U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Adam MP, Feldman J, Mirzaa GM, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2024.

Cover of GeneReviews®

GeneReviews® [Internet].

Show details

Peutz-Jeghers Syndrome

Synonym: PJS

, MD, , PhD, and , PhD.

Author Information and Affiliations

Initial Posting: ; Last Update: September 2, 2021.

Estimated reading time: 36 minutes

Summary

Clinical characteristics.

Peutz-Jeghers syndrome (PJS) is characterized by the association of gastrointestinal (GI) polyposis, mucocutaneous pigmentation, and cancer predisposition. PJS-type hamartomatous polyps are most common in the small intestine (in order of prevalence: jejunum, ileum, and duodenum) but can also occur in the stomach, large bowel, and extraintestinal sites including the renal pelvis, bronchus, gall bladder, nasal passages, urinary bladder, and ureters. GI polyps can result in chronic bleeding, anemia, and recurrent obstruction and intussusception requiring repeated laparotomy and bowel resection. Mucocutaneous hyperpigmentation presents in childhood as dark blue to dark brown macules around the mouth, eyes, and nostrils, in the perianal area, and on the buccal mucosa. Hyperpigmented macules on the fingers are common. The macules may fade in puberty and adulthood. Recognition of the distinctive skin manifestations is important especially in individuals who have PJS as the result of a de novo pathogenic variant as these skin findings often predate GI signs and symptoms. Individuals with PJS are at increased risk for a wide variety of epithelial malignancies (colorectal, gastric, pancreatic, breast, and ovarian cancers). Females are at risk for sex cord tumors with annular tubules (SCTAT), a benign neoplasm of the ovaries, and adenoma malignum of the cervix, a rare aggressive cancer. Males occasionally develop large calcifying Sertoli cell tumors of the testes, which secrete estrogen and can lead to gynecomastia, advanced skeletal age, and ultimately short stature, if untreated.

Diagnosis/testing.

The diagnosis of PJS is based on clinical findings. Identification of a heterozygous pathogenic variant in STK11 by molecular genetic testing confirms the diagnosis and allows for testing of at-risk relatives.

Management.

Treatment of manifestations: Routine endoscopic surveillance with polypectomy decreases the frequency of emergency laparotomy and bowel loss resulting from intussusception. Small-bowel imaging includes video capsule endoscopy (VCE), CT enterography, and/or magnetic resonance enterography (MRE). Balloon-assisted enteroscopy allows for removal of deep small-bowel polyps. Occasionally intraoperative enteroscopy and enterotomy is needed for removal of large distal small-bowel polyps. Intussusception and malignancies should be treated in the standard manner.

Prevention of primary manifestations: Although not specifically studied in individuals with PJS, prophylactic mastectomy to decrease the risk of breast cancer could be considered based on family history or other clinical factors. Similarly, there is no prospective data on gynecologic prophylactic surgery for the elevated gynecologic cancer risk in females with PJS.

Surveillance: In children and adolescents: colonoscopy and upper endoscopy at age eight years; if negative, follow up at age 18 years. If polyps are detected repeat every one to three years based on size, number, and histopathology of polyps. Small-bowel surveillance by MRE or VCE every one to three years beginning at age eight years. Examination for precocious puberty in females annually beginning at age eight years. Testicular examination and examination for feminizing changes in males annually beginning at age ten years.

In adults: Colonoscopy, upper endoscopy, and small-bowel examination by MRE or VCE every two to three years beginning at age 18 years; clinical breast examination in women every six months beginning at age 30 years; mammogram and breast MRI in women annually beginning at age 30 years; pelvic examination and pap smear in women annually beginning at age 18 to 20 years. Pancreatic imaging with endoscopic ultrasound or MRI/MRCP annually beginning at age 30 to 35 years.

Evaluation of relatives at risk: If the pathogenic variant in the family is known, offer molecular genetic testing to at-risk relatives so that morbidity and mortality can be reduced by early diagnosis and prevention of disease through appropriate surveillance and consideration of prophylactic measures in affected family members. If the family variant is not known, offer clinical diagnostic evaluations to all at-risk family members, who will benefit from early treatment and appropriate surveillance.

Genetic counseling.

PJS is inherited in an autosomal dominant manner. The majority of individuals diagnosed with PJS have an affected parent; however, many individuals with PJS represent apparently simplex cases. The exact proportion of individuals who have PJS as the result of a de novo pathogenic variant is unknown. If a parent of the proband is affected and/or is known to have the STK11 pathogenic variant identified in the proband, the risk to the sibs of inheriting the pathogenic variant is 50%. Once the STK11 pathogenic variant has been identified in an affected family member, predictive testing for at-risk family members, prenatal testing, and preimplantation genetic testing are possible.

Diagnosis

Suggestive Findings

Peutz-Jeghers syndrome (PJS) should be suspected in individuals with the following:

  • Two or more PJS-type hamartomatous polyps of the gastrointestinal (GI) tract.
  • Characteristic mucocutaneous pigmentation and hyperpigmented macules (periorbital, lips, fingers, nose, toes, and anus)
  • Gynecomastia in males as a result of estrogen-producing Sertoli cell testicular tumors
  • History of intussusception, especially in a child or young adult

PJS-type GI polyps. The sine qua non of PJS diagnosis is the hamartomatous GI polyp, which is histopathologically characterized by distinctive interdigitating smooth muscle bundles in a characteristic arborizing (branching tree) appearance throughout the lamina propria. Small-bowel polyps are typically pedunculated and demonstrate a lobular architecture. The overlying epithelium is typically nondysplastic, although dysplasia can occur. In the stomach, PJS polyps may be indistinguishable from juvenile or hyperplastic polyps. In the large intestine, PJS polyps resemble mucosal prolapse polyps. Their lobular architecture and desmin-positive smooth muscle fiber favor the diagnosis of PJS [Rosty 2018]. Pseudo invasion of misplaced crypts is an innate property of the PJS hamartoma, which may reflect the role of STK11 in cell polarity [Tse et al 2013].

Note: Individuals with PJS also develop many other polyps; polyps showing adenomatous changes frequently arise in the colon and may cause confusion with familial adenomatous polyposis. The histology of gastric PJS polyps can be similar to gastric hyperplastic polyps, thus highlighting the importance of a GI pathologist in the review of polyp histology and knowledge of the clinical context [Rosty 2018].

Establishing the Diagnosis

The clinical diagnosis of PJS is established in a proband with one of the following [Beggs et al 2010]:

  • Two or more histologically confirmed PJS-type hamartomatous polyps
  • Any number of PJS-type polyps detected in an individual who has a family history of PJS in at least one close relative
  • Characteristic mucocutaneous pigmentation in an individual who has a family history of PJS in at least one close relative
  • Any number of PJS-type polyps in an individual who also has characteristic mucocutaneous pigmentation

The molecular diagnosis of PJS is established in a proband with suggestive findings and a heterozygous pathogenic variant in STK11 identified by molecular genetic testing (see Table 1).

Note: Identification of a heterozygous STK11 variant of uncertain significance does not establish or rule out the diagnosis.

Molecular genetic testing approaches can include single-gene testing, multigene panel, and comprehensive genomic testing depending on the phenotype.

Single-gene testing. Sequence analysis of STK11 is performed first to detect small intragenic deletions/insertions and missense, nonsense, and splice site variants. Note: Depending on the sequencing method used, single-exon, multiexon, or whole-gene deletions/duplications may not be detected. If no variant is detected by the sequencing method used, the next step is to perform gene-targeted deletion/duplication analysis to detect exon and whole-gene deletions or duplications. Sequence analysis and gene-targeted deletion/duplication analysis can be done simultaneously to decrease turn-around time.

Note: If no STK11 pathogenic variant is identified, testing of an alternate DNA source (e.g., buccal cells) for somatic mosaicism should be considered [Butel-Simoes et al 2019]. To date, seven individuals with somatic mosaicism have been identified [Jelsig et al 2021].

A multigene panel that includes STK11 and other genes of interest (see Differential Diagnosis) may also be considered. Note: (1) The genes included and the sensitivity of multigene panels vary by laboratory and are likely to change over time. (2) Some multigene panels may include genes not associated with the condition discussed in this GeneReview; thus, clinicians need to determine which multigene panel is most likely to identify the genetic cause of the condition while limiting identification of variants of uncertain significance and pathogenic variants in genes that do not explain the underlying phenotype. (3) In some laboratories, panel options may include a custom laboratory-designed panel and/or custom phenotype-focused exome analysis that includes genes specified by the clinician. (4) Methods used in a panel may include sequence analysis, deletion/duplication analysis, and/or other non-sequencing-based tests.

For an introduction to multigene panels click here. More detailed information for clinicians ordering genetic tests can be found here.

More comprehensive genomic testing (when available) including exome sequencing and genome sequencing may be considered if single-gene testing (and/or use of a multigene panel that includes STK11) fails to confirm a diagnosis in an individual with features of PJS. Such testing may provide or suggest a diagnosis not previously considered (e.g., mutation of a different gene or genes that results in a similar clinical presentation).

For an introduction to comprehensive genomic testing click here. More detailed information for clinicians ordering genomic testing can be found here.

Table 1.

Molecular Genetic Testing Used in Peutz-Jeghers Syndrome

Gene 1MethodProportion of Probands with a Pathogenic Variant 2 Detectable by Method
STK11 Sequence analysis 3~80%-85% 4, 5
Gene-targeted deletion/duplication analysis 6~15%-20% 4, 5, 7
UnknownNA<1% 4, 8
1.
2.

See Molecular Genetics for information on variants detected in this gene.

3.

Sequence analysis detects variants that are benign, likely benign, of uncertain significance, likely pathogenic, or pathogenic. Variants may include small intragenic deletions/insertions and missense, nonsense, and splice site variants; typically, exon or whole-gene deletions/duplications are not detected. For issues to consider in interpretation of sequence analysis results, click here.

4.
5.

In individuals without a pathogenic identified, sequence analysis and deletion/duplication testing of an alternate DNA source (e.g., buccal cells) for somatic mosaicism should be considered. To date, seven individuals have been identified with somatic mosaic STK11 pathogenic variants.

6.

Gene-targeted deletion/duplication analysis detects intragenic deletions or duplications. Methods used may include quantitative PCR, long-range PCR, multiplex ligation-dependent probe amplification (MLPA), and a gene-targeted microarray designed to detect single-exon deletions or duplications.

7.

Includes larger deletions, such as whole-gene deletions of STK11 and smaller intragenic deletions [De Rosa et al 2010, Borun et al 2015].

8.

Of 25 individuals who had PJS but did not have a detectable STK11 pathogenic variant, one had a heterozygous pathogenic variant of the DNA repair enzyme MUTYH that was not observed in 1015 controls [Alhopuro et al 2008]. Of note, pathogenic variants in MUTYH ordinarily cause an autosomal recessive form of adenomatous polyposis coli.

Clinical Characteristics

Clinical Description

Peutz-Jeghers syndrome (PJS) is characterized by the association of gastrointestinal (GI) polyposis and mucocutaneous pigmentation. The risk for GI and extraintestinal malignancies is significantly increased. Distinct benign and malignant gonadal and gynecologic tumors can also be seen. Variable expressivity is common; for example, some affected individuals in families with PJS may have only polyps or perioral pigmentation.

GI polyposis. PJS-type hamartomatous polyps can occur anywhere in the GI tract but occur most commonly in the small intestine. The density of polyps is greatest in the jejunum, followed by the ileum, then the duodenum. Polyps also occur in the stomach and large bowel. Polyps have also been reported in the renal pelvis, urinary bladder, ureters, lungs, nares, and gallbladder.

Adenomas also appear with increased prevalence throughout the GI tract.

The malignant potential of PJS-type hamartomatous polyps is unknown; however, the polyps can cause significant complications including bowel obstruction, rectal prolapse, and/or severe GI bleeding with secondary anemia requiring multiple emergency laparotomies and bowel resections. The age of onset of symptoms from polyps is variable, with some children developing symptoms within the first few years of life. The risk of intussusception was estimated to be 44% by age ten years and 50% by age 20 years [van Lier et al 2011a]. Intussusception risk increases with polyp size ≥15 mm [Latchford et al 2019]. One small single-center retrospective study concluded that endoscopic management of small-bowel polyps in PJS using balloon assisted endoscopy (BAE) – or when needed, laparoscopically assisted double-balloon endoscopy – decreased the occurrence of urgent laparotomy [Belsha et al 2017].

Significant interfamilial variability in the age at which polyps first appear is observed, suggesting that the natural history of polyps in a family may be a predictor of severity for offspring. Increasingly, case series have demonstrated a younger onset of GI pathology either through diagnostic testing or evaluation based on a positive family history. These data have prompted recommendations to begin surveillance at a younger age to detect and remove GI polyps, decrease risk of malignancy, and reduce complications of bowel obstruction [Wagner et al 2021, NCCN 2021]. Increasingly, the literature demonstrates the efficacy and safety of BAE and polypectomy in children with PJS.

Mucocutaneous pigmentation. Melanocytic macules (MM) are rarely present at birth; they become pronounced in most children before the fifth year, but then may fade in puberty and adulthood. Children often present with dark blue to dark brown mucocutaneous MM around the mouth, eyes, and nostrils, in the perianal area, and on the buccal mucosa. Hyperpigmented MM on the fingers are also common. In one series, 94% of individuals with PJS had perianal MM, 73% had MM that affected the digits, 65% had MM on the buccal mucosa, and 21% had MM at other sites [Utsunomiya et al 1975].

Histologically, increased melanocytes are observed at the epidermal-dermal junction, with increased melanin in the basal cells. No malignancy risk is associated with MM. In sporadic cases, MM is often the first diagnostic clue to PJS. STK11 molecular analysis has been recommended in children with characteristic PJS freckling even in the absence of additional clinical criteria [Latchford et al 2019].

Gonadal tumors. Females with PJS are at risk for ovarian sex cord tumors with annular tubules (SCTATs) and mucinous tumors of the ovaries and fallopian tubes. Symptoms include irregular or heavy menstrual periods and, occasionally, precocious puberty due to hyperestrogenism. PJS-related SCTATs are bilateral multifocal small tumors with focal calcification and a typically benign course [Wagner et al 2021]. In contrast, sporadic SCTATs are large, unilateral, and associated with a 20% risk of malignancy.

Males occasionally develop large cell calcifying Sertoli cell tumors of the testes derived from sperm cord cells. These tumors may secrete estrogen and can lead to gynecomastia, advanced skeletal age, and ultimately short stature, if untreated. Multifocal calcifications are typically seen on testicular ultrasound. Aromatase inhibitors help reverse the hormonal effects of Sertoli cell tumors including reduction of gynecomastia and slowing of linear bone growth and bone age [Crocker et al 2014].

Malignancy. Individuals with PJS are at increased risk for intestinal and extraintestinal malignancies. A meta analysis of 583 individuals with PJS found an overall cancer risk of 83% by age 70 years [Ishida et al 2016]. A cohort study of 336 individuals with PJS reported an overall cancer diagnosis of 55% by age 60 years [Chen et al 2017].

Table 2.

Cumulative Risk of Cancers in Peutz-Jeghers Syndrome

Cancer SiteGeneral Population RiskPeutz-Jeghers Syndrome
RiskMean Age at Diagnosis
Colorectal5%39%42-46 yrs
Stomach<1%29%30-40 yrs
Small bowel<1%13%37-42 yrs
Breast12.4%32%-54%37-59 yrs
Ovarian (mostly SCTAT)1.6%21%28 yrs
Cervix (adenoma malignum)<1%10%34-40 yrs
Uterine2.7%9%43 yrs
Pancreas1.5%11%-36%41-52 yrs
Testicular (Sertoli cell tumor)<1%9%6-9 yrs
Lung6.9%7%-17%47 yrs

Adapted from Syngal et al [2015]

SCTAT = sex cord tumor with annular tubules

Colorectal and gastric cancers can arise from adenomas that are commonly found in individuals with PJS. The incidence of cancer increases markedly after age 50 years. Colorectal cancer (28% by age 60) was the most common cancer reported by Chen et al [2017] in individuals with PJS.

Breast cancer can occur at early ages in women with PJS. The breast cancer risk in women with PJS may approach that of women who have a pathogenic variant in BRCA1 or BRCA2. Early-onset breast cancer has been reported in multiple families with PJS.

Ovarian cancer. In an Italian series of 61 females with PJS, three had ovarian cancer, one of which was a malignant SCTAT [Resta et al 2013]. In a Dutch series of 69 females with PJS, two females had malignant Sertoli cell ovarian tumors and one had ovarian small cell cancer [van Lier et al 2011b].

Cervical cancer. Minimal deviation adenocarcinoma (adenoma malignum) is a rare well-differentiated adenocarcinoma of the uterine cervix. Presenting symptoms include bleeding or a mucoid, watery vaginal discharge. Histologic diagnosis can be difficult on small pathologic samples and a core biopsy or endo-cervical curettage is often necessary.

The current risk for gynecologic cancer in females with PJS is estimated to range from 18% to 50% by age 50 years [Wagner et al 2021].

Testicular cancer. Malignant transformation of Sertoli cell tumors is unusual. In a series including 64 males with PJS, one testicular seminoma was reported [van Lier et al 2011b].

Pancreatic cancer is among the top causes of cancer death in the US and Europe with a dismal five-year survival after diagnosis. Recent advances in the detection of early pancreatic adenocarcinoma amenable to curative surgical resection and identification of premalignant lesions by endoscopic ultrasound and MRI/MRCP have supported surveillance in high-risk individuals including individuals with PJS [Korsse et al 2013, Goggins et al 2020].

Genotype-Phenotype Correlations

Data on genotype-phenotype correlation related to STK11 pathogenic variants are conflicting. Genotype-phenotype correlations have been extensively reviewed [Daniell et al 2018].

Variants that predict premature protein truncation are thought to predispose to a more severe phenotype. Amos et al [2004] found that individuals who had pathogenic STK11 variants that predicted premature truncation and those who tested negative for pathogenic variants had similar ages of onset for first-reported polyps or polypectomy, and those with missense variants had later onset for these symptoms. Salloch et al [2010] similarly found that persons with pathogenic STK11 variants that predicted premature truncation had more GI surgeries, a higher polyp count, and an earlier age of first polypectomy.

Pathogenic variants affecting protein kinase domain XI coded by exon 7 correlated with a 90% (9/10) incidence of GI polyp dysplasia [Wang et al 2014].

In a study of 297 individuals with PJS, the type or site of the STK11 pathogenic variant did not influence cancer risk [Lim et al 2004]. A review of 419 affected individuals found that the variant type and site within the functional domains of the expressed protein did not affect cancer risk [Hearle et al 2006a]. The risk for small-bowel intussusception was not influenced by STK11 variant status [Hearle et al 2006b]. PJS phenotype did not vary between individuals with large deletions and those with other STK11 variants [Daniell et al 2018].

Penetrance

To date all reported individuals with pathogenic variants in STK11 have shown clinical manifestations.

Nomenclature

The following terms have also been used for PJS:

  • Polyp and spots syndrome
  • Inherited hamartomatous polyps in association with mucocutaneous melanocyte macules
  • Hutchinson Weber-Peutz syndrome
  • Perioral lentiginosis (sometimes used inappropriately as a synonym for PJS)

Prevalence

Birth prevalence has not been reliably established; estimates range widely, from 1:25,000 to 1:280,000 [Tchekmedyian et al 2013].

PJS can occur in any racial or ethnic group.

Differential Diagnosis

Table 3 summarizes the differential diagnosis of Peutz-Jeghers syndrome (PJS).

Table 3.

Autosomal Dominant Genetic Cancer Syndromes Showing Signs and Symptoms that Overlap with Peutz-Jeghers Syndrome

Gene(s)SyndromeClinical Features
OverlappingDistinguishing
BMPR1A
SMAD4
Juvenile polyposis syndrome (JPS) 1GI polyps usually by age 20 yrs: hamartoma +++; adenoma +
Cancers: CRC; gastric; upper GI; pancreatic
In JPS: juvenile polyp histology different from PJS: hamartomas w/nl epithelium w/a dense stroma, an inflammatory infiltrate & smooth surface w/dilated, mucus-filled cystic glands in the lamina propria; HHT in those w/SMAD4-JPS 3; no pigmentary abnl (MM); no ovarian or testicular tumors
15q15.3q22.1 duplication 2
BMPR1A
SMAD4
Hereditary mixed polyposis syndrome (HMPS) (OMIM 601228)Polyps: juvenile hamartoma +; adenoma +; serrated +
Cancers: CRC
In HMPS: no pigmentary abnl; no ovarian or testicular tumors
PTEN PTEN hamartoma tumor syndrome (PHTS)Polyps: hamartoma +++; adenoma +; ganglioneuroma +
Cancers: breast; CRC
In PHTS: different pigmentary abnl - freckles of glans penis & keratoses; extraintestinal manifestations more pronounced than intestinal polyposis: trichilemmoma, lipomas, macrocephaly, breast fibrosis; thyroid, kidney, & endometrial cancer
PRKAR1A Carney complex (CC)Polyps: ± adeno
Hyperpigmented lesions: facial +; mucosal +
Cancers: large-cell calcifying Sertoli cell tumor
In CC: myxomas of heart, skin, breast, oropharynx, & female genital tract; thyroid nodules; acromegaly; thyroid cancer, primary pigmented nodular adrenocortical disease, & schwannomas
APC Familial adenomatous polyposis (See APC-Associated Polyposis Conditions.)Polyps: adenoma +++
Cancers: CRC; GI
In FAP: desmoid tumors, osteomas, CHRPE, brain tumors; no ovarian or testicular tumors
EPCAM
MLH1
MSH2
MSH6
PMS2
Lynch syndrome Polyps: adenoma +
Cancers: CRC; gastric; ovarian
In Lynch syndrome: sebaceous adenoma; addl cancers incl endometrial, renal pelvis & ureter; no testicular tumors

CC = Carney complex; CHRPE = congenital hypertrophy of the retinal pigment epithelium; CRC = colorectal cancer; FAP = familial adenomatous polyposis; GI = gastrointestinal; HHT = hereditary hemorrhagic telangiectasia; JPS = juvenile polyposis syndrome; MM = melanocytic macules; PJS = Peutz-Jeghers syndrome

1.

The term "juvenile" refers to the type of polyp, not the age of onset of polyps.

2.

HMPS can be caused by either a BMPR1A pathogenic variant or a duplication of 15q15.3q22.1 that leads to increased expression of GREM1 [Jaeger et al 2012]. Some families with mixed hereditary polyposis syndrome have SMAD4 pathogenic variants [Valle et al 2019].

3.

JPS may occur with hereditary hemorrhagic telangiectasia (HHT); or the combined entity of JPS-HHT. The JPS-HHT overlap syndrome has been reported in 22% of individuals with JPS due to a SMAD4 pathogenic variant (SMAD4-JPS) [Aretz et al 2007]. In a cohort 41 families with SMAD4-JPS, nearly all affected individuals had overlap syndrome [O'Malley et al 2012].

Constitutional MMR deficiency (CMMRD) – a rare childhood cancer predisposition syndrome caused by biallelic pathogenic variants in MLH1, MSH2, MSH6, or PMS2 – should also be considered in the differential diagnosis of PJS. Similar to PJS, CMMRD is associated with colorectal polyps and duodenal and gastric cancer. Unlike PJS, CMMRD is associated with a diverse malignancy spectrum in childhood and adolescence, T-cell lymphoma gliomas, and pigmentary features similar to neurofibromatosis type I (nearly all affected individuals have café au lait macules), (See Lynch Syndrome, Lynch Syndrome Variants.)

Unexplained hamartomatous mixed polyposis. In a study of 49 unrelated persons with unexplained hamartomatous mixed polyposis, Sweet et al [2005] determined that 22% had various germline pathogenic variants.

  • Of 14 individuals with juvenile-type polyposis: two had pathogenic variants in ENG, a gene associated with hereditary hemorrhagic telangiectasia; one had a hemizygous deletion encompassing PTEN and BMPR1A; and one had a SMAD4 pathogenic variant.
  • Of 23 individuals with hyperplastic/mixed polyposis, two had PTEN pathogenic variants.
  • Of nine individuals with an unknown hamartomatous polyposis, pathogenic variants were seen in STK11 (4 individuals), BMPR1A (2), and SMAD4 (1).

Oral pigmented lesions. The differential diagnosis of oral pigmented lesions includes the following:

  • Laugier-Hunziker syndrome (LHS), characterized by the presence of perioral, digit, and nailbed lentiginosis (small, well-demarcated; dark-brown to blue-black in color). This condition usually develops in adults and the hyperpigmentation is progressive. LHS has not been associated with any known exposure and has not been reported to occur in families. LHS is diagnosed by the absence of diseases associated with mucocutaneous pigmentation such as underlying PJS and Addison's disease [Sputa-Grzegrzolka et al 2020].
  • A fixed drug reaction
  • A normal feature, especially in African Americans [Sputa-Grzegrzolka et al 2020]

Rare tumors. The differential diagnosis of some of the rare tumors observed in PJS includes:

  • Sex cord tumors with annular tubules (SCTAT); 50% are associated with PJS; the remainder may occur as an isolated finding.
  • Calcifying Sertoli tumors of the testes and adenoma malignum of the cervix in women; these may also occur as an isolated finding or in other disorders.

Management

Evaluations Following Initial Diagnosis

To establish the extent of disease and need for clinical follow up in an individual diagnosed with Peutz-Jeghers syndrome (PJS), the evaluations summarized in Table 4 (if not performed as part of the evaluation that led to the diagnosis) are recommended.

Table 4.

Recommended Evaluations Following Initial Diagnosis in Individuals with Peutz-Jeghers Syndrome

System/ConcernEvaluationComment
Gastrointestinal
polyps/cancer
  • Colonoscopy
  • Upper endoscopy
  • Small-bowel exam by MRE or VCE
Beginning at age 8 yrs or earlier if symptomatic
Breast cancer
(females)
Clinical breast examBeginning at age 18 yrs
Breast MRI & mammogramBeginning at age 30 yrs
Gynecologic
cancer (females)
Exam for precocious pubertyBeginning at age 8 yrs
  • Pelvic exam for uterine & ovarian cancer (typically SCTAT)
  • Pap smear for cervical cancer (typically adenoma malignum)
Beginning at age 18-20 yrs
Testicular
cancer (males)
  • Testicular exam
  • Exam for feminizing changes in males
  • Testicular ultrasound exam if clinically indicated
Beginning at age 10 yrs
Pancreatic
cancer
Pancreatic imaging w/endoscopic ultrasound or MRI/MRCP ideally performed at center of expertiseBeginning at age 30 yrs
Genetic
counseling
By genetics professionals 1To inform patients & their families re nature, MOI, & implications of PJS in order to facilitate medical & personal decision making

MOI= mode of inheritance; MRCP = magnetic resonance cholangiopancreatography; MRE = magnetic resonance enterography; PJS = Peutz-Jeghers syndrome; SCTAT = sex cord tumors with annular tubules; VCE = video capsule endoscopy

1.

Medical geneticist, certified genetic counselor, certified advanced genetic nurse

Treatment of Manifestations

Polyps. Once the burden of gastrointestinal (GI) polyps has been established by endoscopy and imaging studies, prophylactic polypectomy of polyps larger than one centimeter is performed. This strategy has two goals:

  • To decrease the sequelae of large polyps including bleeding, anemia, obstruction, and intussusception
  • To reduce the risk for cancer by the malignant transformation of PJS-type polyps

The luminal polyp-related complications arise in childhood whereas cancer in PJS is typically seen in adulthood. Some evidence indicates that routine endoscopy and intraoperative enteroscopy with polypectomy decreases the frequency of emergency laparotomy and bowel loss. From St Mark's PJS registry of 51 affected individuals who underwent surveillance endoscopies, none had emergency surgical interventions and no GI luminal cancers were diagnosed [Latchford et al 2011]. In surveillance endoscopies in affected individuals by age 18 years, 17/28 had large gastroduodenal or colonic polyps (>1 cm). These studies demonstrate that endoscopic surveillance and polypectomy in PJS is safe and effective.

Until recently, distal small-bowel polyps that are beyond the reach of conventional endoscopy have been difficult to manage. In the past, barium contrast upper-GI series with a small-bowel follow through has been recommended. However, recent advances allow better diagnosis and eradication of small-bowel polyps, often without laparotomy and with a decrease in the radiation burden related to prior dependence on CT scan imaging [Belsha et al 2017].

  • Video capsule endoscopy (VCE) allows for better visualization of the small-bowel polyps than barium x-rays and is recommended as a first-line surveillance procedure. In children, the capsule can be deployed in the duodenum after upper endoscopy. The results of the small-bowel imaging studies direct the endoscopic removal of polyps. See Note.
  • Magnetic resonance enterography (MRE) is a reliable procedure for the detection of larger small-bowel polyps with similar sensitivity to VCE, and avoids the radiation exposure of CT enterography. CT and MR enteroclysis are alternative procedures but are less well tolerated. See Note.
  • Balloon-assisted enteroscopy (BAE) can remove distal small-bowel polyps with or without laparotomy [Gao et al 2010]. The efficacy and safety of BAE for small-bowel polyp management in PJS has been established in children with PJS [Korsse et al 2012]. BAE and polypectomy decrease the need for intraoperative enteroscopy and enterotomy, which should be reserved for affected individuals with large and distal small-bowel polyps beyond the reach of BAE.

Note: (1) VCE was preferred by individuals and detected more large polyps than MRE [Urquhart et al 2014]. (2) In three individuals, MRE detected polyps >15 mm that were not detected by VCE [Gupta et al 2010].

Intussusception should be treated in a standard manner.

Malignancies should be treated in a standard manner. Conservative management of gonadal tumors in males and females is appropriate.

Prevention of Primary Manifestations

Although not specifically studied in females with PJS, prophylactic mastectomy may be considered to manage the increased risk for breast cancer based on the family history or other clinical factors. Prophylactic hysterectomy and bilateral salpingo-oophorectomy to prevent gynecologic malignancy in women may be considered. In some disorders with a high risk for malignancy (e.g., Lynch syndrome), evidence supports this strategy [Schmeler et al 2006]. As the benefits of surveillance and prophylactic surgery have not been established, all females with PJS should be offered expert breast and gynecologic care at an established center in the framework of a study or registry [Wagner et al 2021].

Surveillance

Surveillance guidelines for PJS have evolved. With accumulation of clinical data demonstrating a high incidence of polyp-associated complications in young individuals with PJS and the greater availability of endoscopic expertise and pediatric-sized endoscopes, the trend is to begin endoscopic surveillance at age eight years. Some recommend beginning surveillance at age five years [Goldstein & Hoffenberg 2013, NCCN 2021].

Table 5a.

Recommended Surveillance Guidelines for Children and Adolescents with Peutz-Jeghers Syndrome

System/ConcernEvaluationFrequency
Gastrointestinal
polyps/cancer
Colonoscopy & upper endoscopy
  • If the baseline endoscopy at age 8 yrs is negative, rpt at age 18 yrs.
  • If polyps are detected at baseline, endoscopy every 1-3 yrs based on size, number, & histopathology
Small-bowel exam by MRE or VCE
  • Every 1-3 yrs beginning at age 8 yrs
  • Or frequency based on findings; repeat at least by age 18 yrs. 1
Gynecologic
cancer
Exam for precocious puberty in femalesAnnually beginning at age 8 yrs
Testicular
cancer
  • Testicular exam
  • Exam for feminizing changes in males
  • Testicular ultrasound exam if clinically indicated
Annually beginning at age 10 yrs

MRE = magnetic resonance enterography; VCE = video capsule endoscopy

1.

Table 5b.

Recommended Surveillance Guidelines for Adults with Peutz-Jeghers Syndrome

System/ConcernEvaluationFrequency
Gastrointestinal
polyps/cancer
  • Colonoscopy
  • Upper endoscopy
  • Small-bowel exam by MRE or VCE
Every 2-3 yrs beginning at ~18 yrs 1
Breast cancer
(women)
Clinical breast exam2x/yr beginning at age 30 yrs
Mammogram & breast MRIAnnually beginning at age 30 yrs
Gynecologic
cancer
  • Pelvic exam for uterine & ovarian cancer (typically SCTAT)
  • Pap smear for cervical cancer (typically adenoma malignum)
Annually beginning at 18-20 yrs
Pancreatic
cancer
Pancreatic imaging w/endoscopic ultrasound or MRI/MRCP ideally performed at center of expertiseAnnually beginning at ~30-35 yrs
Lung cancer Provide education about symptoms & smoking cessation. No other specific recommendations.
Testicular
cancer
Testicular ultrasound exam if clinically indicatedAnnually beginning at age 10 yrs

MRCP = magnetic resonance cholangiopancreatography; MRE = magnetic resonance enterography; SCTAT = sex cord tumors with annular tubules; VCE = video capsule endoscopy

1.

Shorter intervals may be indicated depending on polyp size, number, and pathology.

Agents/Circumstances to Avoid

No agents that increase the risk for polyps or malignancy in individuals with PJS have been described.

Due to the increased risk for cervical, lung, and pancreatic cancer in individuals with PJS, smoking should be avoided.

Evaluation of Relatives at Risk

It is appropriate to evaluate apparently asymptomatic older and younger at-risk relatives of an affected individual in order to identify as early as possible those who would benefit from initiation of treatment and preventive measures.

If the pathogenic variant in the family is known, it is appropriate to offer molecular genetic testing for the familial STK11 pathogenic variant to at-risk relatives.

  • Morbidity and mortality can be reduced in those individuals identified to have the family-specific pathogenic variant by means of early diagnosis, treatment, and surveillance (see Surveillance).
  • Relatives in whom the familial pathogenic variant is not identified (and who do not meet clinical diagnostic criteria for PJS) should not be considered as requiring PJS surveillance [Wagner et al 2021].

If the pathogenic variant in the family is not known, it is appropriate to offer:

  • Clinical diagnostic evaluations to identify those family members who will benefit from early treatment;
  • Surveillance as outlined in Surveillance to all first-degree relatives whether or not they meet diagnostic criteria.

See Genetic Counseling for issues related to testing of at-risk relatives for genetic counseling purposes.

Therapies Under Investigation

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for access to information on clinical studies for a wide range of diseases and conditions. Note: There may not be clinical trials for this rare disorder.

Other

Several animal models of PJS have been generated using STK11 knockout mice [Wei et al 2005]. Gastrointestinal hamartomatous polyposis in STK11+/– mice mimics human PJS polyps with the unique smooth muscle arborization. In these animal models, upregulation of cyclooxygenase-2 (COX-2) in polyp tissue was noted [Rossi et al 2002]. Overexpression of COX-2 in human PJS hamartomas and PJS-associated cancers has also been detected [McGarrity et al 2003, Wei et al 2003]. COX-2 inhibition in mice using celecoxib suppresses polyp growth [Udd et al 2004]. Polyp burden in STK11 (Lkb1) heterozygous (+/–) knockout mice was reduced by 86% among mice who had developed polyps and were then treated with celecoxib, a Cox inhibitor.

Selective COX-2 inhibitors have been used to inhibit premalignant adenoma formation. To date, however, no clinical trials in the US are studying efficacy of COX-2 inhibitors in reducing polyp formation in individuals with PJS. Increased cardiovascular and cerebrovascular adverse events with selective COX-2 inhibitors limit their use.

Observation of hyperactivation of mTOR in hereditary hamartoma syndrome and a variety of cancers suggests that mTOR inhibitors may be useful in the management of PJS [van Veelen et al 2011]. A significant reduction in tumor burden in STK11+/– mice treated with rapamycin was reported compared with that in mice without rapamycin treatment [Wei et al 2009]. Treatment begun before the onset of polyposis resulted in more dramatic reduction than treatment begun after onset. In another study in STK11+/– mice oral rapamycin intake was associated with a significant reduction in microvessel growth in polyps as well as in tumor burden [Robinson et al 2009].

In addition, in two small trials in persons with tuberous sclerosis complex, treatment with rapamycin induced regression of the astrocytomas [Franz et al 2006] and reduced facial angiofibroma [Hofbauer et al 2008]. Whether rapamycin would decrease polyp growth in PJS has not been documented in human studies. The mTOR inhibitor, everolimus, caused partial regression of a pancreatic cancer in an individual with PJS. Induction of apoptosis in colon polyps was also noted [Klümpen et al 2011]. These findings suggest that mTOR inhibitors are an option to investigate for management of polyposis in PJS. A Phase II trial was initiated in Utah and the Netherlands with a goal of recruiting 15 participants to evaluate chemoprevention with everolimus. The trial was stopped due to low enrollment and poor tolerance of everolimus after only two participants could be recruited [de Brabander et al 2018].

Poffenberger et al [2018] demonstrated that selective deletion of STK11 in T-cells reproduced the PJS phenotype in mice. PJS polyps in experimental mice and human were characterized by intense immune cell infiltration and increased levels of inflammatory cytokines (interleukin 6) and enhanced STAT3 signaling, providing new potential therapeutic targets for PJS.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, mode(s) of inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members; it is not meant to address all personal, cultural, or ethical issues that may arise or to substitute for consultation with a genetics professional. —ED.

Mode of Inheritance

Peutz-Jeghers syndrome (PJS) is inherited in an autosomal dominant manner.

Risk to Family Members

Parents of a proband

  • The majority of individuals diagnosed with PJS have an affected parent. In large series, 60%-78% of individuals with PJS had affected relatives [van Lier et al 2010a, Resta et al 2013].
  • Many individuals diagnosed with PJS represent apparently simplex cases (i.e., a single affected family member).
    • In the series noted above, 17%-40% of probands represent apparently simplex cases. In a separate study, between 30% and 45% of probands represent simplex cases [Daniell et al 2018]. In the absence of sufficient parental clinical and molecular data, the proportion of these individuals who have PJS as the result of a de novo pathogenic variant cannot be determined.
    • In a study of 38 individuals with PJS, 11 of 26 probands had the disorder as the result of a de novo pathogenic variant [Wu et al 2020].
  • If the proband appears to be the only affected family member, recommendations for the evaluation of parents of a proband include:
    • Molecular genetic testing if an STK11 pathogenic variant has been identified in the proband;
    • Examination of the buccal mucosa and skin of the digits and genital area for hyperpigmented macules; upper and lower gastrointestinal endoscopy; mammography; bimanual pelvic examination and ovarian ultrasound examination (females); and testicular examination (males).
  • If the pathogenic variant identified in the proband is not identified in either parent, neither parent has clinical findings consistent with PJS, and parental identity testing has confirmed biological maternity and paternity, the following possibilities should be considered:
  • The family history of some individuals diagnosed with PJS may appear to be negative because of failure to recognize the disorder in family members or early death of the parent (and other relatives) before the onset of symptoms. Therefore, an apparently negative family history cannot be confirmed unless appropriate clinical evaluation has been performed on the parents of the proband and/or molecular genetic testing has demonstrated that neither parent is heterozygous for the pathogenic variant identified in the proband.

Sibs of a proband. The risk to the sibs of the proband depends on the clinical/genetic status of the proband’s parents.

  • If a parent of the proband is affected and/or is known to have the pathogenic variant identified in the proband, the risk to the sibs of inheriting the pathogenic variant is 50%. PJS is marked by a wide range of phenotypic expression in family members with the same STK11 pathogenic variant. Epigenetic modifications, the effects of modifying genes, environmental, and other factors are known to influence the development of PJS-related manifestations in heterozygous individuals [Daniell et al 2018].
  • If the proband has a known STK11 pathogenic variant that cannot be detected in the leukocyte DNA of either parent and both parents are clinically unaffected, the recurrence risk to sibs is slightly greater than that of the general population because of the possibility of parental mosaicism [Butel-Simoes et al 2019].
  • If the genetic status of the parents is unknown but they are clinically unaffected, the risk to the sibs of a proband appears to be low. However, sibs of a proband with clinically unaffected parents are still presumed to be at increased risk for PJS because of the possibility of parental mosaicism.

Offspring of a proband

  • Each child of an individual with PJS and an identified STK11 pathogenic variant has a 50% chance of inheriting the STK11 pathogenic variant. (The offspring of individuals who are mosaic for an STK11 pathogenic variant are also at risk of inheriting an STK11 pathogenic variant.)
  • The risk to the offspring of a proband with a clinical diagnosis of PJS, a negative family history, and no identified STK11 pathogenic variant is unknown.

Other family members. The risk to other family members depends on the status of the proband's first-degree relatives: if a relative is affected and/or is heterozygous for an STK11 pathogenic variant, his or her family members are at risk.

Related Genetic Counseling Issues

See Management, Evaluation of Relatives at Risk for information on evaluating at-risk relatives for the purpose of early diagnosis and treatment.

Predictive testing for at-risk asymptomatic family members requires prior identification of the germline STK11 pathogenic variant in the family. Because early detection of at-risk individuals who have an STK11 pathogenic variant affects medical management – particularly surveillance (see Table 4) – testing of at-risk individuals (with informed parental assent) during childhood is considered beneficial [NCCN 2021, Wagner et al 2021].

Parents often want to know the genetic status of their children prior to initiating screening in order to avoid unnecessary procedures in a child who has not inherited the pathogenic variant. Special consideration should be given to education of the children and their parents prior to genetic testing. A plan should be established for the manner in which results are to be given to the parents and their children.

Genetic cancer risk assessment and counseling. For a comprehensive description of the medical, psychosocial, and ethical ramifications of identifying at-risk individuals through cancer risk assessment with or without molecular genetic testing, see Cancer Genetics Risk Assessment and Counseling – Health Professional Version (part of PDQ®, National Cancer Institute). Limited data on the effects of a PJS diagnosis on family planning and quality of life are available [van Lier et al 2010b, van Lier et al 2012]

Other issues to consider. It is recommended that physicians ordering PJS molecular genetic testing and individuals considering undergoing testing understand the risks, benefits, and limitations of the testing prior to sending a sample to a laboratory. Referral to a genetic counselor and/or a center in which testing is routinely offered is recommended.

Genetic heterogeneity. Individuals with a clinical diagnosis of PJS who do not have a detectable STK11 pathogenic variant may have a pathogenic variant(s) in a gene other than STK11 and, consequently, have a disorder that is inherited in a different manner from STK11-related PJS (see Differential Diagnosis). Of 25 individuals who had PJS but did not have a detectable STK11 pathogenic variant, one had a heterozygous pathogenic variant in MUTYH [Alhopuro et al 2008]. Of note, pathogenic variants in MUTYH ordinarily cause an autosomal recessive form of adenomatous polyposis coli.

Family planning

  • The optimal time for determination of genetic risk and discussion of the availability of prenatal/preimplantation genetic testing is before pregnancy. Similarly, decisions about testing to determine the genetic status of at-risk asymptomatic family members are best made before pregnancy.
  • It is appropriate to offer genetic counseling (including discussion of potential risks to offspring and reproductive options) to young adults who are affected or at risk.

DNA banking. Because it is likely that testing methodology and our understanding of genes, allelic variants, and diseases will improve in the future, consideration should be given to banking DNA from probands in whom a molecular diagnosis has not been confirmed (i.e., the causative genetic alteration/s are unknown).

Prenatal Testing and Preimplantation Genetic Testing

Once the STK11 pathogenic variant has been identified in an affected family member, prenatal testing and preimplantation genetic testing for PJS are possible.

Differences in perspective may exist among medical professionals and within families regarding the use of prenatal testing. While most centers would consider use of prenatal testing to be a personal decision, discussion of these issues may be helpful.

Resources

GeneReviews staff has selected the following disease-specific and/or umbrella support organizations and/or registries for the benefit of individuals with this disorder and their families. GeneReviews is not responsible for the information provided by other organizations. For information on selection criteria, click here.

Molecular Genetics

Information in the Molecular Genetics and OMIM tables may differ from that elsewhere in the GeneReview: tables may contain more recent information. —ED.

Table A.

Peutz-Jeghers Syndrome: Genes and Databases

GeneChromosome LocusProteinLocus-Specific DatabasesHGMDClinVar
STK11 19p13​.3 Serine/threonine-protein kinase STK11 STK11 database STK11 STK11

Data are compiled from the following standard references: gene from HGNC; chromosome locus from OMIM; protein from UniProt. For a description of databases (Locus Specific, HGMD, ClinVar) to which links are provided, click here.

Table B.

OMIM Entries for Peutz-Jeghers Syndrome (View All in OMIM)

175200PEUTZ-JEGHERS SYNDROME; PJS
602216SERINE/THREONINE PROTEIN KINASE 11; STK11

Molecular Pathogenesis

STK11 encodes a serine/threonine-protein kinase (STK11), a multi-tasking tumor suppressor that has a role in apoptosis, cell cycle arrest, cell proliferation, cell polarity, and energy metabolism.

  • STK11 activates TSC2 through an AMP-dependent protein kinase [Corradetti & Guan 2006] leading to accumulation of mTOR, which is critical for protein translation. Loss of STK11 increased tumor cell metabolism via mTOR [Faubert et al 2014].
  • STK11 expression was shown to cause apoptosis in epithelial cells [Marignani 2005].
  • STK11 was reported to phosphorylate AMPK and several other members of the AMPK-related subfamily of kinases including the microtubule affinity-regulating kinases (MARKs) to regulate cell polarity [Tanwar et al 2012].
  • Through activation of AMPK by phosphorylation, STK11 plays a role in energy metabolism [Udd & Mäkelä 2011].
  • Loss of STK11 increased tumor-promoting cytokines with reduced expression of PD-1 ligand in mouse and human tumors [Koyama et al 2016].

More than 300 STK11 pathogenic variants have been reported in persons with Peutz-Jeghers syndrome. All types of variants have been reported, from missense variants to whole-gene deletion.

Mechanism of disease causation. Loss of function

Cancer and Benign Tumors

Non-small cell lung cancer. Sporadic pathogenic variants in STK11 are observed in one third of non-small cell lung cancers; STK11 is the third most commonly mutated gene, following p53 and KRAS. Demographically, STK11 variants in lung tumors appear to be frequent in male smokers of northern European background and are associated with poorly differentiated tumors. In addition, STK11 pathogenic variants are more frequently observed in association with concomitant activating KRAS pathogenic variants. Such digenic variants have been associated with a poorer prognosis compared to a KRAS pathogenic variant alone [Pécuchet et al 2017]. The high prevalence of STK11 pathogenic variants in non-small cell lung cancers suggests that cigarette smoking could be particularly harmful for individuals with pathogenic variants in STK11. The presence of STK11 pathogenic variants in these types of cancers may indicate a more aggressive tumor that has a poorer prognosis and is seen more often in smokers [Pécuchet et al 2017, Wang et al 2020].

Cervical cancer. Sporadic pathogenic variants in STK11 are observed in at least 20% of cervical cancers [Wingo et al 2009]. Among the variants observed in tumors, half are single-nucleotide variants or small indels and the other half are large-fragment deletions in either one allele or both alleles. STK11 inactivation in primary cervical tumors has been associated with accelerated disease progression. These results suggest that STK11 is a major cervical tumor suppressor for sporadic cancers.

Chapter Notes

Author Notes

Dr Amos is a research scholar in the Cancer Prevention Research Institute of Texas supported by RR170048. Dr McGarrity is a gastroenterologist who partially specializes in the diagnosis and treatment of PJS. Dr Baker is a genetic counselor and medical geneticist who specializes in the recognition of hereditary cancer predisposition syndromes.

Author History

Christopher I Amos, PhD (2001-present)
Maria J Baker, PhD (2016-present)
Marsha L Frazier, PhD; University of Texas MD Anderson Cancer Center (2001-2016)
Thomas J McGarrity, MD (2001-present)
Chongjuan Wei, PhD; University of Texas MD Anderson Cancer Center (2001-2016)

Revision History

  • 2 September 2021 (sw) Comprehensive update posted live
  • 14 July 2016 (sw) Comprehensive update posted live
  • 25 July 2013 (me) Comprehensive update posted live
  • 22 February 2011 (me) Comprehensive update posted live
  • 2 November 2010 (me) Comprehensive update posted live
  • 15 May 2007 (me) Comprehensive update posted live
  • 19 May 2004 (ca) Revision: Genetic Counseling
  • 26 November 2003 (me) Comprehensive update posted live
  • 23 February 2001 (me) Review posted live
  • 11 July 2000 (ca) Original submission

References

Published Guidelines / Consensus Statements

  • American Society of Clinical Oncology. Policy statement update: genetic testing for cancer susceptibility. Available online. 2010. Accessed 8-31-21.

Literature Cited

  • Alhopuro P, Phichith D, Tuupanen S, Sammalkorpi H, Nybondas M, Saharinen J, Robinson JP, Yang Z, Chen LQ, Orntoft T, Mecklin JP, Järvinen H, Eng C, Moeslein G, Shibata D, Houlston RS, Lucassen A, Tomlinson IP, Launonen V, Ristimäki A, Arango D, Karhu A, Sweeney HL, Aaltonen LA. Unregulated smooth-muscle myosin in human intestinal neoplasia. Proc Natl Acad Sci U S A. 2008;105:5513–8. [PMC free article: PMC2291082] [PubMed: 18391202]
  • Amos CI, Keitheri-Cheteri MB, Sabripour M, Wei C, McGarrity TJ, Seldin MF, Nations L, Lynch PM, Fidder HH, Friedman E, Frazier ML. Genotype-phenotype correlations in Peutz-Jeghers syndrome. J Med Genet. 2004;41:327–33. [PMC free article: PMC1735760] [PubMed: 15121768]
  • Aretz S, Stienen D, Uhlhaas S, Stolte M, Entius MM, Loff S, Back W, Kaufmann A, Keller KM, Blaas SH, Siebert R, Vogt S, Spranger S, Holinski-Feder E, Sunde L, Propping P, Friedl W. High proportion of large genomic deletions and a genotype phenotype update in 80 unrelated families with juvenile polyposis syndrome. J Med Genet. 2007;44:702–9. [PMC free article: PMC2752176] [PubMed: 17873119]
  • Beggs AD, Latchford AR, Vasen HF, Moslein G, Alonso A, Aretz S, Bertario L, Blanco I, Bülow S, Burn J, Capella G, Colas C, Friedl W, Møller P, Hes FJ, Järvinen H, Mecklin JP, Nagengast FM, Parc Y, Phillips RK, Hyer W, Ponz de Leon M, Renkonen-Sinisalo L, Sampson JR, Stormorken A, Tejpar S, Thomas HJ, Wijnen JT, Clark SK, Hodgson SV. Peutz-Jeghers syndrome: a systematic review and recommendations for management. Gut. 2010;59:975–86. [PubMed: 20581245]
  • Belsha D, Urs A, Attard T, Thomson M. Effectiveness of double-balloon enteroscopy-facilitated polypectomy in pediatric patients with Peutz-Jeghers syndrome. J Pediatr Gastroenterol Nutr. 2017;65:500–2. [PubMed: 28319601]
  • Borun P, DeRosa M, Nedoszytko B, Walkowiak J, Plawski A. Specific Alu elements involved in a significant percentage of copy number variations of the STK11 gene in patients with Peutz-Jeghers syndrome. Fam Cancer. 2015;14:455–61. [PMC free article: PMC4559094] [PubMed: 25841653]
  • Butel-Simoes GI, Spigelman AD, Scott RJ, Vilain RE. Low-level parental mosaicism in an apparent de novo case of Peutz-Jeghers syndrome. Fam Cancer. 2019;18:109–12. [PubMed: 29948449]
  • Chen HY, Jin XW, Li BR, Zhu M, Li J, Mao GP, Zhang YF, Ning SB. Cancer risk in patients with Peutz-Jeghers syndrome: A retrospective cohort study of 336 cases. Tumour Biol. 2017;39:1010428317705131. [PubMed: 28653895]
  • Corradetti MN, Guan KL. Upstream of the mammalian target of rapamycin: do all roads pass through mTOR? Oncogene. 2006;25:6347–60. [PubMed: 17041621]
  • Crocker MK, Gourgari E, Lodish M, Stratakis CA. Use of aromatase inhibitors in large cell calcifying Sertoli cell tumors: effects on gynecomastia, growth velocity and bone age. J Clin Endocrinol Metab. 2014;99:E2673–80. [PMC free article: PMC4255117] [PubMed: 25226294]
  • Daniell J, Plazzer JP, Perera A, Macrae F. An exploration of genotype-phenotype link between Peutz-Jeghers syndrome and STK11: a review. Fam Cancer. 2018;17:421–7. [PubMed: 28900777]
  • de Brabander J, Eskens FALM, Korsse SE, Dekker E, Dewint P, van Leerdam ME, van Eeden S, Klümpen HJ. Chemoprevention in patients with Peutz-Jeghers syndrome: lessons learned. Oncologist. 2018;23:399–e33. [PMC free article: PMC5896716] [PubMed: 29371475]
  • De Rosa M, Galatola M, Quaglietta L, Miele E, De Palma G, Rossi GB, Staiano A, Izzo P. Alu-mediated genomic deletion of the serine/threonine protein kinase 11 (STK11) gene in Peutz-Jeghers syndrome. Gastroenterology. 2010;138:2558–60. [PubMed: 20435009]
  • Faubert B, Vincent E, Griss T, Samborska B, Izreig S, Svensson R, Mamer O, Avizonis D, Shackelford D, Shaw R, Jones R. Loss of the tumor suppressor LKB1 promotes metabolic reprogramming of cancer cells via HIF-1a. Proc Natl Acad Sci U S A. 2014;111:2554–9. [PMC free article: PMC3932920] [PubMed: 24550282]
  • Franz DN, Leonard J, Tudor C, Chuck G, Care M, Sethuraman G, Dinopoulos A, Thomas G, Crone KR. Rapamycin causes regression of astrocytomas in tuberous sclerosis complex. Ann Neurol. 2006;59:490–8. [PubMed: 16453317]
  • Gao H, van Lier MG, Poley JW, Kuipers EJ, van Leerdam ME, Mensink PB. Endoscopic therapy of small-bowel polyps by double-balloon enteroscopy in patients with Peutz-Jeghers syndrome. Gastrointest Endosc. 2010;71:768–73. [PubMed: 20188368]
  • Goggins M, Overbeek KA, Brand R, Syngal S, Del Chiaro M, Bartsch DK, Bassi C, Carrato A, Farrell J, Fishman EK, Fockens P, Grees TM, van Hooft JE, Hruban RH, Kastrinos F, Klein A, Lennon AM, Lucas A, Park W, Rustgi A, Simeone D, Stoffel E, Vasen HFA, Cahen DL, Canto MI, Bruno M, et al. Management of patients with increased risk for familial pancreatic cancer: updated recommendations from the international cancer of the pancreas screening (CAPS) consortium. Gut. 2020;69:7–17. [PMC free article: PMC7295005] [PubMed: 31672839]
  • Goldstein SA, Hoffenberg EJ. Peutz-Jegher syndrome in childhood: need for updated recommendations? J Pediatr Gastroenterol Nutr. 2013;56:191–5. [PubMed: 23325439]
  • Gupta A, Postgate AJ, Burling D, Ilangovan R, Marshall M, Phillips RK, Clark SK, Fraser CH. A prospective study of MR enterography versus capsule endoscopy for the surveillance of adult patients with Peutz-Jeghers syndrome. Am J Roentgenol. 2010;195:108–16. [PubMed: 20566803]
  • Hearle N, Schumacher V, Menko FH, Olschwang S, Boardman LA, Gille JJ, Keller JJ, Westerman AM, Scott RJ, Lim W, Trimbath JD, Giardiello FM, Gruber SB, Offerhaus GJ, de Rooij FW, Wilson JHP, Hansmann A, Moslein G, Royer-Pokora B, Vogel T, Phillips RKS, Spigelman AD, Houlston RS. Frequency and spectrum of cancers in the Peutz-Jeghers syndrome. Clin Cancer Res. 2006a;12:3209–15. [PubMed: 16707622]
  • Hearle N, Schumacher V, Menko FH, Olschwang S, Boardman LA, Gille JJ, Keller JJ, Westerman AM, Scott RJ, Lim W, Trimbath JD, Giardiello FM, Gruber SB, Offerhaus GJ, Rooij FW, Wilson JH, Hansmann A, Moslein G, Royer-Pokora B, Vogel T, Phillips RK, Spigelman AD, Houlston RS. STK11 status and intussusception risk in Peutz-Jeghers syndrome. J Med Genet. 2006b;43:e41. [PMC free article: PMC2564597] [PubMed: 16882735]
  • Hofbauer GF, Marcollo-Pini A, Corsenca A, Kistler AD, French LE, Wüthrich RP, Serra AL. The mTOR inhibitor rapamycin significantly improves facial angiofibroma lesions in a patient with tuberous sclerosis. Br J Dermatol. 2008;159:473–5. [PubMed: 18547304]
  • Ishida H, Tajina Y, Gorsta T, Kumamoto K, Ishibashi K, Iwon T. Update on our investigation of malignant tumors associated with Peutz-Jeghers Syndrome in Japan. Surg Today. 2016;46:1231–42. [PubMed: 26746637]
  • Jaeger E, Leedham S, Lewis A, Segditsas S, Becker M, Cuadrado PR, Davis H, Kaur K, Heinimann K, Howarth K, East J, Taylor J, Thomas H, Tomlinson I. Hereditary mixed polyposis syndrome is caused by a 40-kb upstream duplication that leads to increased and ectopic expression of the BMP antagonist GREM1. Nat Genet. 2012;44:699–703. [PMC free article: PMC4594751] [PubMed: 22561515]
  • Jelsig AM, Bertelsen B, Forss I, Karstensen JG. Two cases of somatic STK11 mosaicism in Danish patients with Peutz-Jeghers syndrome. Familial Cancer. 2021;20:55–9. [PubMed: 32504210]
  • Klümpen HJ, Queiroz KC, Spek CA, van Noesel CJ, Brink HC, de Leng WW, de Wilde RF, Mathus-Vliegen EM, Offerhaus GJ, Alleman MA, Westermann AM, Richel DJ. mTOR inhibitor treatment of pancreatic cancer in a patient with Peutz-Jeghers syndrome. J Clin Oncol. 2011;29:e150–3. [PMC free article: PMC4856022] [PubMed: 21189378]
  • Korsse SE, Dewint P, Kuipers EJ, van Leerdam ME. Small bowel endoscopy and Peutz-Jeghers syndrome. Best Pract Res Clin Gastroenterol. 2012;26:261–78.
  • Korsse SE, Harinck F, van Lier MG, Biermann K, Offerhaud GJ, Krak N, Looman CW, van Veelen W, Kuipers EJ, Wagner A, Deffer E, Mathus-Vliegen EM, Fockens P, van Leerdam ME, Bruno MJ. Pancreatic cancer risk in Peutz-Jeghers syndrome patients: a large cohort study and implications for surveillance. J Med Genet. 2013;50:59–64. [PubMed: 23240097]
  • Koyama S, Akbay E, Li Y, Aref A, Skoulidis F, Herter-Sprie G, Buczkowski K, Liu Y, Awad M, Denning W, Jing Wang J, Parra-Cuentas E, Wistuba I, Soucheray M, Thai T, Asahina H, Kitajima S, Altabef A, Cavanaugh J, Rhee K, Gao P, Zhang H, Fecci P, Shimamura T, Hellmann M, Heymach J, Hodi S, Freeman G, Barbie D, Dranoff G, Hammerman P, Wong K. STK11/LKB1 Deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment. Cancer Res. 2016;76:999–1008. [PMC free article: PMC4775354] [PubMed: 26833127]
  • Latchford A, Cohen S, Auth M, Scaillon M, Viala J, Daniels R, Talbotec C, Attard T, Durno C, Hyer W. Management of Peutz-Jeghers syndrome in children and adolescents: a position paper from the ESPGHAN Polyposis Working Group. J Pediatr Gastroenterol Nutr. 2019;68:442–52. [PubMed: 30585892]
  • Latchford AR, Neale K, Phillips RKS, Clark SK. Peutz-Jeghers syndrome: Intriguing suggestions of gastrointestinal cancer prevention from surveillance. Dis Colon Rectum. 2011;54:1547–51. [PubMed: 22067184]
  • Lim W, Olschwang S, Keller JJ, Westerman AM, Menko FH, Boardman LA, Scott RJ, Trimbath J, Giardiello FM, Gruber SB, Gille JJ, Offerhaus GJ, de Rooij FW, Wilson JH, Spigelman AD, Phillips RK, Houlston RS. Relative frequency and morphology of cancers in STK11 mutation carriers. Gastroenterology. 2004;126:1788–94. [PubMed: 15188174]
  • Marignani PA. LKB1, the multitasking tumor suppression kinase. J Clin Pathol. 2005;58:15–9. [PMC free article: PMC1770539] [PubMed: 15623475]
  • Massa G, Roggen N, Renard M, Gille JJ. Germline mutation in the STK11 gene in a girl with an ovarian Sertoli cell tumour. Eur J Pediatr. 2007;166:1083–5. [PubMed: 17124587]
  • McGarrity TJ, Peiffer LP, Amos CI, Frazier ML, Ward MG, Howett MK. Overexpression of cyclooxygenase 2 in hamartomatous polyps of Peutz-Jeghers syndrome. Am J Gastroenterol. 2003;98:671–8. [PubMed: 12650805]
  • NCCN. Genetic/Familial High-Risk Assessment: Colorectal. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology (NCCN Guidelines). Version 1.2021 (May 11, 2021). Available online. Registration required. 2021.
  • O'Malley M, LaGuardia L, Kalady M, Parambil J, Heald B, Eng C, Church J, Burke C. The prevalence of hereditary hemorrhagic telangiectasia in juvenile polyposis syndrome. Dis Colon Rectum. 2012;55:886–92. [PubMed: 22810475]
  • Pécuchet N, Laurent-Puig P, Mansuet-Lupo A, Legras A, Alifano M, Pallier K, Didelot A, Gibault L, Danel C, Just PA, Riquet M, Le Pimpec-Barthes F, Damotte D, Fabre E, Blons H. Different prognostic impact of STK11 mutations in non-squamous non-small-cell lung cancer. Oncotarget. 2017;8:23831–40. [PMC free article: PMC5410347] [PubMed: 26625312]
  • Poffenberger MC, Metcalfe-Roach A, Aguilar E, Chen J, Hsu BE, Wong AH, Johnson RM, Flynn B, Samborska B, Ma EH, Gravel SP, Tonelli L. LKB1 deficiency in T cells promotes the development of gastrointestinal polyposis. Science. 2018;361:406–11. [PubMed: 30049881]
  • Resta N, Pierannunzio D, Lenato GM, Stella A, Capocaccia R, Bagnulo R, Lastella P, Susca FC, Bozzao C, Loconte DC, Sabbà C, Urso E, Sala P, Fornasarig M, Grammatico P, Piepoli A, Host C, Turchetti D, Viel A, Memo L, Giunti L, Stigliano V, Varesco L, Bertario L, Genuardi M, Lucci Cordisco E, Tibiletti MG, Di Gregorio C, Andriulli A, Ponz de Leon M. AIFEG. Cancer risk associated with STK11/LKB1 germline mutations in Peutz-Jeghers syndrome patients: results of an Italian multicenter study. Dig Liver Dis. 2013;45:606–11. [PubMed: 23415580]
  • Robinson J, Lai C, Martin A, Nye E, Tomlinson I, Silver A. Oral rapamycin reduces tumour burden and vascularization in Lkb1(+/-) mice. J Pathol. 2009;219:35–40. [PubMed: 19434632]
  • Rossi DJ, Ylikorkala A, Korsisaari N, Salovaara R, Luukko K, Launonen V, Henkemeyer M, Ristimaki A, Aaltonen LA, Makela TP. Induction of cyclooxygenase-2 in a mouse model of Peutz-Jeghers polyposis. Proc Natl Acad Sci U S A. 2002;99:12327–32. [PMC free article: PMC129444] [PubMed: 12218179]
  • Rosty C. The role of the surgical pathologist in the diagnosis of gastrointestinal polyposis syndromes. Adv Anat Pathol. 2018;25:1–13. [PubMed: 28901964]
  • Salloch H, Reinacher-Schick A, Schulmann K, Pox C, Willert J, Tannapfel A, Heringlake S, Goecke TO, Aretz S, Stemmler S, Schmiegel W. Truncating mutations in Peutz-Jeghers syndrome are associated with more polyps, surgical interventions and cancers. Int J Colorectal Dis. 2010;25:97–107. [PubMed: 19727776]
  • Schmeler KM, Lynch HT, Chen LM, Munsell MF, Soliman PT, Clark MB, Daniels MS, White KG, Boyd-Rogers SG, Conrad PG, Yang KY, Rubin MM, Sun CC, Slomovitz BM, Gershenson DM, Lu KH. Prophylactic surgery to reduce the risk of gynecologic cancers in the Lynch syndrome. N Engl J Med. 2006;354:261–9. [PubMed: 16421367]
  • Sputa-Grzegrzolka P, Wozniak Z, Akutko K, Pytrus T, Baran W, Calik J, et al. Laugier-Hunziker syndrome: a case report of the pediatric patient and review of the literature. Int J Dermatol. 2020;59:1513–9. [PubMed: 33118627]
  • Sweet K, Willis J, Zhou XP, Gallione C, Sawada T, Alhopuro P, Khoo SK, Patocs A, Martin C, Bridgeman S, Heinz J, Pilarski R, Lehtonen R, Prior TW, Frebourg T, Teh BT, Marchuk DA, Aaltonen LA, Eng C. Molecular classification of patients with unexplained hamartomatous and hyperplastic polyposis. JAMA. 2005;294:2465–73. [PubMed: 16287957]
  • Syngal S, Brand RE, Church JM, Giardiello FM, Hampel HL, Burt RW. Clinical Guideline: Genetic Testing and Management of Hereditary Gastrointestinal Cancer Syndromes. Am J Gastroenterol. 2015;110:223–62. [PMC free article: PMC4695986] [PubMed: 25645574]
  • Tanwar PS, Kaneko-Tarui T, Zhang L, Teixeira JM. Altered LKB1/AMPK/TSC1/TSC2/mTOR signaling causes disruption of Sertoli cell polarity and spermatogenesis. Hum Mol Genet. 2012;21:4394–405. [PMC free article: PMC3459463] [PubMed: 22791749]
  • Tchekmedyian A, Amos CI, Bale SJ, Zhu D, Arold S, Berrueta J, Nabon N, McGarrity T. Findings from the Peutz-Jeghers syndrome registry of Uruguay. PLoS One. 2013;8:e79639. [PMC free article: PMC3834183] [PubMed: 24260271]
  • Tse JY, Wu S, Shinagare SA, Lauwers GY, Yilmaz O, Wu CL, Deshpande V. Peutz-Jeghers syndrome: a critical look at colonic Peutz-Jeghers polyps. Mod Pathol. 2013;26:1235–40. [PubMed: 23599156]
  • Udd L, Katajisto P, Rossi DJ, Lepistö A, Lahesmaa AM, Ylikorkala A, Järvinen HJ, Ristimäki AP, Mäkelä TP. Suppression of Peutz-Jeghers polyposis by inhibition of cyclooxygenase-2. Gastroenterology. 2004;127:1030–7. [PubMed: 15480979]
  • Udd L, Mäkelä TP. LKB1 Signaling in advancing cell differentiation. Fam Cancer. 2011;10:425–35. [PubMed: 21519908]
  • Urquhart P, Grimpen F, Lim GJ, Pizzey C, Stella DL, Tesar PA, Macrae FA, Appleyard MA, Brown GJ. Capsule endoscopy versus magnetic resonance enterography for the detection of small bowel polyps in Peutz-Jeghers syndrome. Fam Cancer. 2014;13:249–55. [PubMed: 24509884]
  • Utsunomiya J, Gocho H, Miyanaga T, Hamaguchi E, Kashimure A. Peutz-Jeghers syndrome: its natural course and management. Johns Hopkins Med J. 1975;136:71–82. [PubMed: 1117595]
  • Valle L, de Voer R, Sjursen W, Försti A, Ruiz-Ponte C, Caldés T, Garré P, Olsen M, Nordling M, Castellvi-Bel S, Hemminki K. Update on genetic predisposition to colorectal cancer and polyposis. Mol Aspects Med. 2019;69:10–26. [PubMed: 30862463]
  • van Lier M, Korsse SE, Mathus-Vliegen E, Kuipers EJ, van den Ouweland A, Vanheusden K, van Leerdam M, Wagner A. Peutz-Jeghers syndrome and family planning: the attitude towards prenatal diagnosis and pre-implantation genetic diagnosis. Eur J Hum Genet. 2012;20:236–9. [PMC free article: PMC3260925] [PubMed: 21829227]
  • van Lier MG, Mathus-Vliegen EM, Wagner A, van Leerdam ME, Kuipers EJ. High cumulative risk of intussusception in patients with Peutz-Jeghers syndrome: time to update surveillance guidelines? Time to update surveillance guideline. Am J Gastroenterol. 2011a;106:940–5. [PubMed: 21157440]
  • van Lier MG, Westerman AM, Wagner A, Looman CW, Wilson JH, de Rooij FW, Lemmens VE, Kuipers EJ, Mathus-Vliegen EM, van Leerdam ME. High cancer risk and increased mortality in patients with Peutz-Jeghers syndrome. Gut. 2011b;60:141–7. [PubMed: 21205875]
  • van Lier MGF, Wagner A, Mathus-Vliegen EMH, Kuipers EJ, Steyerberg EW, van Leerdam ME. High cancer risk in Peutz-Jeghers syndrome: A systematic review and surveillance recommendations. Am J Gastroenterol. 2010a;105:1258–64. [PubMed: 20051941]
  • van Lier MGF, Mathus-Vliegen EMH, van Leerdam ME, Kuipers EJ, Looman CWN, Wagner A, Vanheusden K. Quality of life and psychological distress in patients with Peutz-Jeghers syndrome. Clin Genet. 2010b;78:219–26. [PubMed: 20695872]
  • van Veelen W, Korsse SE, Van de Laar L. The long and winding road to rational treatment of cancer associated with LKB1/AMPK/TSC/mTORC1 signaling. Oncogene. 2011;30:2289–303. [PubMed: 21258412]
  • Wagner A, Aretz S, Auranen A, Bruno MJ, Cavestro GM, Crosbie EJ, Goverde A, Jelsig AM, Latchford AR, Leerdam MEV, Lepisto AH, Puzzono M, Winship I, Zuber V, Möslein G. The management of Peutz-Jeghers Syndrome: European Hereditary Tumour Group (EHTG) guideline. J Clin Med. 2021;10:473. [PMC free article: PMC7865862] [PubMed: 33513864]
  • Wang H, Guo J, Shang X, Wang Z. Less immune cell infiltration and worse prognosis after immunotherapy for patients with lung adenocarcinoma who harbored STK11 mutation. Int Immunopharmacol. 2020;84:1060574.
  • Wang Z, Wu B, Mosig RA, Chen Y, Ye F, Zhang Y, Gong W, Gong L, Huang F, Wang X, Nie B, Zheng H, Cui M, Wang Y, Wang J, Chen C, Polydorides AD, Zhang DY, Martignetti JA, Jiang B. STK11 domain XI mutations: candidate genetic drivers leading to the development of dysplastic polyps in Peutz-Jeghers syndrome. Hum Mutat. 2014;35:851–8. [PubMed: 24652667]
  • Wei C, Amos CI, Rashid A, Sabripour M, Nations L, McGarrity TJ, Frazier ML. Correlation of staining for LKB1 and COX-2 in hamartomatous polyps and carcinomas from patients with Peutz-Jeghers syndrome. J Histochem Cytochem. 2003;51:1665–72. [PubMed: 14623934]
  • Wei C, Amos CI, Stephens LC, Campos I, Deng JM, Behringer RR, Rashid A, Frazier ML. Mutation of Lkb1 and p53 genes exert a cooperative effect on tumorigenesis. Cancer Res. 2005;65:11297–303. [PubMed: 16357136]
  • Wei C, Amos CI, Zhang N, Zhu J, Wang X, Frazier ML. Chemopreventive efficacy of rapamycin on Peutz-Jeghers syndrome in a mouse model. Cancer Lett. 2009;277:149–54. [PMC free article: PMC2966387] [PubMed: 19147279]
  • Wingo SN, Gallardo TD, Akbay EA, Liang MC, Contreras CM, Boren T, Shimamura T, Miller DS, Sharpless NE, Bardeesy N, Kwiatkowski DJ, Schorge JO, Wong KK, Castrillon DH. Somatic LKB1 mutations promote cervical cancer progression. PLoS One. 2009;4:e5137. [PMC free article: PMC2660434] [PubMed: 19340305]
  • Wu BD, Wang YJ, Fan LL, Huang H, Zhou P, Yang M, Shi XL. Clinical and genetic analyses of 38 Chnese patients with Peutz-Jeghers syndrome. Biomed Res Int. 2020;2020:9159315. [PMC free article: PMC7240661] [PubMed: 32462036]
Copyright © 1993-2024, University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

GeneReviews® chapters are owned by the University of Washington. Permission is hereby granted to reproduce, distribute, and translate copies of content materials for noncommercial research purposes only, provided that (i) credit for source (http://www.genereviews.org/) and copyright (© 1993-2024 University of Washington) are included with each copy; (ii) a link to the original material is provided whenever the material is published elsewhere on the Web; and (iii) reproducers, distributors, and/or translators comply with the GeneReviews® Copyright Notice and Usage Disclaimer. No further modifications are allowed. For clarity, excerpts of GeneReviews chapters for use in lab reports and clinic notes are a permitted use.

For more information, see the GeneReviews® Copyright Notice and Usage Disclaimer.

For questions regarding permissions or whether a specified use is allowed, contact: ude.wu@tssamda.

Bookshelf ID: NBK1266PMID: 20301443

Views

Tests in GTR by Gene

Related information

  • MedGen
    Related information in MedGen
  • OMIM
    Related OMIM records
  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed
  • Gene
    Locus Links

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...