Bookshelf » GeneReviews » PTEN Hamartoma Tumor Syndrome (PHTS)
 
gene
GeneReviews
PagonRoberta A
BirdThomas C
DolanCynthia R
SmithRichard JH
StephensKaren
University of Washington, Seattle2009
geneticspublic health

GeneTests Home Page About GeneTests Search GeneReviews on the GeneTests web site Laboratory Directory Clinic Directory Educational Materials Illustrated Glossary

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Genetics clinics are a source of information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

Support groups have been established for individuals and families to provide information, support, and contact with other affected individuals. The Resources section may include disease-specific and/or umbrella support organizations.

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

Genetic counseling is the process of providing individuals and families with information on the nature, 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. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

PTEN Hamartoma Tumor Syndrome (PHTS)

Kevin M Zbuk, MD
Genomic Medicine Institute
Cleveland Clinic Foundation
Jennifer L Stein, MS, CGC
Genomic Medicine Institute
Cleveland Clinic Foundation
Charis Eng, MD, PhD
Genomic Medicine Institute
Cleveland Clinic Foundation
and
Department of Genetics
Case Western Reserve University School of Medicine
Cleveland, OH
10012006phts
Initial Posting: November 29, 2001.
Last Update: January 10, 2006.

*

*

*

Summary

Disease characteristics. The PTEN hamartoma tumor syndrome (PHTS) includes Cowden syndrome (CS), Bannayan-Riley-Ruvalcaba syndrome (BRRS), Proteus syndrome (PS), and Proteus-like syndrome. CS is a multiple hamartoma syndrome with a high risk of benign and malignant tumors of the thyroid, breast, and endometrium. Affected individuals usually have macrocephaly, trichilemmomas, and papillomatous papules and present by the late 20s. The lifetime risk of developing breast cancer is 25%-50%, with an average age of diagnosis between 38 and 46 years; the lifetime risk for thyroid cancer (usually follicular, rarely papillary, but never medullary thyroid cancer) is around 10%, and the risk for endometrial cancer may approach 5%-10%. BRRS is a congenital disorder characterized by macrocephaly, intestinal polyposis, lipomas, and pigmented macules of the glans penis. PS is a complex, highly variable disorder involving congenital malformations and hamartomatous overgrowth of multiple tissues, as well as connective tissue nevi, epidermal nevi, and hyperostoses. Proteus-like syndrome is undefined but refers to individuals with significant clinical features of PS who do not meet the diagnostic criteria for PS.

Diagnosis/testing. The diagnosis of PHTS is made only when a PTEN mutation is identified. Approximately 80% of individuals who meet the diagnostic criteria for CS and 60% of individuals with a clinical diagnosis of BRRS have a detectable PTEN gene mutation. Preliminary data also suggest that up to 50% of individuals with a Proteus-like syndrome and up to 20% of individuals with Proteus syndrome have PTEN mutations. Full sequencing of the PTEN gene is available on a clinical basis.

Management. Because the most serious consequences of PHTS relate to the increased risk of breast, thryoid, endometrial, and renal cancers, the most important aspect of management of an individual with a PTEN mutation is increased cancer surveillance. Surveillance in general for individuals with PTEN mutations (with CS, BRRS, PS, or Proteus-like syndromes) includes annual physical examination from age 18 years, annual urinalysis, and baseline colonoscopy at age 50 years. Specific surveillance for breast cancer in individuals with CS includes monthly self-examination beginning at age 18 years (for females and males), annual clinical breast examinations beginning at age 25 years, and annual mammography and breast MRI beginning at age 30-35 years; surveillance for thyroid cancer includes baseline thyroid ultrasound examination at age 18 years and annual thyroid ultrasound examinations; surveillance for endometrial cancer includes annual suction biopsies beginning at age 35-40 years for premenopausal women and annual transvaginal ultrasound examination for postmenopausal women. Topical agents (e.g., 5-fluorouracil), curettage, cryosurgery, or laser ablation may alleviate the mucocutaneous manifestations of CS; cutaneous lesions should be excised only if malignancy is suspected or symptoms (e.g., pain, deformity) are significant. Molecular testing of asymptomatic at-risk relatives can identify those who have a family-specific PTEN mutation and ensure appropriate surveillance.

Genetic counseling. PHTS is inherited in an autosomal dominant manner. Because CS is likely underdiagnosed, the actual proportion of simplex cases (defined as individuals with no obvious family history) and familial cases (defined as two or more related affected individuals) cannot be determined. The majority of CS cases are simplex. Perhaps 10%-50% of individuals with CS have an affected parent. If a parent of the proband has PHTS, the risk to sibs is 50%. Each child of an affected individual has a 50% chance of inheriting the mutation and developing PHTS. Prenatal testing is available.

Diagnosis

Clinical Diagnosis

A presumptive diagnosis of PHTS is based on clinical signs; by definition, however, the diagnosis of PHTS is made only when a PTEN mutation is identified.

Cowden syndrome. Consensus diagnostic criteria for Cowden syndrome have been developed [Eng 2000] and updated each year by the National Comprehensive Cancer Network (NCCN). Clinical criteria have been divided into three categories:

Pathognomonic criteria

  • Adult Lhermitte-Duclos disease (LDD), defined as the presence of a cerebellar dysplastic gangliocytoma [Zhou et al 2003a]

  • Mucocutaneous lesions:

    • Trichilemmomas (facial)

    • Acral keratoses

    • Papillomatous lesions

    • Mucosal lesions

Major criteria

  • Breast cancer

  • Thyroid cancer (non-medullary), especially follicular thyroid epithelial cancer

  • Macrocephaly (occipital frontal circumference ≥97th percentile)

  • Endometrial carcinoma

Minor criteria

  • Other thyroid lesions (e.g., adenoma, multinodular goiter)

  • Mental retardation (IQ ≤75)

  • Hamartomatous intestinal polyps

  • Fibrocystic disease of the breast

  • Lipomas

  • Fibromas

  • Genitourinary tumors (especially renal cell carcinoma)

  • Genitourinary malformation

  • Uterine fibroids

An operational diagnosis of Cowden syndrome is made if an individual meets any one of the following criteria:

  • Pathognomonic mucocutaneous lesions alone if there are:

    • Six or more facial papules, of which three or more must be trichilemmoma, or

    • Cutaneous facial papules and oral mucosal papillomatosis, or

    • Oral mucosal papillomatosis and acral keratoses, or

    • Six or more palmo-plantar keratoses

  • One of the following:

    • Two or more major criteria

    • One major and at least three minor criteria

    • At least four minor criteria

In a family in which one individual meets the diagnostic criteria for Cowden syndrome listed above, other relatives are considered to have a diagnosis of CS if they meet any of the following criteria:

  • The pathognomonic criteria OR

  • Any one major criterion with or without minor criteria OR

  • Two minor criteria OR

  • History of Bannayan-Riley-Ruvalcaba syndrome

Bannayan-Ruvalcaba-Riley syndrome. Diagnostic criteria for BRRS have not been set but are based heavily on the presence of the cardinal features of macrocephaly, hamartomatous intestinal polyposis, lipomas, and pigmented macules of the glans penis [Gorlin et al 1992, Jones 1997].

Proteus syndrome. Proteus syndrome (PS) is highly variable and appears to affect individuals in a mosaic distribution (i.e., only some organs/tissues are affected). Thus, it is frequently misdiagnosed despite the development of consensus diagnostic criteria [Biesecker et al 1999]. Mandatory general criteria for diagnosis include mosaic distribution of lesions, progressive course, and sporadic occurrence.

Additional specific criteria for diagnosis include:

  • Connective tissue nevi (pathognomonic)

OR two of the following:

  • Epidermal nevus

  • Disproportionate overgrowth (one or more)

    • Limbs (arms/legs, hands/feet/digits)

    • Skull (hyperostoses)

    • External auditory meatus (hyperostosis)

    • Vertebrae (megaspondylodysplasia)

    • Viscera (spleen/thymus)

  • Specific tumors before end of second decade (either one)

    • Bilateral ovarian cystadenomas

    • Parotid monomorphic adenoma

OR three of the following:

  • Dysregulated adipose tissue (either one)

    • Lipomas

    • Regional absence of fat

  • Vascular malformations (one or more)

    • Capillary malformation

    • Venous malformation

    • Lymphatic malformation

  • Facial phenotype

    • Dolichocephaly

    • Long face

    • Minor downslanting of palpebral fissures and/or minor ptosis

    • Low nasal bridge

    • Wide or anteverted nares

    • Open mouth at rest

Proteus-like syndrome. Proteus-like syndrome is undefined but refers to individuals with significant clinical features of PS but who do not meet the diagnostic criteria.

Testing

Pathologic review is essential in confirming the appropriate histopathology of the characteristic dermatologic, thyroid, breast, endometrial, and colonic lesions that can be seen with PHTS.

Molecular Genetic Testing

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Gene. PTEN is the only gene known to be associated with PTEN hamartoma tumor syndrome (PHTS).

Clinical uses

  • Confirmation of the diagnosis. Failure to detect a mutation does not exclude a clinical diagnosis of CS, BRRS, or Proteus/Proteus-like syndromes in an individual with significant signs associated with these disorders.

  • Predictive testing

  • Prenatal diagnosis

Clinical testing

  • Sequence analysis. Virtually all missense mutations in PTEN are believed to be deleterious [Eng, unpublished data]:

    • Approximately 85% of individuals who meet the diagnostic criteria for CS [Marsh et al 1998, Zhou et al 2003b] and 65% of individuals with a clinical diagnosis of BRRS [Marsh et al 1999, Zhou et al 2003b] have a detectable PTEN

      gene mutation.


      Note: To date, no individuals with CS have had large deletions.

    • Data suggest that up to 50% of individuals with a Proteus-like syndrome and up to 20% of individuals with Proteus syndrome have PTEN mutations [Zhou et al 2001a, Eng 2003].


      Note: (1) These observations were confirmed by Smith et al (2002), who found a germline PTEN mutation in a child with Proteus syndrome who met published diagnostic criteria. (2) In the Thiffault et al (2004) study, no PTEN mutations were detected in individuals with Proteus syndrome, potentially signaling the existence of other genes in this syndrome or the relative insensitivity of the mutation detection technique used.

Research testing

  • Deletion analysis. Southern blotting, monochromosomal hybrid analysis, real-time PCR, and semiquantitative multiplex PCR can each be used to detect PTEN

    deletions on a research basis only.


    Approximately 10% of individuals with BRRS who do not have a mutation detected in the PTEN coding sequence have large deletions within or encompassing PTEN [Zhou et al 2003b].

  • Promoter analysis. Direct sequencing of the promoter region detects mutations that alter the function of the gene in approximately 10% of individuals with CS who do not have an identifiable mutation in the PTEN coding region [Zhou et al 2003b].

Table 1 summarizes molecular genetic testing for this disorder.

Table 1. Molecular Genetic Testing Used in PTEN Hamartoma Tumor Syndrome

Test MethodGenetic MechanismMutation Detection Frequency by Test MethodTest Availability
Sequence analysisPTEN mutation80% with Cowden syndromeClinical
graphic element
60% with BRRS
50% with Proteus-like syndrome
20% with Proteus syndrome
Deletion analysisPTEN deletion~11% with BRRS 1Research only
Promoter analysisPTEN promoter mutations~10% with CS

1. Zhou et al 2003b

Interpretation of test results. For issues to consider in interpretation of sequence analysis results, click here.

Clinical Description

Natural History

The PTEN hamartoma tumor syndrome (PHTS) is characterized by hamartomatous tumors and germline PTEN mutations. Clinically, PHTS includes Cowden syndrome (CS), Bannayan-Riley-Ruvalcaba syndrome (BRRS), Proteus syndrome (PS), and Proteus-like syndrome. CS is a multiple hamartoma syndrome with a high risk of benign and malignant tumors of the thyroid, breast, and endometrium. BRRS is a congenital disorder characterized by macrocephaly, intestinal polyposis, lipomas, and pigmented macules of the glans penis. PS is a complex, highly variable disorder involving congenital malformations and overgrowth of multiple tissues. Proteus-like syndrome is undefined but refers to individuals with significant clinical features of PS who do not meet the diagnostic criteria for PS.

Cowden syndrome. Over 90% of individuals with CS have some clinical manifestation of the disorder by the late 20s [Nelen et al 1996, Eng 2000]. By the third decade, 99% of affected individuals develop the mucocutaneous stigmata, primarily trichilemmomas and papillomatous papules, as well as acral and plantar keratoses. In addition, individuals with Cowden syndrome usually have macrocephaly and dolicocephaly. Hamartomatous gastrointestinal polyps can be seen in CS but are usually minute and cause few symptoms. Based on anecdotal observations, glycogenic acanthosis in the presence of features of CS appears to be associated with a high likelihood of finding a PTEN mutation [Eng 2003, McGarrity et al 2003]:

  • Tumor risk. Individuals with CS have a high risk of breast, thyroid, and endometrial cancers. As with other hereditary cancer syndromes, the risk of multifocal and bilateral (in paired organs such as the breasts) cancer is increased.

  • Breast disease. Women with Cowden syndrome have as high as a 67% risk for benign breast disease. The lifetime risk to females of developing breast cancer is 25%-50%, with an average age of diagnosis between 38 and 46 years [Brownstein et al 1978, Starink et al 1986]. Breast cancer has been described in PTEN mutation-positive males [Fackenthal et al 2001].

  • Thyroid disease. Benign multinodular goiter of the thyroid as well as adenomatous nodules and follicular adenomas are common, occurring in up to 75% of individuals with CS [Harach et al 1999]. The lifetime risk for thyroid cancer (usually follicular, rarely papillary, but never medullary thyroid cancer) is around 10% [Eng 1997]. It is not clear if the age of diagnosis of thyroid cancer is earlier than in the general population.

  • Endometrial disease. Benign uterine fibroids are common. Risk for endometrial cancer, although not well defined, may approach 5%-10%.

  • Other

    • Skin cancers, renal cell carcinomas, and brain tumors as well as vascular malformations affecting any organ are occasionally seen in individuals with CS.


      Note: Because meningioma is so common in the general population, it is not yet clear if meningioma is a true manifestation of CS.


      A rare central nervous system tumor, cerebellar dyplastic gangliocytoma (Lhermitte-Duclos disease) is also found in CS and may be pathognomonic.

    • Although hamartomatous polyps may occur in the gastrointestinal tract, it is felt that the risk for colorectal cancer is not increased; unlike BRRS polyps, the polyps in CS are rarely symptomatic.

Bannayan-Ruvalcaba-Riley syndrome (BRRS). Common features of BRRS, in addition to those mentioned above, include high birth weight, developmental delay, and mental deficiency (50% of affected individuals), a myopathic process in proximal muscles (60%), joint hyperextensibility, pectus excavatum, and scoliosis (50%) [Gorlin et al 1992, Jones 1997]. Although cancer was initially not believed to be a component of the syndrome, individuals with BRRS and PTEN gene mutations are currently thought to have the same cancer risks as individuals with CS [Marsh et al 1999]. It is not clear whether these risks apply to individuals with BRRS without PTEN gene mutations. The gastrointestinal hamartomatous polyps in BRRS (seen in 45% of affected individuals) may occasionally be associated with intussusception, but rectal bleeding and oozing of "serum" is more common. These polyps are not believed to increase the risk for colorectal cancer. PHTS hamartomatous polyps are different in histomorphology from the polyps seen in Peutz-Jeghers syndrome.

Proteus syndrome (PS). Proteus syndrome is a complex disorder comprising malformations and hamartomatous overgrowth of multiple tissues, connective tissue nevi, epidermal nevi, and hyperostoses. The manifestations are commonly present at birth and persist or progress over postnatal life. Tumors or malignancies are not frequently reported in PS. However, certain unusual tumor types, such as cystadenoma of the ovary, various types of testicular tumors, central nervous system tumors, and parotid monomorphic adenomas, are occasionally associated with PS and therefore can be of diagnostic value when present. PS is uncommon; only approximately 120 affected individuals have been reported [Cohen 1999].

Proteus-like syndrome. Proteus-like syndrome is undefined but refers to individuals with significant clinical features of PS who do not meet the diagnostic criteria.

Genotype-Phenotype Correlations

For purposes of genotype-phenotype analyses, a series of 37 unrelated probands with CS were ascertained by the operational diagnostic criteria of the International Cowden Consortium, 1995 version [Nelen et al 1996, Eng 2000]. Association analyses revealed that families with CS and germline PTEN mutations are more likely to develop malignant breast disease when compared to families that do not have a PTEN mutation [Marsh et al 1998]. In addition, missense mutations and mutations 5' to or within the phosphatase core motif appeared to be associated with involvement of five or more organs, a surrogate phenotype for severity of disease [Marsh et al 1998].

The mutational spectra of BRRS and CS have been shown to overlap, thus lending formal proof that CS and BRRS are allelic [Marsh et al 1999]. No difference in mutation frequencies was observed between BRRS occurring in a single individual in a family and BRRS occurring in multiple family members. Over 90% of families with CS-BRRS overlap were found to have germline PTEN mutations. In addition, the presence of PTEN mutations in BRRS was found to be associated with the development of lipomas and tumors of the breast [Marsh et al 1999]. Therefore, individuals with BRRS and PTEN mutations may have increased cancer risks (despite the fact that this syndrome was previously not believed to be associated with malignancy).

An individual representing a simplex case (i.e., one with no known family history) of Proteus-like syndrome comprising hemihypertrophy, macrocephaly, lipomas, connective tissue nevi, and multiple arteriovenous malformations was found to have a germline R335X PTEN mutation and the same somatic mutation (R130X) in three separate tissues, possibly representing germline mosaicism [Zhou et al 2000]. Both these mutations have been previously described in classic CS and BRRS. Two of nine individuals (22%) with Proteus syndrome and three of six (50%) individuals with Proteus-like syndrome were found to have germline PTEN mutations [Zhou et al 2001a].

Penetrance

More than 90% of individuals with CS have some clinical manifestation of the disorder by the late 20s [Nelen et al 1996, Eng 2000]. By the third decade, 99% of affected individuals develop the mucocutaneous stigmata, primarily trichilemmomas and papillomatous papules, as well as acral and plantar keratoses.

Anticipation

Anticipation is not observed.

Nomenclature

Cowden syndrome, Cowden disease, and multiple hamartoma syndrome have been used interchangeably.

Bannayan-Riley-Ruvalcaba syndrome, Bannayan-Zonana syndrome, and Myhre-Riley-Smith syndrome refer to a similar constellation of signs that comprise what the authors refer to as BRRS. When a PTEN mutation is found, the gene-related name, PHTS, should be used.

Prevalence

Because the diagnosis of CS is difficult to establish, the true prevalence is unknown. The prevalence has been estimated at one in 200,000 [Nelen et al 1997, Nelen et al 1999]; this is likely an underestimate. Because of the variable and often subtle external manifestations of CS/BRRS, many individuals remain undiagnosed [Haibach et al 1992; Schrager et al 1998; Eng, unpublished].

Differential Diagnosis

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

The primary differential diagnoses to consider are other hamartoma syndromes, including juvenile polyposis syndrome (JPS) and Peutz-Jeghers syndrome (PJS), both inherited in an autosomal dominant manner.

Juvenile polyposis syndrome (JPS) is characterized by predisposition for hamartomatous polyps in the gastrointestinal tract, specifically in the stomach, small intestine, colon, and rectum. The term "juvenile" refers to the type of polyp, not the age of onset of polyps. Juvenile polyps are hamartomas that show a normal epithelium with a dense stroma, an inflammatory infiltrate, and a smooth surface with dilated, mucus-filled cystic glands in the lamina propria.

Most individuals with JPS have some polyps by age 20 years. Some individuals may have only four or five polyps over their lifetimes, whereas others in the same family may have more than one hundred. If the polyps are left untreated, they may cause bleeding and anemia. Most juvenile polyps are benign; however, malignant transformation can occur.

Approximately 20% of individuals with JPS have mutations in the MADH4 gene; approximately 20% of individuals with JPS have mutations in the BMPR1A gene [Howe et al 1998, Howe et al 2001]:

Peutz-Jeghers syndrome (PJS) is characterized by the association of gastrointestinal polyposis and mucocutaneous pigmentation. PJS-type hamartomatous polyps are most prevalent in the small intestine, but also occur in the stomach and large bowel in the majority of affected individuals. The Peutz-Jeghers polyp has a diagnostic appearance and is quite different from the hamartomatous polyps seen in CS or JPS. Clinically, Peutz-Jeghers polyps are often symptomatic (intussusception, rectal bleeding), whereas CS polyps are rarely so.

The pigmentation of the peri-oral region is pathognomonic, particularly if it crosses the vermilion border [Eng & Blackstone 1988, Rustgi 1994]. Hyperpigmented macules on the fingers are also common.

Molecular genetic testing of STK11/LKB1 reveals disease-causing mutations in approximately 70% of individuals who have a positive family history and 20%-70% of individuals who have no family history of PJS.

Other less likely differential diagnoses to consider for PHTS include the following:

Management

Treatment of Manifestations

The mucocutaneous manifestations of Cowden syndrome are rarely life threatening:

  • If asymptomatic, observation alone is prudent.

  • When symptomatic, topical agents (e.g., 5-fluorouracil), curettage, cryosurgery, or laser ablation may provide only temporary relief [Hildenbrand et al 2001]. Surgical excision is sometimes complicated by cheloid formation and recurrence (often rapid) of the lesions [Eng, unpublished data].

Treatment for the benign and malignant manifestations of PHTS is the same as for their sporadic counterparts.

Prevention of Primary Manifestations

Some women at increased risk for breast cancer consider prophylactic mastectomy, especially if breast tissue is dense or if repeated breast biopsies have been necessary. Prophylactic mastectomy reduces the risk of breast cancer by 90% women at high risk [Hartmann et al 1999]. Note: the recommendation of prophylactic mastectomy is a generalization for women at increased risk of breast cancer from a variety of causes, not just from PTHS.

There is no direct evidence to support the routine use of agents such as tamoxifen or raloxifene in individuals with PHTS to reduce the risk of developing breast cancer. Physicians should discuss the limitations of the evidence and the risks and benefits of chemoprophylaxis with each individual. In addition, the clinician must discuss the increased risk of endometrial cancer associated with tamoxifen use in a population already at increased risk for endometrial cancer.

Surveillance

The most serious consequences of PHTS relate to the increased risk of cancers including breast, thyroid, endometrial, and to a lesser extent, renal. In this regard, the most important aspect of management of any individual with a PTEN mutation is increased cancer surveillance.

Cowden Syndrome

General

  • Annual comprehensive physical examination starting at age 18 years (or five years before the youngest component cancer diagnosis in the family), with attention paid to skin changes and the neck region

  • Consider annual dermatologic examination

  • Annual urinalysis. Consider annual cytololgy and renal ultrasound examination if the family history is positive for renal cell carcinoma

  • Baseline colonoscopy at age 50 years (unless symptoms arise earlier). If only hamartomas are found, the American Cancer Society guidelines for colon cancer screening (i.e., annual fecal occult blood testing and sigmoidoscopy every five years or colonoscopy every ten years) should be followed.

Breast cancer

  • Women [Eng 2000, National Comprehensive Cancer Network 2006]

    • Monthly breast self-examination beginning at age 18 years

    • Annual clinical breast examinations beginning at age 25 years or 5-10 years earlier than earliest known breast cancer diagnosis in the family (whichever is earliest)

    • Annual mammography and breast MRI beginning at age 30-35 years or 5-10 years before the earliest known breast cancer diagnosis in the family (whichever is earliest)

  • Men. Monthly breast self-examination

Thyroid cancer

  • Baseline thyroid ultrasound examination at age 18 years

  • Consider annual thyroid ultrasound examination thereafter

Endometrial cancer

  • Premenopausal women. Annual blind repel (suction) biopsies beginning at age 35-40 years (or five years before the youngest endometrial cancer diagnosis in the family)

  • Postmenopausal women. Annual transvaginal ultrasound examination with biopsy of suspicious areas

Bannayan-Ruvalcaba-Riley Syndrome

Screening recommendations have not been established for BRRS. Given recent molecular epidemiologic studies, however, individuals with BRRS and a germline PTEN mutation should undergo the same surveillance as individuals with CS.

Individuals with BRRS should also be monitored for complications related to gastrointestinal hamartomatous polyposis, which can be more severe than in CS.

Proteus Syndrome/Proteus-Like Syndrome

Although the observation of germline PTEN mutations in Proteus and Proteus-like syndromes is relatively new, clinicians should consider instituting the CS surveillance recommendations for individuals with these disorders who have germline PTEN mutations.

Agents/Circumstances to Avoid

Because of the propensity for rapid tissue regrowth and the propensity to form cheloid tissue, it is recommended that cutaneous lesions be excised only if malignancy is suspected or symptoms (e.g., pain, deformity) are significant.

Testing of Relatives at Risk

When a PTEN mutation has been identified in a proband, testing of asymptomatic at-risk relatives can identify those who have the family-specific mutation and, therefore, have PHTS. These individuals are in need of ongoing surveillance, as discussed above.

Molecular testing is appropriate for at-risk children, given the possible early disease presentation in individuals with BRRS and Proteus syndrome.

Relatives who have not inherited the PTEN mutation found in an affected relative do not have PHTS or its associated cancer risks.

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

Therapies Under Investigation

Although mTOR inhibitors show promise for treatment of malignancies in individuals who have a germline PTEN mutation, use should be limited to clinical trials. At this time no clinical trials are specifically directed at individuals with PHTS.

Search ClinicalTrials.gov for access to information on clinical studies for a wide range of diseases and conditions.

Other

Genetics clinics are a source of information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

Support groups have been established for individuals and families to provide information, support, and contact with other affected individuals. The Resources section may include disease-specific and/or umbrella support organizations.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, 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. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

Mode of Inheritance

PHTS is inherited in an autosomal dominant manner.

Risk to Family Members

Parents of a proband

  • Because Cowden syndrome is likely underdiagnosed, the actual proportion of simplex cases (defined as individuals with no obvious family history) and familial cases (defined as two or more related affected individuals) cannot be determined.

  • From the literature and the experience of both major US Cowden syndrome centers, the majority of individuals with CS have no obvious family history. As a broad estimate, perhaps 10%-50% of individuals with Cowden syndrome have an affected parent [Marsh et al 1999].

  • The majority of evidence suggests that PTEN mutations occur in both simplex and familial occurrences of BRRS [Eng 2003].

  • If a PTEN mutation is identified in the proband, the parents should be offered molecular genetic testing to determine if one of them has previously unidentified PHTS. If no mutation is identified in the proband, both parents should undergo thorough clinical examination to help determine if either parent has signs of PHTS.

Note: Although some individuals diagnosed with PHTS have an affected parent, the family history may appear to be negative because of failure to recognize the disorder in family members, early death of the parent before the onset of symptoms, or late onset of the disease in the affected parent.

Sibs of a proband

  • The risk to the sibs of the proband depends on the genetic status of the parents.

  • If a parent of the proband has PHTS, the risk to sibs is 50%.

  • If it has been shown that neither parent has the PTEN mutation found in the proband, the risk to sibs is probably negligible, since germline mosaicism has not been reported in PHTS.

  • If a mutation cannot be identified in the proband, PHTS can be excluded on clinical grounds. Normal clinical examinations in parents in their thirties, done looking specifically for signs of CS/BRRS, would make the risk to sibs of the proband minimal, since an estimated 99% of affected individuals would have signs by that age.

Offspring of a proband. Each child of an affected individual has a 50% chance of inheriting the mutation and developing PHTS.

Other family members of a proband. The risk to other family members depends on the genetic status of the proband's parents. If a parent is found to be affected, his or her family members are at-risk.

Related Genetic Counseling Issues

Testing of at-risk relatives. When a mutation has been identified in a proband, testing of asymptomatic at-risk relatives can identify those who also have the mutation and have PHTS. These individuals are in need of ongoing surveillance, as discussed above. Molecular testing is appropriate for at-risk individuals younger than age 18 years, given the possible early disease presentation in individuals with BRRS and Proteus syndrome.

Considerations in families with an apparent de novo mutation. When neither parent of a proband with an autosomal dominant condition has the disease-causing mutation or clinical evidence of the disorder, it is likely that the proband has a de novo mutation. However, possible non-medical explanations, including alternate paternity or undisclosed adoption, could also be explored.

Genetic cancer risk assessment and counseling. For comprehensive descriptions of the medical, psychosocial, and ethical ramifications of identifying at-risk individuals through cancer risk assessment with or without molecular genetic testing, see:

Family planning. The optimal time for determination of genetic risk and discussion of the availability of prenatal testing is before pregnancy.

DNA banking. DNA banking is the storage of DNA (typically extracted from white blood cells) for possible future use. Because it is likely that testing methodology and our understanding of genes, mutations, and diseases will improve in the future, consideration should be given to banking DNA of affected individuals. DNA banking is particularly relevant when the sensitivity of currently available testing is less than 100%. See graphic element for a list of laboratories offering DNA banking.

Prenatal Testing

Prenatal diagnosis for pregnancies at increased risk is possible by analysis of DNA extracted from fetal cells obtained by amniocentesis usually performed at approximately 15-18 weeks' gestation or chorionic villus sampling (CVS) at approximately ten to 12 weeks' gestation. The disease-causing allele of an affected family member must be identified before prenatal testing can be performed.

Note: Gestational age is expressed as menstrual weeks calculated either from the first day of the last normal menstrual period or by ultrasound measurements.

Preimplantation genetic diagnosis (PGD). Although successful PGD for PHTS has not been reported in the medical literature, PGD may be available for families in which the disease-causing mutation has been identified in an affected family member in a research or clinical laboratory. For laboratories offering PGD, see graphic element.

Molecular Genetics

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Table A. Molecular Genetics of PTEN Hamartoma Tumor Syndrome

Gene SymbolChromosomal LocusProtein Name
PTEN10q23.3Phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN

Data are compiled from the following standard references: Gene symbol from HUGO; chromosomal locus, locus name, critical region, complementation group from OMIM; protein name from Swiss-Prot.

Table B. OMIM Entries for PTEN Hamartoma Tumor Syndrome

 153480 BANNAYAN-RILEY-RUVALCABA SYNDROME; BRRS
 158350 COWDEN DISEASE; CD
 176920 PROTEUS SYNDROME
 601728 PHOSPHATASE AND TENSIN HOMOLOG; PTEN

Table C. Genomic Databases for PTEN Hamartoma Tumor Syndrome

Gene SymbolEntrez GeneHGMD
PTEN5728 (MIM No. 601728)PTEN

For a description of the genomic databases listed, click here.

Note: HGMD requires registration.

Molecular Genetic Pathogenesis

The complete function of PTEN is not yet fully understood. PTEN belongs to a sub-class of phosphatases called dual-specificity phosphatases that remove phosphate groups from tyrosine as well as serine and threonine. In addition, PTEN is the major phosphatase for phosphoinositide-3,4,5-triphosphate, and thus downregulates the PI3K/Akt pathway.

Somatic PTEN mutations and loss of gene expression are frequently found in both endometrioid endometrial adenocarcinoma and precancerous endometrial lesions (intraepithelial neoplasia), confirming the critical role that PTEN must play in endometrial tissues [Mutter et al 2000].

Normal allelic variants: The gene comprises nine exons and likely spans a genomic distance of more than 120 kb. The 1209-bp coding sequence is predicted to encode a 403-amino acid protein.

Pathologic allelic variants: Germline mutations have been found throughout PTEN (with the exception of exon 9) and include missense and nonsense mutations, splice site mutations, small deletions, insertions, and several large deletions. More than 150 unique mutations are currently listed in the Human Gene Mutation Database (see Genomic Databases table). Nearly 40% of mutations are found in exon 5, which encodes the phosphate core motif [Eng 2003]. Most mutations are unique, although a number of recurrent mutations have been reported, particularly R130X, R233X, and R335X [Bonneau & Longy 2000]. Approximately 10% of individuals with CS who do not have a mutation detected in the PTEN coding sequence have heterozygous germline mutations in the PTEN promoter [Zhou et al 2003b]. In contrast, 10% of individuals with BRRS who do not have an identifiable PTEN mutation on sequence analysis have large deletions within or encompassing PTEN [Zhou et al 2003b].

Normal gene product: PTEN encodes an almost ubiquitously expressed dual specificity phosphatase. The PTEN protein localizes to specific nuclear and cytoplasmic components. The wild-type protein is a major lipid phosphatase that downregulates the PI3K/Akt pathway to cause G1 arrest and apoptosis. In addition, the protein phosphatase appears to play an important role in inhibition of cell migration and spreading, as well as downregulating several cell cyclins [Eng 2003]. It appears that nuclear PTEN mediates cell cycle arrest, while cytoplasmic PTEN is required for apoptosis [Chung & Eng 2005].

Abnormal gene product: The majority (76%) of germline mutations in PTEN result in either truncated protein, lack of protein (haploinsufficiency), or dysfunctional protein. Many missense mutations are functionally null and several act as dominant negatives. When PTEN is absent, decreased, or dysfunctional, phosphorylation of Akt is uninhibited, leading to the inability to activate cell cycle arrest and/or to undergo apoptosis. In addition, through lack of protein phosphatase activity, the mitogen-activated protein kinase (MAPK) pathway is dysregulated, leading to abnormal cell survival [Eng 2003].

Resources

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

Cowdens Syndrome & Bannayan-Riley-Ruvalcaba Syndrome Foundation
1394 Wedgewood Drive
Salne MI 48176
Phone: 734-944-8313
Email: Rosalita@comcast.net
Cowden Syndrome

Genetics of Breast and Ovarian Cancer (PDQ)
A service of the National Cancer Institute
Cowden syndrome

National Library of Medicine Genetics Home Reference
Cowden syndrome

American Cancer Society
Provides contact information for regional support.
1599 Clifton Road NE
Atlanta GA 30322
Phone: 800-227-2345
www.cancer.org

CancerCare
275 Seventh Avenue Floor 22
New York NY 10001
Phone: 800-813-HOPE (800-813-4673); 212-712-8400
Fax: 212-712-8495
Email: info@cancercare.org
www.cancercare.org

National Alliance of Breast Cancer Organizations
An advocacy group that serves as an umbrella for 370 breast cancer groups nationwide. Provides information, a newsletter, and treatment information. Also provides grants for programs on early detection and education.
9 East 37th Street 10th Floor
New York NY 10016
Phone: 888-806-2226; 212-889-0606
Fax: 212-689-1213
Email: nbcamquestions@yahoo.com
www.nbcam.org

The National Breast Cancer Coalition/Fund
An advocacy group seeking public policy change to benefit breast cancer patients and survivors.
1101 17th Street Northwest Suite 1300
Washington DC 20036
Phone: 800-622-2838 (toll-free); 202-296-7477
Fax: 202-265-6854
www.stopbreastcancer.org

The National Coalition for Cancer Survivorship
A consumer organization that advocates on behalf of all people with cancer.
1010 Wayne Avenue Suite 770
Silver Spring MD 20910
Phone: 888-650-9127 (toll-free); 301-650-9127
Fax: 301-565-9670
Email: info@canceradvocacy.org
www.canceradvocacy.org

The Susan G. Komen Breast Cancer Foundation
Information, referrals to treatment centers. Answers questions from recently diagnosed women and provides emotional support. Funds research programs for women who do not have adequate medical service and support.
5005 LBJ Freeway Suite 250
Dallas TX 75244
Phone: 877-465-6636 (toll-free); 972-855-1600
Fax: 972-855-1605
Email: helpline@komen.org
http://cms.komen.org/komen/index.htm

Y-Me National Breast Cancer Organization
Hotline staffed by counselors and volunteers who have had breast cancer. Information, referrals, support.
212 West Van Buren Street Suite 1000
Chicago IL 60607
Phone: 800-221-2141; 312-986-8338
Fax: 312-294-8597
www.y-me.org

References

Medical Genetic Searches: A specialized PubMed search designed for clinicians that is located on the PubMed Clinical Queries page. graphic element

Literature Cited

Biesecker LG, Happle R, Mulliken JB, Weksberg R, Graham JM, Viljoen DL, Cohen MM. Proteus syndrome: diagnostic criteria, differential diagnosis, and patient evaluation. Am J Med Genet. 1999; 84: 38995. [PubMed]
Bonneau D, Longy M. Mutations of the human PTEN gene. Hum Mutat. 2000; 16: 10922. [PubMed]
Brownstein MH, Wolf M, Bikowski JB. Cowden's disease: a cutaneous marker of breast cancer. Cancer. 1978; 41: 23938. [PubMed]
Butler MG, Dasouki MJ, Zhou XP, Talebizadeh Z, Brown M, Takahashi TN, Miles JH, Wang CH, Stratton R, Pilarski R, Eng C. Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations. J Med Genet. 2005; 42: 31821. [PubMed]
Chung JH, Eng C. Nuclear-cytoplasmic partitioning of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) differentially regulates the cell cycle and apoptosis. Cancer Res. 2005; 65: 8096100. [PubMed]
Cohen MM. Overgrowth syndromes: an update. Adv Pediatr. 1999; 46: 44191. [PubMed]
Dasouki MJ, Ishmael H, Eng C. Macrocephaly, macrosomia and autistic behavior due to a de novo PTEN germline mutation. Am J Hum Genet. 2001; 69: A564.
Eng C. Cowden syndrome. J Genet Couns. 1997; 6: 181.
Eng C. Will the real Cowden syndrome please stand up: revised diagnostic criteria. J Med Genet. 2000; 37: 82830. [PubMed]
Eng C. PTEN: one gene, many syndromes. Hum Mutat. 2003; 22: 18398. [PubMed]
Eng C, Blackstone MO. Peutz-Jeghers syndrome. Med Rounds. 1988; 1: 165.
Fackenthal JD, Marsh DJ, Richardson AL, Cummings SA, Eng C, Robinson BG, Olopade OI. Male breast cancer in Cowden syndrome patients with germline PTEN mutations. J Med Genet. 2001; 38: 15964. [PubMed]
Goffin A, Hoefsloot LH, Bosgoed E, Swillen A, Fryns JP. PTEN mutation in a family with Cowden syndrome and autism. Am J Med Genet. 2001; 105: 5214. [PubMed]
Gorlin RJ, Cohen MM, Condon LM, Burke BA. Bannayan-Riley-Ruvalcaba syndrome. Am J Med Genet. 1992; 44: 30714. [PubMed]
Haibach H, Burns TW, Carlson HE, Burman KD, Deftos LJ. Multiple hamartoma syndrome (Cowden's disease) associated with renal cell carcinoma and primary neuroendocrine carcinoma of the skin (Merkel cell carcinoma). Am J Clin Pathol. 1992; 97: 70512. [PubMed]
Harach HR, Soubeyran I, Brown A, Bonneau D, Longy M. Thyroid pathologic findings in patients with Cowden disease. Ann Diagn Pathol. 1999; 3: 33140. [PubMed]
Hartmann LC, Schaid DJ, Woods JE, Crotty TP, Myers JL, Arnold PG, Petty PM, Sellers TA, Johnson JL, McDonnell SK, Frost MH, Jenkins RB. Efficacy of bilateral prophylactic mastectomy in women with a family history of breast cancer. N Engl J Med. 1999; 340: 7784. [PubMed]
Hildenbrand C, Burgdorf WH, Lautenschlager S. Cowden syndrome-diagnostic skin signs. Dermatology. 2001; 202: 3626. [PubMed]
Howe JR, Bair JL, Sayed MG, Anderson ME, Mitros FA, Petersen GM, Velculescu VE, Traverso G, Vogelstein B. Germline mutations of the gene encoding bone morphogenetic protein receptor 1A in juvenile polyposis. Nat Genet. 2001; 28: 1847. [PubMed]
Howe JR, Roth S, Ringold JC, Summers RW, Jarvinen HJ, Sistonen P, Tomlinson IP, Houlston RS, Bevan S, Mitros FA, Stone EM, Aaltonen LA. Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science. 1998; 280: 10868. [PubMed]
Huang SC, Chen CR, Lavine JE, Taylor SF, Newbury RO, Pham TT, Ricciardiello L, Carethers JM. Genetic heterogeneity in familial juvenile polyposis. Cancer Res. 2000; 60: 68825. [PubMed]
Jones KL (1997) Smith's Recognizable Patterns of Human Malformation, 5 ed. WB Saunders, Philadelphia.
Kurose K, Araki T, Matsunaka T, Takada Y, Emi M. Variant manifestation of Cowden disease in Japan: hamartomatous polyposis of the digestive tract with mutation of the PTEN gene. Am J Hum Genet. 1999; 64: 30810. [PubMed]
Marsh DJ, Coulon V, Lunetta KL, Rocca-Serra P, Dahia PL, Zheng Z, Liaw D, Caron S, Duboué B, Lin AY, Richardson AL, Bonnetblanc JM, Bressieux JM, Cabarrot-Moreau A, Chompret A, Demange L, Eeles RA, Yahanda AM, Fearon ER, Fricker JP, Gorlin RJ, Hodgson SV, Huson S, Lacombe D, Eng C. et al. Mutation spectrum and genotype-phenotype analyses in Cowden disease and Bannayan-Zonana syndrome, two hamartoma syndromes with germline PTEN mutation. Hum Mol Genet. 1998; 7: 50715. [PubMed]
Marsh DJ, Kum JB, Lunetta KL, Bennett MJ, Gorlin RJ, Ahmed SF, Bodurtha J, Crowe C, Curtis MA, Dasouki M, Dunn T, Feit H, Geraghty MT, Graham JM, Hodgson SV, Hunter A, Korf BR, Manchester D, Miesfeldt S, Murday VA, Nathanson KL, Parisi M, Pober B, Romano C, Eng C. et al. PTEN mutation spectrum and genotype-phenotype correlations in Bannayan- Riley-Ruvalcaba syndrome suggest a single entity with Cowden syndrome. Hum Mol Genet. 1999; 8: 146172. [PubMed]
McGarrity TJ, Wagner Baker MJ, Ruggiero FM, Thiboutot DM, Hampel H, Zhou XP, Eng C. GI polyposis and glycogenic acanthosis of the esophagus associated with PTEN mutation positive Cowden syndrome in the absence of cutaneous manifestations. Am J Gastroenterol. 2003; 98: 142934. [PubMed]
Mutter GL, Lin MC, Fitzgerald JT, Kum JB, Baak JP, Lees JA, Weng LP, Eng C. Altered PTEN expression as a diagnostic marker for the earliest endometrial precancers. J Natl Cancer Inst. 2000; 92: 92430. [PubMed]
National Comprehensive Cancer Network (2006) Genetic/Familial High-Risk Assessment: Breast and Ovarian. Clinical Practice Guidelines in Oncology. Version 1.2008 (pdf).
Nelen MR, Kremer H, Konings IB, Schoute F, van Essen AJ, Koch R, Woods CG, Fryns JP, Hamel B, Hoefsloot LH, Peeters EA, Padberg GW. Novel PTEN mutations in patients with Cowden disease: absence of clear genotype-phenotype correlations. Eur J Hum Genet. 1999; 7: 26773. [PubMed]
Nelen MR, Padberg GW, Peeters EA, Lin AY, van den Helm B, Frants RR, Coulon V, Goldstein AM, van Reen MM, Easton DF, Eeles RA, Hodgsen S, Mulvihill JJ, Murday VA, Tucker MA, Mariman EC, Starink TM, Ponder BA, Ropers HH, Kremer H, Longy M, Eng C. Localization of the gene for Cowden disease to chromosome 10q22-23. Nat Genet. 1996; 13: 1146. [PubMed]
Nelen MR, van Staveren WC, Peeters EA, Hassel MB, Gorlin RJ, Hamm H, Lindboe CF, Fryns JP, Sijmons RH, Woods DG, Mariman EC, Padberg GW, Kremer H. Germline mutations in the PTEN/MMAC1 gene in patients with Cowden disease. Hum Mol Genet. 1997; 6: 13837. [PubMed]
Olschwang S, Serova-Sinilnikova OM, Lenoir GM, Thomas G. PTEN germ-line mutations in juvenile polyposis coli. Nat Genet. 1998; 18: 124. [PubMed]
Rustgi AK. Hereditary gastrointestinal polyposis and nonpolyposis syndromes. N Engl J Med. 1994; 331: 1694702. [PubMed]
Schrager CA, Schneider D, Gruener AC, Tsou HC, Peacocke M. Clinical and pathological features of breast disease in Cowden's syndrome: an underrecognized syndrome with an increased risk of breast cancer. Hum Pathol. 1998; 29: 4753. [PubMed]
Smith JM, Kirk EP, Theodosopoulos G, Marshall GM, Walker J, Rogers M, Field M, Brereton JJ, Marsh DJ. Germline mutation of the tumour suppressor PTEN in Proteus syndrome. J Med Genet. 2002; 39: 93740. [PubMed]
Starink TM, van der Veen JP, Arwert F, de Waal LP, de Lange GG, Gille JJ, Eriksson AW. The Cowden syndrome: a clinical and genetic study in 21 patients. Clin Genet. 1986; 29: 22233. [PubMed]
Thiffault I, Schwartz CE, Der Kaloustian V, Foulkes WD. Mutation analysis of the tumor suppressor PTEN and the glypican 3 (GPC3) gene in patients diagnosed with Proteus syndrome. Am J Med Genet A. 2004; 130A: 1237. [PubMed]
Zhou X, Hampel H, Thiele H, Gorlin RJ, Hennekam RC, Parisi M, Winter RM, Eng C. Association of germline mutation in the PTEN tumour suppressor gene and Proteus and Proteus-like syndromes. Lancet. 2001a; 358: 2101. [PubMed]
Zhou XP, Marsh DJ, Hampel H, Mulliken JB, Gimm O, Eng C. Germline and germline mosaic PTEN mutations associated with a Proteus- like syndrome of hemihypertrophy, lower limb asymmetry, arteriovenous malformations and lipomatosis. Hum Mol Genet. 2000; 9: 7658. [PubMed]
Zhou XP, Marsh DJ, Morrison CD, Chaudhury AR, Maxwell M, Reifenberger G, Eng C. Germline Inactivation of PTEN and Dysregulation of the Phosphoinositol-3-Kinase/Akt Pathway Cause Human Lhermitte-Duclos Disease in Adults. Am J Hum Genet. 2003a; 73: 11911198. [PubMed]
Zhou XP, Waite KA, Pilarski R, Hampel H, Fernandez MJ, Bos C, Dasouki M, Feldman GL, Greenberg LA, Ivanovich J, Matloff E, Patterson A, Pierpont ME, Russo D, Nassif NT, Eng C. Germline PTEN promoter mutations and deletions in Cowden/Bannayan-Riley-Ruvalcaba syndrome result in aberrant PTEN protein and dysregulation of the phosphoinositol-3-kinase/Akt pathway. Am J Hum Genet. 2003b; 73: 40411. [PubMed]
Zhou XP, Woodford-Richens K, Lehtonen R, Kurose K, Aldred M, Hampel H, Launonen V, Virta S, Pilarski R, Salovaara R, Bodmer WF, Conrad BA, Dunlop M, Hodgson SV, Iwama T, Jarvinen H, Kellokumpu I, Kim JC, Leggett B, Markie D, Mecklin JP, Neale K, Phillips R, Piris J, Rozen P, Houlston RS, Aaltonen LA, Tomlinson IP, Eng C. Germline mutations in BMPR1A/ALK3 cause a subset of cases of juvenile polyposis syndrome and of Cowden and Bannayan-Riley-Ruvalcaba syndromes. Am J Hum Genet. 2001b; 69: 70411. [PubMed]

Published Statements and Policies Regarding Genetic Testing

American Society of Clinical Oncology (2003) Statement on genetic testing for cancer susceptibility.

Suggested Reading

Scriver CR, Beaudet AL, Sly WS, Valle D, Childs B, Kinzler KW, Vogelstein B (2001) The Metabolic and Molecular Basis of Inherited Disease, 8 ed. McGraw-Hill, New York.
Vogelstein B, Kinzler KW (2001) The Genetic Basis of Human Cancer. McGraw-Hill, New York.

Chapter Notes

Author Notes

Dr. Eng is the director of the International Cowden Syndrome Consortium and a primary researcher in the field of PTEN-related disorders. Dr. Zbuk is a clinical cancer genetics fellow and Ms Stein is a genetic counselor in Cleveland Clinic's Genomic Medicine Institute, which is directed by Dr. Eng. The Cleveland Clinic Genomic Medicine Institute program features the only Cowden Syndrome center in the US, with ongoing clinical and molecular research protocols in PHTS.

Author History

Charis Eng, MD, PhD (2001-present)
Heather Hampel, MS; Ohio State University (2001-2006)
Robert Pilarski, MS; Ohio State University (2001-2006)
Jennifer L Stein, MS, CGC (2006-present)
Kevin M Zbuk, MD (2006-present)

Revision History

  • 10 January 2006 (me) Comprehensive update posted to live Web site

  • 19 May 2004 (ce) Revision: Genetic Counseling posted to live Web site

  • 17 December 2003 (me) Comprehensive update posted to live Web site

  • 23 May 2003 (ce) Revision: Differential Diagnosis

  • 29 November 2001 (me) Review posted to live Web site

  • 10 July 2001 (ce) Original submission

 

Next
GeneReviews2009
(navigation arrows) Go to previous chapter Go to next chapter Go to top of this page Go to bottom of this page Go to Table of Contents