of PJS consisting of a tive endoscopy with polypectomy or, in the case of larger polyps,
branching framework of connective tissue and smooth muscle lined enterotomy. The clean sweep approach appears to decrease the
by the normal intestinal epithelium. Further detailed family history need for recurrent small bowel surgery. Recently, the use of double-
was obtained, and similar skin lesions were detected on her child balloon enteroscopy for removal of small bowel PJS polyps has been
(since birth). Although endoscopy screening performed identified reported and might decrease the need for laparotomy [14].
multiple polyps (< 5 mm) in the ileum and large bowel, the child In our case, the patient was admitted to the hospital with intus-
was not suffering from abdominal symptoms (Fig. 5). susception. The exploratory laparotomy revealed intussusception
with the small bowel segment not viable. Small bowel resection
3. Discussion was performed and continued with the Bishop-Koop procedure
due to prevent short bowel syndrome. We did not perform an end-
PJS is an inherited polyposis syndrome in which multiple char- to-end anastomosis because the patient had hypovolemic shock
acteristic polyps occur in the gastrointestinal (GI) tract, associated

287
CASE REPORT – OPEN ACCESS
E. Syarifuddin et al. International Journal of Surgery Case Reports 79 (2021) 286–290

Fig. 2. (A) Intussusception ileojejenal and (B) hamartoma of the ileum.

Fig. 3. (A) The Bishop-Koop procedure of the jejunum; (B) The histopathological findings reveal hamartoma, with a typical PJS polyp demonstrating an arborizing pattern of
growth of smooth-muscle (HE staining, magnification 4×); and (C) Histopathological findings revealed an edematous appearance of the intestinal mucosa with inflammatory
lymphocytes, histiocytes, and very dense plasma cells. In the layer below, there appears pigment hemosiderin. There is no evidence of dysplasia or malignancy (HE staining,
magnification 40×).

Fig. 4. Colonoscopy with more than 50 polyps scattered throughout the colon and rectum.

288
CASE REPORT – OPEN ACCESS
E. Syarifuddin et al. International Journal of Surgery Case Reports 79 (2021) 286–290

Fig. 5. On the patient’s male child, we detected: (A) pigmentations of the lips and oral mucosa; and (B) polyp in the large bowel detected by endoscopy screening.

preoperative and anemia, and we found edema in the small bowel Guarantor
mucosa.
Erwin Syarifuddin.
4. Conclusion
Provenance and peer review
In patients with intussusception at a young age, a hamartoma
polyp might have been the trigger for intussusception. If there are Not commissioned, externally peer-reviewed.
multiple polyps found in the GI mucosa together with other pathog-
nomonic signs, such as hyperdense macular lesions on the lip and Acknowledgment
buccal mucosa, such cases should be confirmed as PJS.
We acknowledge Robert Christeven, M.D, for her help in provid-
Declaration of Competing Interest ing us with the linguistic assistance for this case report.

The authors declare that they have no conflict of interests. References

[1] S.-X. Duan, G.-H. Wang, J. Zhong, W.-H. Ou, M.-X. Fu, F.-S. Wang, S.-H. Ma, J.-H.
Funding Li, Peutz–Jeghers syndrome with intermittent upper intestinal obstruction,
Medicine (Baltimore) 96 (2017) e6538, http://dx.doi.org/10.1097/MD.
0000000000006538.
No funding or sponsorship was received for this study or publi- [2] M. Kopacova, I. Tacheci, S. Rejchrt, J. Bures, Peutz-Jeghers syndrome:
cation of this article. diagnostic and therapeuticapproach, World J. Gastroenterol. 15 (2009) 5397,
http://dx.doi.org/10.3748/wjg.15.5397.
[3] J.H. Tweedie, B.G. McCann, Peutz-Jeghers syndrome and metastasising colonic
Ethical Approval adenocarcinoma, Gut 25 (1984) 1118–1123, http://dx.doi.org/10.1136/gut.25.
10.1118.
[4] S. Chowdhry, D.D. Umrigar, N. Yadav, Peutz-jeghers syndrome in a child
The study is exempt from ethical approval in our institution. presenting with acute abdomen: a case report, Asian J. Dermatol. 8 (2015)
20–24, http://dx.doi.org/10.3923/ajd.2016.20.24.
[5] T.R. Foley, T.J. McGarrity, A.B. Abt, Peutz-Jeghers syndrome: a
Consent
clinicopathologic survey of the “Harrisburg family” with a 49-year follow-up,
Gastroenterology 95 (1988) 1535–1540, http://dx.doi.org/10.1016/s0016-
Written informed consent was obtained from the patient for 5085(88)80074-x.
publication of this case report and accompanying images. A copy [6] F.M. Giardiello, J.D. Trimbath, Peutz-Jeghers syndrome and management
recommendations, Clin. Gastroenterol. Hepatol. 4 (2006) 408–415, http://dx.
of the written consent is available for review by the Editor-in-Chief doi.org/10.1016/j.cgh.2005.11.005.
of this journal on request. [7] N. Butt, M. Salih, M. Khan, R. Ahmed, Z. Haider, S.A. Shah, An incidentally
discovered asymptomatic para-aortic paraganglioma with Peutz-Jeghers
syndrome, Saudi J. Gastroenterol. 18 (2012) 388, http://dx.doi.org/10.4103/
Author contribution 1319-3767.103432.
[8] A. Latchford, S. Cohen, M. Auth, M. Scaillon, J. Viala, R. Daniels, C. Talbotec, T.
Attard, C. Durno, W. Hyer, Management of Peutz-Jeghers syndrome in
Erwin Syarifuddin, Julianus A. Uwuratuw, and Muhammad children and adolescents, J. Pediatr. Gastroenterol. Nutr. 68 (2019) 442–452,
Faruk: study concept, surgical therapy for this patient. Erwin http://dx.doi.org/10.1097/MPG.0000000000002248.
Syarifuddin, Rina Masadah, and Muhammad Faruk: Data collec- [9] R.A. Agha, T. Franchi, C. Sohrabi, G. Mathew, A. Kerwan, A. Thoma, A.J.
Beamish, A. Noureldin, A. Rao, B. Vasudevan, B. Challacombe, B. Perakath, B.
tion, Writing-Original draft preparation. Ronald E. Lusikooy and
Kirshtein, B. Ekser, C.S. Pramesh, D.M. Laskin, D. Machado-Aranda, D. Miguel,
Warsinggih: senior author and the manuscript reviewer. Erwin D. Pagano, F.H. Millham, G. Roy, H. Kadioglu, I.J. Nixon, I. Mukhejree, J.A.
Syarifuddin and Muhammad Faruk: Editing and Writing. All McCaul, J. Chi-Yong Ngu, J. Albrecht, J.G. Rivas, K. Raveendran, L. Derbyshire,
M.H. Ather, M.A. Thorat, M. Valmasoni, M. Bashashati, M. Chalkoo, N.Z. Teo, N.
authors read and approved the final manuscript.
Raison, O.J. Muensterer, P.J. Bradley, P. Goel, P.S. Pai, R.Y. Afifi, R.D. Rosin, R.
Coppola, R. Klappenbach, R. Wynn, R.L. De Wilde, S. Surani, S. Giordano, S.
Registration of research studies Massarut, S.G. Raja, S. Basu, S.A. Enam, T.G. Manning, T. Cross, V.K. Karanth, V.
Kasivisvanathan, Z. Mei, The SCARE 2020 guideline: updating consensus
surgical CAse REport (SCARE) guidelines, Int. J. Surg. 84 (2020) 226–230,
Not applicable. http://dx.doi.org/10.1016/j.ijsu.2020.10.034.

289
CASE REPORT – OPEN ACCESS
E. Syarifuddin et al. International Journal of Surgery Case Reports 79 (2021) 286–290

[10] I. Rebsdorf Pedersen, A. Hartvigsen, B. Fischer Hansen, C. Toftgaard, K. [13] M. Jansen, L.A.A. Brosens, G.J.A. Offerhaus, Gastrointestinal polyposis
Konstantin-Hansen, S. Büllow, Management of Peutz-Jeghers syndrome. syndromes: early tumor evolution through the looking glass, in: Pathobiol.
Experience with patients from the Danish Polyposis Register, Int. J. Colorectal Hum. Dis., Elsevier, 2014, pp. 1319–1331, http://dx.doi.org/10.1016/B978-0-
Dis. 9 (1994) 177–179, http://dx.doi.org/10.1007/BF00292244. 12-386456-7.03809-0.
[11] J. Loureiro, G.L. Menegazzo, L. Vergamini, R.C. Pestana, F.B. Formiga, M.G.C. de [14] H.-Y. Chen, X.-W. Jin, B.-R. Li, M. Zhu, J. Li, G.-P. Mao, Y.-F. Zhang, S.-B. Ning,
Sousa, T.Y. Takahashi, F. Silveira, P. de A.P. Candelária, D.M. Filho, F.C. Bin, Cancer risk in patients with Peutz-Jeghers syndrome: a retrospective cohort
Diagnostic difficulty in Peutz–Jeghers syndrome, Colorectal Dis. 35 (2015) study of 336 cases, Tumour Biol. 39 (2017), http://dx.doi.org/10.1177/
67–71, http://dx.doi.org/10.1016/j.jcol.2014.08.012. 1010428317705131, 1010428317705131.
[12] F.M. Giardiello, Hereditary colorectal cancer and polyp syndromes, in: Early
Diagnosis Treat. Cancer Ser. Color. Cancer, Elsevier, 2011, pp. 21–30, http://dx.
doi.org/10.1016/B978-1-4160-4686-8.50008-7.

Open Access
This article is published Open Access at sciencedirect.com. It is distributed under the IJSCR Supplemental terms and conditions, which
permits unrestricted non commercial use, distribution, and reproduction in any medium, provided the original authors and source are
credited.

290

You might also like