Bookshelf » GeneReviews » APC-Associated Polyposis Conditions
 
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.

APC-Associated Polyposis Conditions
[Includes: Familial Adenomatous Polyposis (Familial Polyposis Coli), Gardner Syndrome, Turcot Syndrome, Attenuated FAP]

Randall W Burt, MD
Huntsman Cancer Institute
Salt Lake City
Kory W Jasperson, MS
Huntsman Cancer Institute
Salt Lake City
24072008fap
Initial Posting: December 18, 1998.
Last Update: July 24, 2008.

*

*

*

Summary

Disease characteristics. APC-associated polyposis conditions include: familial adenomatous polyposis (FAP), attenuated FAP, Gardner syndrome, and Turcot syndrome. FAP is a colon cancer predisposition syndrome in which hundreds to thousands of precancerous colonic polyps develop, beginning, on average, at age 16 years (range 7-36 years). By age 35 years, 95% of individuals with FAP have polyps; without colectomy, colon cancer is inevitable. The mean age of colon cancer diagnosis in untreated individuals is 39 years (range 34-43 years). Extracolonic manifestations are variably present and include: polyps of the gastric fundus and duodenum, osteomas, dental anomalies, congenital hypertrophy of the retinal pigment epithelium (CHRPE), soft tissue tumors, desmoid tumors, and associated cancers. Attenuated FAP is characterized by a significant risk for colon cancer but fewer colonic polyps (average of 30), more proximally located polyps, and diagnosis of colon cancer at a later age; management may be substantially different. Gardner syndrome is characterized by colonic polyposis typical of FAP together with osteomas and soft tissue tumors. Turcot syndrome is the association of colonic polyposis and central nervous system (CNS) tumors. Differences in phenotype may relate to the location of the mutation within the APC gene.

Diagnosis/testing. APC-associated polyposis conditions are caused by mutations in the APC gene. The diagnosis relies primarily on clinical findings. Clinically available molecular genetic testing of APC detects disease-causing mutations in up to 90% of probands with typical FAP. Molecular genetic testing is most often used in the early diagnosis of at-risk family members, as well as in confirming the diagnosis of FAP or attenuated FAP in individuals with equivocal findings (e.g., <100 adenomatous polyps).

Management. Treatment of manifestations: Colectomy is advised when more than 20 or 30 adenomas or multiple adenomas with advanced histology have occurred. Nonsteroidal anti-inflammatory drugs (NSAIDs), especially sulindac, have caused regression of adenomas in FAP and decreased the number of polyps requiring ablation in the remaining rectum of persons with a subtotal colectomy. Endoscopic or surgical removal of duodenal adenomas is considered if polyps exhibit villous change or severe dysplasia, exceed one centimeter in diameter, or cause symptoms. Osteomas may be removed for cosmetic reasons. Desmoid tumors may be surgically excised or treated with NSAIDs, anti-estrogens, cytotoxic chemotherapy, or radiation. Surveillance: screening for hepatoblastoma by liver ultrasound and measurement of serum alpha-fetoprotein concentration (until age five years); sigmoidoscopy or colonoscopy beginning at age ten to 12 years; annual colonoscopy once polyps are detected until colectomy; esophagogastroduodenoscopy by age 25 years or prior to colon surgery; small bowel x-ray or CT when duodenal adenomas are detected; and regular physical examinations including thyroid palpation. Testing of relatives at risk: Molecular genetic testing for early identification of at-risk family members improves diagnostic certainty and reduces the need for costly screening procedures in those at-risk family members who have not inherited the disease-causing mutation.

Genetic counseling. APC-associated polyposis conditions are inherited in an autosomal dominant manner. Approximately 75%-80% of individuals with APC-associated polyposis conditions have an affected parent. Offspring of an affected individual have a 50% risk of inheriting the altered APC gene. Prenatal testing and preimplantation genetic diagnosis are possible if a disease-causing mutation is identified in an affected family member.

Diagnosis

Clinical Diagnosis

The APC-associated polyposis conditions include: (1) the overlapping, often indistinguishable phenotypes of familial adenomatous polyposis (FAP), Gardner syndrome, and Turcot syndrome and (2) attenuated FAP, which has a lower colonic polyp burden and lower cancer risk:

Familial adenomatous polyposis (FAP) is diagnosed clinically in an individual with one of the following:

  • One hundred or more colorectal adenomatous polyps

    Note: The diagnosis of FAP is generally considered in individuals with polyposis occurring before age 40 years.

  • Fewer than 100 adenomatous polyps and a relative with FAP

Gardner syndrome is the association of colonic adenomatous polyposis, osteomas, and soft tissue tumors (epidermoid cysts, fibromas, desmoid tumors) [Gardner & Richards 1953].

Turcot syndrome is the association of colonic adenomatous polyposis and CNS tumors, usually medulloblastoma.

Attenuated FAP (AFAP) is considered in an individual with one of the following:

  • Ten to 99 colonic adenomatous polyps.

    Note: Individuals with 100 or more polyps occurring at “advanced” ages (35 to 40 years or older) may be found to have attenuated FAP.

  • A personal history of colorectal cancer before age 60 years and a family history of multiple adenomatous polyps

Currently, there is a lack of consensus regarding the exact diagnostic criteria that should be used for attenuated FAP. Nielsen et al [2007b) propose the following diagnostic criteria for attenuated FAP:

  • No family member with more than 100 polyps before age 30 years

    AND

  • At least two individuals with 10 to 99 adenomas diagnosed after age 30 years

    OR

  • One individual with 10 to 99 adenomas diagnosed after age 30 years and a first-degree relative with colorectal cancer with a few adenomas

Note: (1) This proposed definition takes into account the variability in colonic phenotype seen in attenuated FAP (i.e., some individuals may have ≥100 polyps at a later age, although most have <100 polyps) [Burt et al 2004]. (2) One limitation in the proposed criteria is that they do not take into account APC mutation status. A significant proportion of persons with polyposis who do not have an identified APC mutation have biallelic MYH mutations and therefore should be classified as having MYH-associated polyposis (see Differential Diagnosis).

Variable features not included in the diagnostic criteria of an APC-associated polyposis condition but potentially helpful in establishing the clinical diagnosis include: gastric polyps, duodenal adenomatous polyps, osteomas, dental abnormalities (especially supernumerary teeth and/or odontomas), congenital hypertrophy of the retinal pigment epithelium (CHRPE), soft tissue tumors (specifically epidermoid cysts and fibromas), desmoid tumors, and associated cancers.

Histology of adenomatous polyps

Dysplasia. Adenomatous polyps (often referred to as adenomas) are precancerous growths in which the surface epithelium of the gastrointestinal tract exhibits features of dysplasia. Dysplasia is characterized by branching of the microscopic glands, loss of goblet cells, and the following cellular features: loss of basilar polarity of the nucleus, increased nuclear/cytoplasmic ratio, increased basophilia of the cytoplasm, and loss of cytoplasmic glycogen. Dysplasia is graded as mild, moderate, or severe. Severe dysplasia is more likely to have cancer found somewhere within it and is more likely to progress to cancer.

Villous changes. In addition to the dysplastic features of an adenomatous polyp, villous changes, characterized by elongated villi at the surface of the polyp, may develop:

  • Villous adenomas exhibit villous changes on the majority of the polyp surface. A greater risk of cancer is associated with villous features within an adenomatous polyp than with adenomatous changes alone.

  • Tubular adenomas have no villous features.

  • Tubulovillous adenomas have some villous features.

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. APC is the gene associated with APC-associated polyposis conditions.

Clinical testing

  • Full gene sequencing of all APC exons and intron-exon boundaries appears to be the most accurate clinical test available to detect APC mutations [Giardiello et al 2001]. Most mutations in APC are nonsense or frameshift mutations that cause premature truncation of the APC protein.

    The likelihood of detecting an APC mutation is highly dependent on the severity of colonic polyposis:

    • Individuals with classic FAP are significantly more likely to have an APC mutation than individuals with a less severe colonic phenotype (i.e., <100 polyps) [Sieber et al 2002, Aretz et al 2005, Michils et al 2005, Aretz et al 2006].

    • Fewer than 30% of individuals with attenuated phenotypes are expected to have an identifiable APC mutation [Lefevre et al 2006].

    • Approximately 20% of individuals with an apparent de novo APC mutation have somatic mosaicism [Hes et al 2007].

      Note: In individuals with somatic mosaicism, sequencing of the APC gene may fail to detect disease-causing mutations because of weak mutation signals in peripheral blood lymphocytes [Aretz et al 2007, Hes et al 2007]. This, in part, may explain the lower mutation detection rate in simplex cases (i.e., a single occurrence in a family) than in persons with an affected parent.

  • Protein truncation testing, which is positive in approximately 80% of individuals with classic FAP [Powell et al 1993], has largely been replaced by more sensitive full gene sequencing techniques.

  • Duplication/deletion analysis. Methods commonly used to detect partial and whole-gene deletions or other large rearrangements include Southern blot analysis, multiplex ligation-dependent probe amplification (MLPA), and quantitative PCR. Approximately 8%-12% of individuals with an APC-associated polyposis condition and 100 or more polyps have a partial or whole APC gene deletion [Sieber et al 2002, Bunyan et al 2004, Aretz et al 2005, Michils et al 2005]. In one study, 19 of 296 (6%) individuals with ten or more adenomatous polyps who had no mutations in MYH (see Differential Diagnosis) or APC using sequencing, protein truncation testing, and denaturing gradient gel electrophoresis had a large APC deletion detected by MLPA [Nielsen et al 2007b].

    Interstitial deletions of chromosome 5q that include the APC gene have been identified on routine chromosome analysis in several individuals with colonic polyposis and mental retardation [Heald et al 2007]. In at least one individual, array comparative genomic hybridization (array CGH) detected a deletion that was not visible on routine cytogenetic studies [Heald et al 2007].

    Note: Cytogenetic analysis and/or array CGH are generally pursued only when adenomatous polyposis is accompanied by developmental delays.

Table 1 summarizes molecular genetic testing for this disorder.

Table 1. Molecular Genetic Testing Used in APC-Associated Polyposis Conditions

Gene SymbolTest MethodMutations DetectedMutation Detection Frequency by Test Method 1Test Availability
APCSequence analysisSequence variants≤90% 2Clinical graphic element
Duplication/deletion analysisDuplication/deletion of one or more exons~8-12% 3

1. Detection rates are higher in classic polyposis than in attenuated colonic phenotypes [Sieber et al 2002, Aretz et al 2005, Michils et al 2005, Aretz et al 2006] and in individuals with a family history of polyposis than in those without affected family members in the previous generation [Truta et al 2005, Aretz et al 2007, Hes et al 2007].

  • Linkage analysis. When no disease-causing mutation is identified in an affected individual, linkage analysis can be considered in families with more than one affected family member belonging to different generations. Linkage studies are based on an accurate clinical diagnosis of an APC-associated polyposis condition in the affected family members and accurate understanding of genetic relationships in the family. Linkage analysis is dependent on the availability and willingness of family members to be tested. The markers used for linkage analysis of APC-associated polyposis conditions are highly informative and very tightly linked to the APC locus; thus, they can be used with greater than 98% accuracy in more than 95% of families with an APC-associated polyposis condition [Petersen et al 1991, Burt et al 1992]. Linkage testing is not available to families with a single affected individual, a situation that often occurs when an individual has a de novo gene mutation and no affected offspring.

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

Testing Strategy

To confirm the diagnosis in a proband

  • In individuals meeting the diagnostic criteria for FAP or individuals suspected of having an APC-associated polyposis condition, sequence analysis and duplication/deletion analysis of APC are performed.

  • If no disease causing APC mutation is found, molecular genetic testing of the MYH gene (see Differential Diagnosis) should be considered.

Predictive testing for at-risk asymptomatic family members requires prior identification of the disease-causing mutation in the family.

Note: If no alteration in the APC gene is identified in a family with more than one affected relative belonging to different generations, linkage analysis can be considered.

Prenatal diagnosis and preimplantation genetic diagnosis (PGD) for at-risk pregnancies require prior identification of the disease-causing mutation in the family.

Clinical Description

Natural History

APC-associated polyposis conditions include classic FAP, the two overlapping phenotypes Gardner syndrome and Turcot syndrome, and attenuated FAP.

Classic FAP

Colorectal adenomatous polyps begin to appear, on average, at age 16 years (range 7-36 years) [Petersen et al 1991]. By age 35 years, 95% of individuals with classic FAP have polyps. Once they appear, the polyps rapidly increase in number; when colonic expression is fully developed, hundreds to thousands of colonic adenomatous polyps are typically observed. Without colectomy, colon cancer is inevitable. The average age of colon cancer diagnosis in untreated individuals is 39 years (range 34-43 years). Seven percent of untreated individuals with FAP develop colon cancer by age 21 years, 87% by 45 years, and 93% by 50 years [Bussey 1975]. Although rare, asymptomatic individuals in their 50s have been reported [Evans et al 1993]. Inter- and intrafamilial phenotypic variability are common [Giardiello et al 1994, Rozen et al 1999].

Other features variably present in FAP

Gastric polyps. Gastric polyps can be either fundic gland or adenomatous [Bülow et al 1995]:

  • Gastric fundic gland polyps, hamartomatous tumors located in the fundus and body of the stomach, occur in approximately half of individuals with FAP [Offerhaus et al 1999]. For a complete review of gastric fundic gland polyps and their relationship to FAP and attenuated FAP, see Burt [2003].

  • Adenomatous polyps, the second most prevalent gastric lesion in individuals with FAP [Bülow et al 1995, Wallace & Phillips 1998], are usually confined to the gastric antrum [Offerhaus et al 1999].

The risk for gastric cancer in individuals with FAP living in Western cultures is low, although it has been reported [Offerhaus et al 1999, Garrean et al 2008]. The rates of gastric cancer in persons of Japanese and Korean heritage with FAP may be tenfold higher than the general population [Garrean et al 2008]. Gastric adenocarcinoma is believed to arise most often from adenomas but may also develop from fundic gland polyps [Zwick et al 1997, Hofgartner et al 1999, Attard et al 2001].

Adenomatous polyps of the small bowel. Adenomatous polyps of the duodenum, observed in 50%-90% of individuals with FAP, are commonly found in the second and third portions of the duodenum [Kadmon et al 2001] and rarely in the distal small bowel [Wallace & Phillips 1998]. A classification system for duodenal polyps, based on number and size of polyps, histology, and degree of dysplasia, has been developed [Spigelman et al 1989]. No clear association between the number of colonic polyps and the number of upper gastrointestinal polyps has been identified [Kadmon et al 2001]. The lifetime risk of small bowel malignancy is 4%-12%; the majority occurs in the duodenum.

Adenomatous polyps of the periampullary region (including the duodenal papilla and ampulla of Vater) are seen in at least 50% of individuals with classic FAP. Polyps in this area can cause obstruction of the pancreatic duct resulting in pancreatitis or biliary obstruction, both of which occur at increased frequency in FAP. These polyps are often small and require a side-viewing endoscope for visualization. Some theorize that pancreaticobiliary secretions (e.g., bile) affect the development of adenomas [Wallace & Phillips 1998], and may account for the observed increased risk of malignancy of polyps in the periampullary region [Kadmon et al 2001].

Extraintestinal manifestations

Osteomas are bony growths found most commonly on the skull and mandible; however, they may occur in any bone of the body. Osteomas do not usually cause clinical problems and do not become malignant; they may appear in children prior to the development of colonic polyps.

Dental abnormalities. Unerupted teeth, congenital absence of one or more teeth, supernumerary teeth, dentigerous cysts (an odontogenic cyst associated with the crown of an unerupted tooth), and odontomas have been reported in approximately 17% of individuals with FAP compared to 1%-2% of the general population [Brett et al 1994].

Congenital hypertrophy of the retinal pigment epithelium (CHRPE) refers to discrete, flat, pigmented lesions of the retina that are not age dependent and do not cause clinical problems. Visualization of CHRPE may require examination of the ocular fundus with an indirect ophthalmoscope through a dilated pupil. Observation of multiple or bilateral CHRPE may be an indication that an at-risk family member has inherited FAP, whereas isolated lesions may be seen in the general population [Chen et al 2006].

Benign cutaneous lesions include epidermoid cysts and fibromas that may be found on any part of the body, including the face, and are mainly of cosmetic concern. Multiple pilomatricomas, although rare, have also been reported [Pujol et al 1995].

Desmoid tumors develop in approximately 10% of children and adults with FAP [Gurbuz et al 1994, Clark & Phillips 1996]. The risk of desmoid tumors in individuals with FAP is 852 times the risk in the general population [Gurbuz et al 1994]. These poorly understood, benign fibrous tumors are clonal proliferations of myofibroblasts that are locally invasive but do not metastasize [Clark et al 1999]. A pathologically distinct fibromatous lesion called a Gardner-associated fibroma (GAF) is hypothesized to be a precursor lesion [Wehrli et al 2001].

Desmoid tumors form predominantly within the abdomen or in the abdominal wall but may also occur extra-abdominally. Desmoid tumors may compress abdominal organs or complicate abdominal surgery. About 5% of individuals with FAP experience morbidity and/or mortality from desmoid tumors. Abdominal desmoid tumors may occur spontaneously or following abdominal surgery [Bertario et al 2001]. Hyper-estrogen states such as pregnancy or oral contraceptive use may also increase risk. Independent predictors for desmoid tumor development include: an APC mutation 3' of codon 1444, family history of desmoids, female gender, and the presence of osteomas [Bertario et al 2001].

Desmoid tumors are best evaluated by CT scan [Clark & Phillips 1996] or MRI. A CT scoring system for desmoid tumors in FAP has been developed [Middleton et al 2003].

Adrenal masses. Although not thoroughly studied, a statistically significant association between adrenal masses and FAP has been reported. Adrenal masses are found in 1%-3% of the general population; a retrospective analysis identified adrenal masses in 7.4% of individuals with FAP [Marchesa et al 1997], and a prospective study of 107 individuals with FAP found 13% with an adrenal mass greater than or equal to 1.0 cm on abdominal CT scan [Smith et al 2000b]. Most of these masses appeared to be adrenocortical adenomas without endocrinopathy or hypertension.

Extracolonic cancers. Several extracolonic cancers occur with a higher incidence in individuals with FAP than in the general population (Table 2) [Burt 2000].

Table 2. Lifetime Risk of Extracolonic Cancer in FAP

SiteType of CancerLifetime Risk of Cancer
Small bowel: duodenum or periampullaCarcinoma4%-12%
Small bowel: distal to the duodenumRare
PancreasAdenocarcinoma~2%
ThyroidPapillary thyroid carcinoma1%-2%
CNSUsually medulloblastoma<1%
LiverHepatoblastoma1.6%
Bile ductsAdenocarcinomaLow, but increased

Duodenal adenocarcinoma has been reported between ages 17 and 81 years, with the mean age of diagnosis between 45 and 52 years [Wallace & Phillips 1998, Kadmon et al 2001]. It occurs most commonly in the periampullary area. Small bowel cancer distal to the duodenum occurs but is rare.

Thyroid cancers affect approximately 1%-2% of individuals with FAP [Cetta et al 2000, Truta et al 2003]. Familial occurrence and a female preponderance have been observed. In one small series of females with FAP, the prevalence was 12% [Herraiz et al 2007]. Papillary histology predominates and may commonly have a cribriform pattern.

The risk of hepatoblastoma in FAP is 750 to 7500 times higher than in the general population, although the absolute risk is estimated at less than 2% [Aretz et al 2007]. The majority of hepatoblastomas occur prior to age three years [Aretz et al 2007].

Pregnancy/hormone use. Limited information is available on the effect of pregnancy on females with FAP. In one study of 58 Danish women with FAP, the same frequency of fertility, pregnancy, and delivery was observed as in a control population [Johansen et al 1990]. A larger study of 162 women with FAP compared fertility rates before and after two types of colorectal surgery with a control population. Women with FAP who had not yet undergone surgery had the same fertility as a control population of normal women. Additionally, those women with FAP who had a colectomy with ileorectal anastomosis (IRA) had the same fertility as the control population. Fertility was significantly reduced in women with FAP who had a proctocolectomy with ileal pouch-anal anastomosis (IPAA) compared to the control population [Olsen et al 2003].

Women who have undergone colectomy are considered to have the same risk of obstetrical complications as any other woman who has had major abdominal surgery.

As anti-estrogen medications have been successfully used in the treatment of desmoid tumors, the development of desmoid tumors is thought to be affected by hormones important in pregnancy. However, one study has shown that women who had a previous pregnancy and developed a desmoid tumor had significantly fewer complications from the desmoid tumor than those who had never had a pregnancy [Church & McGannon 2000].

In a study of women with FAP at the time of their colectomy, no association was found between pregnancy history and colonic polyp severity; however, the proportion of parous women with severe duodenal disease was significantly higher than the proportion of nulliparous women [Suraweera et al 2007].

Some studies have suggested that female hormones protect against colorectal cancer development in the general population. In one woman, reduction in polyps after use of oral contraceptives was observed [Giardiello et al 2005].

Gardner Syndrome

Gardner syndrome (GS) is the association of colonic adenomatous polyposis of classic FAP with osteomas and soft tissue tumors (epidermoid cysts, fibromas, desmoid tumors) [Gardner & Richards 1953]. These benign, extraintestinal growths occur in about 20% of individuals and families with FAP. When these findings are prominent, many clinicians continue to use the term Gardner syndrome.

Osteomas are most commonly found on the mandible and skull, although any bone of the body may be involved. Epidermoid cysts occur on any cutaneous surface and are mainly of cosmetic concern, as they do not appear to have malignant potential. Supernumerary teeth, odontomas, and desmoid tumors were originally described as a part of Gardner syndrome; however, like osteomas and epidermoid cysts, they can occur in any individual with FAP, whether or not other extraintestinal findings are present.

Gardner syndrome was once thought to be a distinct clinical entity; however, it is now known that mutations in the APC gene give rise to both classic FAP and Gardner syndrome. Other manifestations of FAP, such as upper gastrointestinal polyposis, are also found in Gardner syndrome. Some correlation exists between extraintestinal growths and mutation location in APC. See Genotype-Phenotype Correlations.

Turcot Syndrome

Turcot syndrome is the association of colonic polyposis or colorectal cancer and CNS tumors. The molecular basis of most Turcot syndrome is either a mutation in the APC gene associated with FAP or a mutation in one of the mismatch repair genes associated with hereditary non-polyposis colon cancer (HNPCC) [Hamilton et al 1995]. The CNS tumors in individuals with APC mutations are typically medulloblastoma, whereas those with mismatch repair mutations are usually glioblastoma multiforme. The risk of CNS tumors is substantially increased in persons with FAP generally, although the absolute risk is only approximately 1%. Families with APC-associated polyposis conditions in which multiple individuals have CNS tumors raise the possibility of mutation specificity or modifying genes.

Attenuated FAP

Attenuated FAP is characterized by fewer colonic polyps (average of 30) than classic FAP but a significant risk for colorectal cancer. Polyps tend to be found more proximally in the colon than in classic FAP.

The exact lifetime risk of colorectal cancer in attenuated FAP is unclear; the cumulative risk by age 80 years is estimated to be approximately 70% [Neklason et al 2008]. The average age of colon cancer diagnosis in individuals with attenuated FAP is 50 to 55 years — ten to 15 years later than in those with classic FAP, but earlier than in those with sporadically occurring colon cancer [Spirio et al 1993, Giardiello et al 1997].

In two large kindreds with attenuated FAP and an identical APC germline mutation [Burt et al 2004, Neklason et al 2008]:

  • The median number of adenomatous polyps in 120 mutation-positive individuals was 25 (range 0-470).

  • Forty-four of 120 (~37%) mutation-positive individuals with detailed colonoscopy records available had fewer than ten adenomatous polyps.

  • Three of the 44 mutation-positive individuals with fewer than ten polyps had colorectal cancer; one of the three was diagnosed before age 30 years.

Additional findings in attenuated FAP can include:

Genotype-Phenotype Correlations

Although variation occurs among and within individuals and among and within families with identical APC mutations [Giardiello et al 1994, Friedl et al 2001], much effort has gone into making genotype-phenotype correlations. Some have suggested basing management strategies on these associations [Vasen et al 1996], whereas others feel that therapeutic decisions should not be based on genotype [Friedl et al 2001].

While not in routine use at present, the following correlations may become important in management decisions in the future (see Table 3 for reference sequences for mutations discussed in this section):

  • The most frequent APC mutation is located at codon 1309 (c.3927_3931delAAAGA) [Friedl & Aretz 2005]. Mutations at this codon lead to a high number of colonic adenomas at an early age [Friedl et al 2001, Bertario et al 2003].

  • The average age of onset in individuals with colonic symptoms [Friedl et al 2001] varied by mutation location:

    • At codon 1309: age 20 years

    • Between codon 168 and 1580 (excluding 1309): age 30 years

    • 5' of codon 168 and 3' of codon 1580: age 52 years

  • Profuse polyposis (an average of 5000 polyps) has been reported with mutations in codons 1250-1464 [Nagase et al 1992].

  • Attenuated FAP is associated with the following:

  • A fourfold increased risk for duodenal adenomas was found in individuals with mutations between codons 976 and 1067 in one study of Italian individuals with FAP [Bertario et al 2003].

  • Prominent extracolonic manifestations often correlate (though not completely) with more distal APC mutations. A retrospective study of 190 individuals with FAP that evaluated nine extracolonic manifestations (desmoid tumors, osteomas, epidermoid cysts, duodenal adenomas, gastric polyps, hepatoblastoma, dental anomalies, periampullary cancers, and brain tumors) [Wallis et al 1999] revealed that:

    • Individuals with mutations in codons 1395-1493 have significantly higher rates of desmoid tumors, osteomas, and epidermoid cysts than those with mutations in codons 177-452.

    • Individuals with mutations in codons 1395-1493 have significantly higher rates of desmoid tumors and osteomas than those with mutations in codons 457-1309.

    • No individuals with mutations in codons 177-452 developed osteomas or periampullary cancers.

    • Only individuals with mutations in codons 457-1309 developed hepatoblastoma and/or brain tumors.

  • Desmoid tumors show the following correlations:

    • APC mutations between codons 1444 and 1580 are associated with a higher incidence of desmoid tumors than mutations in other codons [Caspari et al 1995, Davies et al 1995].

    • A study of 269 individuals with identifiable APC mutations found desmoid tumors in 20% of individuals with mutations 5' to codon 1444, 49% of individuals with mutations 3' to codon 1444, and 61% of individuals with mutations in codons 1445-1580 [Friedl et al 2001].

    • Several families with severe desmoid tumors with mutations at the extreme 3' end of the gene have been reported [Eccles et al 1996, Scott et al 1996, Couture et al 2000].

    • A study of Italian individuals with FAP found that mutations between codons 1310 and 2011 are associated with a sixfold risk of desmoid tumors compared to mutations between codons 159 and 495 [Bertario et al 2003].

    • A review of the literature by Nieuwenhuis & Vasen [2007] revealed a consistent association of desmoid tumors with mutations distal to codon 1444.

  • CHRPE is associated with:

  • In individuals with thyroid cancer and FAP:

    • In 24 individuals, the majority of mutations identified were 5' to codon 1220 [Cetta et al 2000].

    • Nine of 12 individuals had APC mutations identified proximal to the mutation cluster region (codons 1286-1513) [Truta et al 2003].

  • A review of the literature up to August 2006 and a report of additional cases by Nielsen et al [2007a] revealed 89 submicroscopic APC gene deletions (42 partial and 47 whole-gene deletions). Most partial and whole APC gene deletions are associated with 100-2000 colonic adenomas, although attenuated FAP has been seen [Nielsen et al 2007a]. Extracolonic findings were seen in 36% of cases, with no significant differences between those with partial and whole-gene deletions [Nielsen et al 2007a].

Penetrance

In FAP, the penetrance of colonic adenomatous polyposis and colon cancer is virtually 100% in untreated individuals.

The penetrance of other intestinal and extraintestinal manifestations is less well understood and may depend in part on the mutation location in the APC gene.

Anticipation

Although a recent observation has suggested the possibility of anticipation in APC-associated polyposis conditions [Heald et al 2007], true genetic anticipation (in which subsequent generations have an increased risk of more severe disease manifestations because of the underlying mutational mechanism) has not been observed in APC-associated polyposis conditions. Rather, milder disease manifestations in the first person to have the disorder in a family are most often the result of somatic mosaicism for the disease-causing mutation in that individual.

Nomenclature

Another term used historically for FAP is adenomatous polyposis coli (i.e., APC), which is now used for the relevant gene.

The term Gardner syndrome is mainly of historical interest as it is now known to arise from mutations of the APC gene like FAP. Furthermore, with sufficient investigation, subtle extraintestinal manifestations can be found in almost all individuals with FAP. Nonetheless, individuals and families with particularly prominent extracolonic manifestations will undoubtedly continue to be referred to as having Gardner syndrome.

In some families with FAP, multiple individuals have CNS tumors, making Turcot syndrome a historical term of uncertain significance as it relates to FAP.

Attenuated FAP appears to be the same as the “hereditary flat adenoma syndrome” [Lynch et al 1992].

Prevalence

The prevalence data reported from national registries include all of the APC-associated polyposis conditions (except possibly some cases of attenuated FAP); reported prevalence is 2.29 to 3.2 per 100,000 individuals [Burn et al 1991, Jarvinen 1992, Bülow et al 1996].

Attenuated FAP is likely underdiagnosed, given the lower number of colonic polyps and lower risk of colorectal cancer compared to classic FAP [Neklason et al 2008].

APC-associated polyposis conditions historically accounted for about 0.5% of all colorectal cancers; this figure is declining as more at-risk family members undergo successful treatment following early polyp detection and prophylactic colectomy.

Differential Diagnosis

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

APC-associated polyposis conditions may be distinguished from other inherited colon cancer conditions and other gastrointestinal polyposis syndromes by molecular genetic testing, histopathologic findings, and phenotypic characteristics. Conditions to consider in the differential diagnosis include the following hereditary disorders:

Conditions to be considered in the differential diagnosis include the following acquired disorders:

Management

Evaluations Following Initial Diagnosis

To establish the extent of disease in an individual diagnosed with an APC-associated polyposis condition, the following evaluations are recommended:

  • Personal medical history with particular attention to features of APC-associated polyposis (colon cancer, colon polyps, rectal bleeding, diarrhea, abdominal pain)

  • Family history with particular attention to features of APC-associated polyposis

  • Physical examination with particular attention to extraintestinal manifestations of APC-associated polyposis

  • Ophthalmologic evaluation for presence of congenital hypertrophy of the retinal pigment epithelium (CHRPE) (optional)

  • Colonoscopy with review of pathology

  • Consideration of upper GI tract evaluation, including endoscopy with a side-viewing scope; if symptomatic, small-bowel imaging such as small-bowel enteroclysis (an x-ray that looks at how contrast moves through the area) or abdominal and pelvic CT with contrast

Note: Smith et al [2000b] and Ferrández et al [2006] found no evidence to warrant screening for adrenal masses in FAP.

Treatment of Manifestations

Colonic polyps. Practice parameters, including information on surgery, have been outlined by the American Society of Colon and Rectal Surgeons [Church et al 2003b], in addition to the American Society of Clinical Oncology and the Society of Surgical Oncology [Guillem et al 2006].

For individuals with classic FAP, colectomy is recommended after adenomas emerge; colectomy may be delayed depending on the size and number of adenomatous polyps. Colectomy is usually advised when more than 20 or 30 adenomas or multiple adenomas with advanced histology have developed.

For individuals with attenuated FAP, colectomy may be necessary, but in approximately one-third of individuals the colonic polyps are limited enough in number that surveillance with periodic colonoscopic polypectomy is sufficient (See Surveillance).

The types of colectomy include the following:

  • Restorative proctocolectomy

  • Proctocolectomy with ileal pouch anal anastomosis

  • Total colectomy with ileorectal anastomosis; often used for individuals with attenuated FAP or in instances in which the rectum is spared of polyps

  • Total proctocolectomy with permanent ileostomy.

    Note: This procedure is rarely needed.

A study of individuals with FAP and ileal pouches found that 57% had adenomatous polyps in the ileal pouch. No apparent relationship between the development of pouch adenomas and the severity of polyps in the colon or duodenum was found [Groves et al 2005].

The risk of cancer in the surgical transition zone is very low but has been reported [Ooi et al 2003].

Small bowel polyps. Endoscopic or surgical removal of duodenal and/or ampullary adenomas should be considered if polyps exhibit villous change or severe dysplasia, exceed one centimeter in diameter, or cause symptoms [Wallace & Phillips 1998, Saurin et al 1999, Kadmon et al 2001].

Pancreaticoduodenectomy (Whipple procedure) may occasionally be necessary to treat severe duodenal adenomas.

Osteomas may be removed for cosmetic reasons.

Desmoid tumors. Available treatments include surgical excision (associated with high rates of recurrence), nonsteroidal anti-inflammatory drugs (NSAIDs), anti-estrogens, cytotoxic chemotherapy, and radiation [Griffioen et al 1998, Clark et al 1999, Smith et al 2000a, Tonelli et al 2003, Gega et al 2006]. A review of desmoid treatments can be found in Guillem et al [2006].

Nonsteroidal anti-inflammatory drugs (NSAIDs), especially sulindac, [Steinbach et al 2000, Higuchi et al 2003, Keller & Giardiello 2003], have been shown to cause regression of adenomas in FAP and to decrease the number of polyps requiring ablation in the remaining rectum of individuals who have had a colectomy with ileorectal anastomosis.

Note: NSAID use before colectomy remains experimental.

Sulindac appears to be the only option. Withdrawal from the market of rofecoxib in 2005 because of untoward cardiovascular and cerebrovascular events and the observation that similar events occur with the doses of celecoxib needed for adenoma regression has brought into question the long-term use of these agents for treatment of FAP.

Surveillance

Recommended surveillance of individuals known to have FAP or an APC disease-causing mutation and individuals at risk for FAP who have not undergone molecular genetic testing or who are members of families in which molecular genetic testing did not identify a disease-causing mutation [Giardiello et al 2001]:

  • Sigmoidoscopy or colonoscopy every one to two years, beginning at age ten to 12 years

  • Colonoscopy, once polyps are detected

  • Annual colonoscopy, if colectomy is delayed more than a year after polyps emerge. In individuals age ten to 20 years in whom adenomas are smaller than 6.0 mm and without villous component, delay in colectomy may be considered.

  • Esophagogastroduodenoscopy (EGD) beginning by age 25 years or prior to colectomy and repeated every one to three years

    Note: (1) The frequency of EGD depends on the severity of duodenal adenomas; Spigelman staging criteria can help determine the frequency of EGD.

    (2) A side-viewing instrument should be used to visualize the duodenal papilla.

    (3) As adenomatous tissue is commonly found at the papilla, biopsy may be justified if no polyps are visualized but the papilla seems enlarged.

    (4) In some cases, endoscopic retrograde cholangiopancreatography (ERCP) may be necessary to evaluate for adenomas of the common bile duct.

    (5) The utility of video capsule endoscopy (VCE) in screening for small-bowel lesions in FAP is unclear. Inaccurate identification of large polyps in the proximal small bowel and the inability to view the ampulla call into question the use of VCE in APC-associated polyposis conditions [Wong et al 2006].

  • Small-bowel imaging (small-bowel enteroclysis or abdominal and pelvic CT with orally administered contrast) when duodenal adenomas are detected or prior to colectomy, repeated every one to three years depending on findings and presence of symptoms

  • Screening for hepatoblastoma: efficacy in individuals with FAP is unclear. Screening protocols in Beckwith-Wiedemann syndrome, in which the risk of hepatoblastoma is also increased, often include frequent (every 2-3 months) abdominal ultrasound examinations and measurement of serum alpha-fetoprotein concentrations and have resulted in early detection of hepatoblastomas [Tan & Amor 2006]. Screening for hepatoblastoma in FAP using the same protocol may be considered from infancy to age five years. However, the optimal interval for hepatoblastoma screening in FAP is not known, although it has been recommended that screening should occur at least every three months [Hirschman et al 2004, Aretz et al 2007].

  • Annual physical examination, including evaluation for extraintestinal manifestations, usually for cosmetic concerns, and palpation of the thyroid with consideration of follow-up ultrasound examination and fine-needle aspiration if thyroid nodules are present [Herraiz et al 2007]

Testing of Relatives at Risk

Recommended genetic testing for at-risk family members. Early recognition of APC-associated polyposis conditions may allow for timely intervention and improved final outcome; thus, surveillance of asymptomatic, at-risk children for early manifestations is appropriate (see American Gastroenterological Association Medical Position Statement and American College of Medical Genetics/American Society of Human Genetics Joint Statement).

Use of molecular genetic testing for early identification of at-risk family members (see Genetic Counseling) improves diagnostic certainty and reduces the need for costly screening procedures in those at-risk family members who have not inherited the disease-causing mutation. A cost analysis comparing molecular genetic testing and sigmoidoscopy screening for individuals at risk for APC-associated polyposis conditions shows that genetic testing is more cost effective than sigmoidoscopy in determining who in the family is affected [Cromwell et al 1998]. Additionally, individuals diagnosed with APC-associated polyposis conditions as a result of having an affected relative have a significantly greater life expectancy than those individuals diagnosed on the basis of symptoms [Heiskanen et al 2000].

As colon screening for those at risk for classic FAP begins as early as age ten to12 years, molecular genetic testing is generally offered to children at risk for classic FAP by age ten years. Genetic testing at birth may also be warranted, as some parents and pediatricians may consider hepatoblastoma screening from infancy to age five years in affected offspring. Colon screening for those with attenuated FAP begins at age 18 to 20 years; thus, molecular genetic testing should be offered to those at risk for attenuated FAP at approximately age 18 years.

Note: No evidence points to an optimal age at which to begin screening; thus, the ages at which testing is performed and screening initiated may vary by center, family history, hepatoblastoma screening, and/or parents'/child's needs.

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

Therapies Under Investigation

Search ClinicalTrials.gov for access to information on clinical studies for a wide range of diseases and conditions. Note: There may not be clinical trials for this disorder.

Other

NSAIDs have been used unsuccessfully in an attempt to prevent the emergence of colonic adenomatous polyposis [Giardiello et al 2002].

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

APC-associated polyposis conditions are inherited in an autosomal dominant manner.

Risk to Family Members

Parents of a proband

  • Approximately 20%-25% of individuals with an APC-associated polyposis condition have the altered gene as the result of a de novo gene mutation [Bisgaard et al 1994].

  • Investigations to determine the parental origin of a de novo APC gene mutation suggest a slight preponderance of mutations of paternal origin (12/16 families; not statistically significant) [Aretz et al 2004] while another report shows equal maternal and paternal origin [Ripa et al 2002]. Thus, de novo APC mutations do not appear to demonstrate an advanced paternal age effect [Ripa et al 2002, Aretz et al 2004].

  • It is appropriate to evaluate the parents of an affected individual (a) with molecular genetic testing of the APC gene if the disease-causing mutation is known in the proband or (b) for clinical manifestations of APC-associated polyposis conditions.

Note: Although most individuals diagnosed with an APC-associated polyposis condition 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 depends on the genetic status of the parents.

  • If a parent is affected or has the disease-causing mutation, the risk to the sibs of inheriting the mutation is 50%.

  • If neither parent has the APC mutation identified in the proband, the risk to the sibs is low but greater than that of the general population because of the possibility of germline mosaicism. Thus, molecular genetic testing should be offered to the sibs of an individual with an apparent de novo mutation.

  • Germline mosaicism has been documented in an asymptomatic 79 year old woman who had two sons with thousands of adenomatous colonic polyps and an APC mutation [Hes et al 2007]. Another unaffected woman was demonstrated to have germline mosaicism, as two of her children had colonic adenomatous polyposis and were subsequently found to have an APC mutation [Schwab et al 2007].

Offspring of a proband. Every child of an individual with an APC-associated polyposis condition has a 50% chance of inheriting the mutation.

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 or to have an APC disease-causing mutation, his or her family members are at risk.

Related Genetic Counseling Issues

Testing of at-risk asymptomatic individuals during adulthood and childhood. Consideration of molecular genetic testing of young, at-risk family members is appropriate for guiding medical management (see Management).

Molecular genetic testing can be used with certainty to clarify the genetic status of at-risk family members when a clinically diagnosed relative has undergone molecular genetic testing and is found to have a mutation in the APC gene.

The use of molecular genetic testing for determining the genetic status of at-risk relatives when a clinically diagnosed relative is not available for testing is problematic, and test results need to be interpreted with caution. A positive test result in the at-risk family member indicates the presence of an APC disease-causing mutation and also indicates that the same molecular genetic testing method can be used to assess the genetic status of other, at-risk family members. In contrast, when genetic testing is offered to an at-risk family member prior to testing a family member known to be affected, the failure to identify a disease-causing mutation in the at-risk family member does not eliminate the possibility that an APC disease-causing mutation is present. The genetic status of such individuals cannot be determined through molecular genetic testing, and they need to follow the recommendations for clinical surveillance of at-risk family members.

Because colon screening for those at risk for classic FAP begins as early as age ten years, molecular genetic testing is generally offered to individuals by this age. Colon screening for those at risk for attenuated FAP begins at age 18 to 20 years; thus, molecular genetic testing should be offered at about age 18 years. Molecular genetic testing may be performed earlier if it alters medical management of the child, as is the case when parents are considering hepatoblastoma screening for their at-risk offspring. Predictive genetic testing may be considered within the first few months of life as a result of the increased risk of hepatoblastoma in FAP.

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 altered gene. 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. Although most children do not show evidence of clinically significant psychological problems after presymptomatic testing, Codori et al [2003] recommend that long-term psychological support be available to these families.

Other issues to consider. It is recommended that physicians ordering APC 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. A study demonstrated that for almost one-third of individuals assessed for FAP, the physician misinterpreted the test results [Giardiello et al 1997]. In addition, Michie et al [2002] found that at-risk relatives who were found to be mutation-negative were more likely to request continued bowel surveillance when results were relayed to them by non-geneticist physicians than by genetics professionals. In a follow-up study evaluating why some at-risk individuals are not reassured by negative molecular genetic test results and request continued surveillance, Michie et al [2003] conclude that effective communication is key to facilitating adaptive behavior. Referral to a genetic counselor and/or a center in which testing is routinely offered is recommended.

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:

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 maternity (e.g., with assisted reproduction) or undisclosed adoption could also be explored.

Family planning

  • The optimal time for determination of genetic risk is 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 are carriers, or are at risk of being affected or carriers.

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 50% risk for APC-associated polyposis conditions is possible by analysis of DNA extracted from fetal cells obtained by amniocentesis usually performed at about 15 to 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. The criteria for use of molecular genetic testing discussed in Testing of at-risk asymptomatic individuals during adulthood and childhood apply to prenatal testing as well. It should be noted that detection of an APC mutation in a fetus at risk does not predict the time of onset or severity of the disease.

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

Requests for prenatal testing for conditions such as the APC-associated polyposis that do not affect intellect and have treatment available are not common. A pilot study of 20 individuals with FAP revealed that 100% believed it was ethical to provide prenatal testing for FAP, and 95% (19/20) would consider it themselves [Kastrinos et al 2007]. Differences in perspective may exist among medical professionals and within families regarding the use of prenatal testing, particularly if the testing is being considered for the purpose of pregnancy termination rather than early diagnosis. Although most centers would consider decisions about prenatal testing to be the choice of the parents, discussion of these issues is appropriate.

Preimplantation genetic diagnosis (PGD) has been successfully used in pregnancies at risk for several inherited cancer predisposition syndromes and may be an option for couples at risk of having offspring with an APC-associated polyposis condition [Rechitsky et al 2002, Davis et al 2006, Moutou et al 2007]. The parent's disease-causing allele must be identified before PGD can be performed. 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 APC-Associated Polyposis Conditions

Gene SymbolChromosomal LocusProtein Name
APC5q21-q22Adenomatous polyposis coli protein

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 APC-Associated Polyposis Conditions

 175100 ADENOMATOUS POLYPOSIS OF THE COLON; APC
 276300 MISMATCH REPAIR CANCER SYNDROME

Table C. Genomic Databases for APC-Associated Polyposis Conditions

Gene SymbolLocus SpecificEntrez GeneHGMD
APCAPC324 (MIM No. 175100)APC

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

Note: HGMD requires registration

Normal allelic variants: The gene is alternatively spliced in multiple coding and noncoding regions; the main transcript has 15 exons with 8532 base pairs that code for 2843 amino acids and result in a 311.8-kd protein. Exon 15 is large and comprises over three-quarters of the coding region of the gene.

Pathologic allelic variants: Over 826 germline mutations have been found in families with an APC-associated polyposis condition [Beroud et al 2000]. Mutations almost always cause a premature truncation of the APC protein, usually through single amino-acid substitutions or frameshifts. While mutations have been found scattered throughout the gene, they are predominantly located in the 5' end of the gene. The most common germline APC mutation is c.3927_3931delAAAGA. (For more information, see Genomic Databases table.)

Table 3. APC Allelic Variants Discussed in This GeneReview

Class of Variant Allele
DNA Nucleotide Change
(Alias1)
Protein Amino
Acid Change
(Alias1)
Reference
Sequence
Normal 2c.5465T>Ap.Val1822Asp
(D1822V)
NM_000038.3NP_000029.2
Uncertain clinical significance 1c.3949G>Cp.Glu1317Gln
Predisposition to colon cancer 1c.3920T>Ap.Ile1307Lys
Pathologicc.3927_3931delAAAGAp.Glu1309AspfsX4

See Quick Reference for an explanation of nomenclature. GeneReviews follows the standard naming conventions of the Human Genome Variation Society (http://www.hgvs.org).
1. Variant designation that does not conform to current naming conventions
2. See Genetically Related Disorders.

Normal gene product: The APC protein has been localized to the nucleus and membrane/cytoskeleton in human epithelial cells [Neufeld & White 1997]. It has also been shown to homodimerize [Joslyn et al 1993] and bind to other proteins including GSK3b [Rubinfeld et al 1996], b-catenin [Rubinfeld et al 1993, Su et al 1993], g-catenin [Hulsken et al 1994, Rubinfeld et al 1995], tubulin [Munemitsu et al 1994, Smith et al 1994], EB1 [Su et al 1995], and hDLG, a homolog of the Drosophila discs large tumor-suppressor protein [Matsumine et al 1996]. The APC protein product is a tumor suppressor. APC protein forms a complex with glycogen synthase kinase 3b (GSK-3b) [Rubinfeld et al 1996], which targets b-catenin, a protein involved in both cell adhesion and intracellular signal transduction [Korinek et al 1997, Morin et al 1997, Nakamura 1997, Peifer 1997, Rubinfeld et al 1997]. The presence of normal APC protein appears to maintain normal apoptosis and may also decrease cell proliferation, probably through its regulation of b-catenin. This pathway is normally involved with Wingless-Wnt signaling, which participates in several known cell growth functions.

The APC protein has been shown to accumulate at the kinetochore during mitosis, contribute to kinetochore-microtubule attachment, and play a role in chromosome segregation in mouse embryonic stem cells [Fodde et al 2001, Kaplan et al 2001]. The APC protein may play a role in chromosomal instability, the presence of which is often observed when APC function is lost.

Other possible roles for the APC protein include: regulation of cell migration up the colonic crypt and cell adhesion through association with E-cadherin, regulation of cell polarity through association with GSK3b and other functions related to association with microtubule bundles [Nathke et al 1996, Barth et al 1997, Etienne-Manneville & Hall 2003]. Goss & Groden [2000] provide an excellent review of the function of the APC protein.

Abnormal gene product: Disease-causing mutations in the APC gene most often result in truncated protein products. Experiments have localized normal full-length APC protein to the membrane/cytoskeleton and nuclear fractions of human epithelial cells but demonstrated that colon cancer cells containing only mutant APC genes revealed no truncated APC protein in nuclear fractions [Neufeld & White 1997].

When the APC gene is mutated and abnormal protein is present, high levels of free cytosolic b-catenin result. Free b-catenin migrates to the nucleus, binds to a transcription factor Tcf-4 or Lef-1 (T cell factor-lymphoid enhancer factor), and may activate expression of genes such as the oncogenes c-Myc and cyclin D1 [Chung 2000]. The specific genes targeted are not yet known but may include those increasing proliferation or decreasing apoptosis. Because APC may be important in cell migration, abnormal APC protein may disrupt normal cellular positioning in the colonic crypt. Additionally, mutations in the APC gene are thought to contribute to chromosomal instability in colorectal cancers [Fodde et al 2001].

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.

Collaborative Group of the Americas on Inherited Colorectal Cancer
www.cgaicc.com

Genetics of Colorectal Cancer (PDQ)
A service of the National Cancer Institute
Genetics of colorectal cancer

IMPACC (Intestinal Multiple Polyposis and Colorectal Cancer)
PO Box 11
Conyngham, PA 18219
Phone: 570-788-3712
Fax: 717-788-1818
Email: impacc@epix.net

National Library of Medicine Genetics Home Reference
Familial adenomatous polyposis

American Cancer Society
Provides contact information for regional support
1599 Clifton Road NE
Atlanta, GA 30329
Phone: 800-227-2345
www.cancer.org

C3: Colorectal Cancer Coalition
1225 King Street 2nd Floor
Alexandria, VA 22314
Phone: 703-548-1225
Fax: 202-315-3871
Email: info@FightColorectalCancer.org
www.FightColorectalCancer.org

Colon Cancer Alliance
1200 G Street, NW Suite 800
Washington, DC 20005
Phone: 877-422-2030 (toll-free helpline)
Fax: 866-304-9075
Email: kelly@ccalliance.org
www.ccalliance.org

Colorectal Cancer Network
PO Box 182
Kensington, MD 20895-0182
Phone: 301-879-1500
Fax: 301-879-1901
Email: CCNetwork@colorectal-cancer.net
www.colorectal-cancer.net

United Ostomy Association, Inc
PO Box 66
Fairview, TN 37062-0066
Phone: 800-826-0826
Email: info@uoa.org
www.uoa.org

Teaching Case-Genetic Tools
Cases designed for teaching genetics in the primary care setting
Case 9. Colorectal Cancer in a 28-Year-Old Woman

References

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

Literature Cited

Aretz S, Stienen D, Friedrichs N, Stemmler S, Uhlhaas S, Rahner N, Propping P, Friedl W. Somatic APC mosaicism: a frequent cause of familial adenomatous polyposis (FAP). Hum Mutat. 2007; 28: 98592. [PubMed]
Aretz S, Stienen D, Uhlhaas S, Pagenstecher C, Mangold E, Caspari R, Propping P, Friedl W. Large submicroscopic genomic APC deletions are a common cause of typical familial adenomatous polyposis. J Med Genet. 2005; 42: 18592. [PubMed]
Aretz S, Uhlhaas S, Caspari R, Mangold E, Pagenstecher C, Propping P, Friedl W. Frequency and parental origin of de novo APC mutations in familial adenomatous polyposis. Eur J Hum Genet. 2004; 12: 528. [PubMed]
Aretz S, Uhlhaas S, Goergens H, Siberg K, Vogel M, Pagenstecher C, Mangold E, Caspari R, Propping P, Friedl W. MUTYH-associated polyposis: 70 of 71 patients with biallelic mutations present with an attenuated or atypical phenotype. Int J Cancer. 2006; 119: 80714. [PubMed]
Attard TM, Giardiello FM, Argani P, Cuffari C. Fundic gland polyposis with high-grade dysplasia in a child with attenuated familial adenomatous polyposis and familial gastric cancer. J Pediatr Gastroenterol Nutr. 2001; 32: 2158. [PubMed]
Barth AI, Pollack AL, Altschuler Y, Mostov KE, Nelson WJ. NH2-terminal deletion of beta-catenin results in stable colocalization of mutant beta-catenin with adenomatous polyposis coli protein and altered MDCK cell adhesion. J Cell Biol. 1997; 136: 693706. [PubMed]
Beroud C, Collod-Beroud G, Boileau C, Soussi T, Junien C. UMD (Universal mutation database): a generic software to build and analyze locus-specific databases. Hum Mutat. 2000; 15: 8694. [PubMed]
Bertario L, Russo A, Sala P, Eboli M, Giarola M, D amico F, Gismondi V, Varesco L, Pierotti MA, Radice P (2001) Genotype and phenotype factors as determinants of desmoid tumors in patients with familial adenomatous.
Bülow S, Faurschou Nielsen T, Bülow C, Bisgaard ML, Karlsen L, Moesgaard F. The incidence rate of familial adenomatous polyposis. Results from the Danish Polyposis Register. Int J Colorectal Dis. 1996; 11: 8891. [PubMed]
Bülow S, Alm T, Fausa O, Hultcrantz R, Järvinen H, Vasen H, Bülow S, Alm T, Fausa O, Hultcrantz R, Jarvinen H, Vasen H. Duodenal adenomatosis in familial adenomatous polyposis. DAF Project Group. Int J Colorectal Dis. 1995; 10: 436. [PubMed]
Bunyan DJ, Eccles DM, Sillibourne J, Wilkins E, Thomas NS, Shea-Simonds J, Duncan PJ, Curtis CE, Robinson DO, Harvey JF, Cross NC. Dosage analysis of cancer predisposition genes by multiplex ligation-dependent probe amplification. Br J Cancer. 2004; 91: 11559. [PubMed]
Burn J, Chapman P, Delhanty J, Wood C, Lalloo F, Cachon-Gonzalez MB, Tsioupra K, Church W, Rhodes M, Gunn A. The UK Northern region genetic register for familial adenomatous polyposis coli: use of age of onset, congenital hypertrophy of the retinal pigment epithelium, and DNA markers in risk calculations. J Med Genet. 1991; 28: 28996. [PubMed]
Burt RW. Colon cancer screening. Gastroenterology. 2000; 119: 83753. [PubMed]
Burt RW. Gastric fundic gland polyps. Gastroenterology. 2003; 125: 14629. [PubMed]
Burt RW, Leppert MF, Slattery ML, Samowitz WS, Spirio LN, Kerber RA, Kuwada SK, Neklason DW, Disario JA, Lyon E, Hughes JP, Chey WY, White RL. Genetic testing and phenotype in a large kindred with attenuated familial adenomatous polyposis. Gastroenterology. 2004; 127: 44451. [PubMed]
Burt RW, Ward K, Spirio L. et al. Accurate identification of familial adenomatous polyposis coli using newly developed genetic markers. Gastroenterology. 1992; 102: A347.
Cao Y, Pieretti M, Marshall J, Khattar NH, Chen B, Kam-Morgan L, Lynch H. Challenge in the differentiation between attenuated familial adenomatous polyposis and hereditary nonpolyposis colorectal cancer: case report with review of the literature. Am J Gastroenterol. 2002; 97: 18227. [PubMed]
Caspari R, Olschwang S, Friedl W, Mandl M, Boisson C, Boker T, Augustin A, Kadmon M, Moslein G, Thomas G. et al. Familial adenomatous polyposis: desmoid tumours and lack of ophthalmic lesions (CHRPE) associated with APC mutations beyond codon 1444. Hum Mol Genet. 1995; 4: 33740. [PubMed]
Cetta F, Montalto G, Gori M, Curia MC, Cama A, Olschwang S. Germline mutations of the APC gene in patients with familial adenomatous polyposis-associated thyroid carcinoma: results from a European cooperative study. J Clin Endocrinol Metab. 2000; 85: 28692. [PubMed]
Chen CS, Phillips KD, Grist S, Bennet G, Craig JE, Muecke JS, Suthers GK. Congenital hypertrophy of the retinal pigment epithelium (CHRPE) in familial colorectal cancer. Fam Cancer. 2006; 5: 397404. [PubMed]
Chung DC. The genetic basis of colorectal cancer: insights into critical pathways of tumorigenesis. Gastroenterology. 2000; 119: 85465. [PubMed]
Church J, Burke C, McGannon E, Pastean O, Clark B. Risk of rectal cancer in patients after colectomy and ileorectal anastomosis for familial adenomatous polyposis: a function of available surgical options. Dis Colon Rectum. 2003a; 46: 117581. [PubMed]
Church J, Simmang C. Standards Task Force, American Society of Colon and Rectal Surgeons, Collaborative Group of the Americas on Inherited Colorectal Cancer and the Standards Committee of The American Society of Colon and Rectal Surgeons; Practice parameters for the treatment of patients with dominantly inherited colorectal cancer (familial adenomatous polyposis and hereditary nonpolyposis colorectal cancer). Dis Colon Rectum. 2003b; 46: 100112. [PubMed]
Church JM, McGannon E. Prior pregnancy ameliorates the course of intra-abdominal desmoid tumors in patients with familial adenomatous polyposis. Dis Colon Rectum. 2000; 43: 44550. [PubMed]
Clark SK, Phillips RK. Desmoids in familial adenomatous polyposis. Br J Surg. 1996; 83: 1494504. [PubMed]
Clark SK, Neale KF, Landgrebe JC, Phillips RK. Desmoid tumours complicating familial adenomatous polyposis. Br J Surg. 1999; 86: 11859. [PubMed]
Codori AM, Zawacki KL, Petersen GM, Miglioretti DL, Bacon JA, Trimbath JD, Booker SV, Picarello K, Giardiello FM. Genetic testing for hereditary colorectal cancer in children: long-term psychological effects. Am J Med Genet. 2003; 116A: 11728.
Couture J, Mitri A, Lagace R, Smits R, Berk T, Bouchard HL, Fodde R, Alman B, Bapat B. A germline mutation at the extreme 3' end of the APC gene results in a severe desmoid phenotype and is associated with overexpression of beta-catenin in the desmoid tumor. Clin Genet. 2000; 57: 20512. [PubMed]
Cromwell DM, Moore RD, Brensinger JD, Petersen GM, Bass EB, Giardiello FM. Cost analysis of alternative approaches to colorectal screening in familial adenomatous polyposis. Gastroenterology. 1998; 114: 893901. [PubMed]
Davies DR, Armstrong JG, Thakker N, Horner K, Guy SP, Clancy T, Sloan P, Blair V, Dodd C, Warnes TW. et al. Severe Gardner syndrome in families with mutations restricted to a specific region of the APC gene. Am J Hum Genet. 1995; 57: 11518. [PubMed]
Davis T, Song B, Cram DS. Preimplantation genetic diagnosis of familial adenomatous polyposis. Reprod Biomed Online. 2006; 13: 70711. [PubMed]
De Vos M, Hayward B, Bonthron DT, Sheridan E. Phenotype associated with recessively inherited mutations in DNA mismatch repair (MMR) genes. Biochem Soc Trans. 2005; 33: 71820. [PubMed]
Eccles DM, van der Luijt R, Breukel C, Bullman H, Bunyan D, Fisher A, Barber J, du Boulay C, Primrose J, Burn J, Fodde R. Hereditary desmoid disease due to a frameshift mutation at codon 1924 of the APC gene. Am J Hum Genet. 1996; 59: 1193201. [PubMed]
Etienne-Manneville S, Hall A. Cdc42 regulates GSK-3beta and adenomatous polyposis coli to control cell polarity. Nature. 2003; 421: 7536. [PubMed]
Evans DG, Guy SP, Thakker N, Armstrong JG, Dodd C, Davies DR, Babbs C, Clancy T, Warnes T, Sloan P. et al. Non-penetrance and late appearance of polyps in families with familial adenomatous polyposis. Gut. 1993; 34: 138993. [PubMed]
Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990; 61: 75967. [PubMed]
Felton KE, Gilchrist DM, Andrew SE. Constitutive deficiency in DNA mismatch repair: is it time for Lynch III? Clin Genet. 2007; 71: 499500. [PubMed]
Ferrández A, Pho L, Solomon C, Samowitz WS, Kuwada SK, Knecht TP, Gilfeather M, Burt RW. An evidence-based, multidisciplinary approach to the clinical considerations, management, and surveillance of adrenal lesions in familial adenomatous polyposis: report of three cases. Dis Colon Rectum. 2006; 49: 178190. [PubMed]
Fidder HH, Figer A, Geva R, Flex D, Schayek H, Avidan B, Meir SB, Friedman E. Genetic analyses in consecutive israeli jewish colorectal cancer patients. Am J Gastroenterol. 2005; 100: 137680. [PubMed]
Fodde R, Kuipers J, Rosenberg C, Smits R, Kielman M, Gaspar C, van Es JH, Breukel C, Wiegant J, Giles RH, Clevers H. Mutations in the APC tumour suppressor gene cause chromosomal instability. Nat Cell Biol. 2001; 3: 4338. [PubMed]
Frayling IM, Beck NE, Ilyas M, Dove-Edwin I, Goodman P, Pack K, Bell JA, Williams CB, Hodgson SV, Thomas HJ, Talbot IC, Bodmer WF, Tomlinson IP. The APC variants I1307K and E1317Q are associated with colorectal tumors, but not always with a family history. Proc Natl Acad Sci USA. 1998; 95: 107227. [PubMed]
Friedl W, Aretz S. Familial Adenomatous Polyposis: Experience from a Study of 1164 Unrelated German Polyposis Patients. Hereditary Cancer in Clinical Practice. 2005; 3: 95114.
Friedl W, Caspari R, Sengteller M, Uhlhaas S, Lamberti C, Jungck M, Kadmon M, Wolf M, Fahnenstich J, Gebert J, Moslein G, Mangold E, Propping P. Can APC mutation analysis contribute to therapeutic decisions in familial adenomatous polyposis? Experience from 680 FAP families. Gut. 2001; 48: 51521. [PubMed]
Friedl W, Meuschel S, Caspari R, Lamberti C, Krieger S, Sengteller M, Propping P. Attenuated familial adenomatous polyposis due to a mutation in the 3' part of the APC gene. A clue for understanding the function of the APC protein. Hum Genet. 1996; 97: 57984. [PubMed]
Gardner EJ, Richards RC. Multiple cutaneous and subcutaneous lesions occurring simultaneously with hereditary polyposis and osteomatosis. Am J Hum Genet. 1953; 5: 13947. [PubMed]
Garrean S, Hering J, Saied A, Jani J, Espat NJ. Gastric adenocarcinoma arising from fundic gland polyps in a patient with familial adenomatous polyposis syndrome. Am Surg. 2008; 74: 7983. [PubMed]
Gega M, Yanagi H, Yoshikawa R, Noda M, Ikeuchi H, Tsukamoto K, Oshima T, Fujiwara Y, Gondo N, Tamura K, Utsunomiya J, Hashimoto-Tamaoki T, Yamamura T. Successful chemotherapeutic modality of doxorubicin plus dacarbazine for the treatment of desmoid tumors in association with familial adenomatous polyposis. J Clin Oncol. 2006; 24: 1025. [PubMed]
Giardiello FM, Brensinger JD, Petersen GM. AGA technical review on hereditary colorectal cancer and genetic testing. Gastroenterology. 2001; 121: 198213. [PubMed]
Giardiello FM, Brensinger JD, Petersen GM, Luce MC, Hylind LM, Bacon JA, Booker SV, Parker RD, Hamilton SR. The use and interpretation of commercial APC gene testing for familial adenomatous polyposis. N Engl J Med. 1997; 336: 8237. [PubMed]
Giardiello FM, Brensinger JD, Tersmette AC, Goodman SN, Petersen GM, Booker SV, Cruz-Correa M, Offerhaus JA. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology. 2000; 119: 144753. [PubMed]
Giardiello FM, Hylind LM, Trimbath JD, Hamilton SR, Romans KE, Cruz-Correa M, Corretti MC, Offerhaus GJ, Yang VW. Oral contraceptives and polyp regression in familial adenomatous polyposis. Gastroenterology. 2005; 128: 107780. [PubMed]
Giardiello FM, Krush AJ, Petersen GM, Booker SV, Kerr M, Tong LL, Hamilton SR. Phenotypic variability of familial adenomatous polyposis in 11 unrelated families with identical APC gene mutation. Gastroenterology. 1994; 106: 15427. [PubMed]
Giardiello FM, Yang VW, Hylind LM, Krush AJ, Petersen GM, Trimbath JD, Piantadosi S, Garrett E, Geiman DE, Hubbard W, Offerhaus GJ, Hamilton SR. Primary chemoprevention of familial adenomatous polyposis with sulindac. N Engl J Med. 2002; 346: 10549. [PubMed]
Goss KH, Groden J. Biology of the adenomatous polyposis coli tumor suppressor. J Clin Oncol. 2000; 18: 196779. [PubMed]
Griffioen G, Bus PJ, Vasen HF, Verspaget HW, Lamers CB. Extracolonic manifestations of familial adenomatous polyposis: desmoid tumours, and upper gastrointestinal adenomas and carcinomas. Scand J Gastroenterol Suppl. 1998; 225: 8591. [PubMed]
Groves CJ, Beveridge G, Swain DJ, Saunders BP, Talbot IC, Nicholls RJ, Phillips RK. Prevalence and morphology of pouch and ileal adenomas in familial adenomatous polyposis. Dis Colon Rectum. 2005; 48: 81623. [PubMed]
Guerreiro CS, Cravo ML, Brito M, Vidal PM, Fidalgo PO, Leitão CN. The D1822V APC polymorphism interacts with fat, calcium, and fiber intakes in modulating the risk of colorectal cancer in Portuguese persons. Am J Clin Nutr. 2007; 85: 15927. [PubMed]
Guillem JG, Wood WC, Moley JF, Berchuck A, Karlan BY, Mutch DG, Gagel RF, Weitzel J, Morrow M, Weber BL, Giardiello F, Rodriguez-Bigas MA, Church J, Gruber S, Offit K. ASCO/SSO review of current role of risk-reducing surgery in common hereditary cancer syndromes. J Clin Oncol. 2006; 24: 464260. [PubMed]
Gurbuz AK, Giardiello FM, Petersen GM, Krush AJ, Offerhaus GJ, Booker SV, Kerr MC, Hamilton SR. Desmoid tumours in familial adenomatous polyposis. Gut. 1994; 35: 37781. [PubMed]
Hahnloser D, Petersen GM, Rabe K, Snow K, Lindor NM, Boardman L, Koch B, Doescher D, Wang L, Steenblock K, Thibodeau SN. The APC E1317Q variant in adenomatous polyps and colorectal cancers. Cancer Epidemiol Biomarkers Prev. 2003; 12: 10238. [PubMed]
Hamilton SR, Liu B, Parsons RE, Papadopoulos N, Jen J, Powell SM, Krush AJ, Berk T, Cohen Z, Tetu B. et al. The molecular basis of Turcot's syndrome. N Engl J Med. 1995; 332: 83947. [PubMed]
Heald B, Moran R, Milas M, Burke C, Eng C. Familial adenomatous polyposis in a patient with unexplained mental retardation. Nat Clin Pract Neurol. 2007; 3: 694700. [PubMed]
Heiskanen I, Luostarinen T, Jarvinen HJ. Impact of screening examinations on survival in familial adenomatous polyposis. Scand J Gastroenterol. 2000; 35: 12847. [PubMed]
Herraiz M, Barbesino G, Faquin W, Chan-Smutko G, Patel D, Shannon KM, Daniels GH, Chung DC. Prevalence of thyroid cancer in familial adenomatous polyposis syndrome and the role of screening ultrasound examinations. Gastroenterol Hepatol. 2007; 5: 36773.
Hes FJ, Nielsen M, Bik EC, Konvalinka D, Wijnen JT, Bakker E, Vasen HF, Breuning MH, Tops CM. Somatic APC mosaicism: an underestimated cause of polyposis coli. Gut. 2007; 57: 716. [PubMed]
Higuchi T, Iwama T, Yoshinaga K, Toyooka M, Taketo MM, Sugihara K. A randomized, double-blind, placebo-controlled trial of the effects of rofecoxib, a selective cyclooxygenase-2 inhibitor, on rectal polyps in familial adenomatous polyposis patients. Clin Cancer Res. 2003; 9: 475660. [PubMed]
Hirschman BA, Pollock BH, Tomlinson GE. The spectrum of APC mutations in children with hepatoblastoma from familial adenomatous polyposis kindreds. J Pediatr. 2004; 147: 2636. [PubMed]
Hofgartner WT, Thorp M, Ramus MW, Delorefice G, Chey WY, Ryan CK, Takahashi GW, Lobitz JR. Gastric adenocarcinoma associated with fundic gland polyps in a patient with attenuated familial adenomatous polyposis. Am J Gastroenterol. 1999; 94: 227581. [PubMed]
Hulsken J, Behrens J, Birchmeier W. Tumor-suppressor gene products in cell contacts: the cadherin-APC-armadillo connection. Curr Opin Cell Biol. 1994; 6: 7116. [PubMed]
Jaeger EE, Woodford-Richens KL, Lockett M, Rowan AJ, Sawyer EJ, Heinimann K, Rozen P, Murday VA, Whitelaw SC, Ginsberg A, Atkin WS, Lynch HT, Southey MC, Debinski H, Eng C, Bodmer WF, Talbot IC, Hodgson SV, Thomas HJ, Tomlinson IP. An ancestral Ashkenazi haplotype at the HMPS/CRAC1 locus on 15q13-q14 is associated with hereditary mixed polyposis syndrome. Am J Hum Genet. 2003; 72: 12617. [PubMed]
Jarvinen HJ. Epidemiology of familial adenomatous polyposis in Finland: impact of family screening on the colorectal cancer rate and survival. Gut. 1992; 33: 35760. [PubMed]
Jass JR, Williams CB, Bussey HJ, Morson BC. Juvenile polyposis--a precancerous condition. Histopathology. 1988; 13: 61930. [PubMed]
Johansen C, Bitsch M, Bülow S. Fertility and pregnancy in women with familial adenomatous polyposis. Int J Colorectal Dis. 1990; 5: 2036. [PubMed]
Joslyn G, Richardson DS, White R, Alber T. Dimer formation by an N-terminal coiled coil in the APC protein. Proc Natl Acad Sci USA. 1993; 90: 1110913. [PubMed]
Kadmon M, Tandara A, Herfarth C. Duodenal adenomatosis in familial adenomatous polyposis coli. A review of the literature and results from the Heidelberg Polyposis Register. Int J Colorectal Dis. 2001; 16: 6375. [PubMed]
Kaplan KB, Burds AA, Swedlow JR, Bekir SS, Sorger PK, Nathke IS. A role for the Adenomatous Polyposis Coli protein in chromosome segregation. Nat Cell Biol. 2001; 3: 42932. [PubMed]
Kastrinos F, Stoffel EM, Balmaña J, Syngal S. Attitudes toward prenatal genetic testing in patients with familial adenomatous polyposis. Am J Gastroenterol. 2007; 102: 128490. [PubMed]
Keller JJ, Giardiello FM. Chemoprevention strategies using NSAIDs and COX-2 inhibitors. Cancer Biol Ther. 2003; 2: S1409. [PubMed]
Knudsen AL, Bisgaard ML, Bülow S. Attenuated familial adenomatous polyposis (AFAP). A review of the literature. Fam Cancer. 2003; 2: 4355. [PubMed]
Korinek V, Barker N, Morin PJ, van Wichen D, de Weger R, Kinzler KW, Vogelstein B, Clevers H. Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC-/- colon carcinoma. Science. 1997; 275: 17847. [PubMed]
Laken SJ, Petersen GM, Gruber SB, Oddoux C, Ostrer H, Giardiello FM, Hamilton SR, Hampel H, Markowitz A, Klimstra D, Jhanwar S, Winawer S, Offit K, Luce MC, Kinzler KW, Vogelstein B. Familial colorectal cancer in Ashkenazim due to a hypermutable tract in APC. Nat Genet. 1997; 17: 7983. [PubMed]
Lamlum H, Al Tassan N, Jaeger E, Frayling I, Sieber O, Reza FB, Eckert M, Rowan A, Barclay E, Atkin W, Williams C, Gilbert J, Cheadle J, Bell J, Houlston R, Bodmer W, Sampson J, Tomlinson I. Germline APC variants in patients with multiple colorectal adenomas, with evidence for the particular importance of E1317Q. Hum Mol Genet. 2000; 9: 221521. [PubMed]
Lefevre JH, Rodrigue CM, Mourra N, Bennis M, Flejou JF, Parc R, Tiret E, Gespach C, Parc YR. Implication of MYH in colorectal polyposis. Ann Surg. 2006; 244: 8749. [PubMed]
Locker GY, Kaul K, Weinberg DS, Gatalica Z, Gong G, Peterman A, Lynch J, Klatzco L, Olopade OI, Bomzer CA, Newlin A, Keenan E, Tajuddin M, Knezetic J, Coronel S, Lynch HT. The I1307K APC polymorphism in Ashkenazi Jews with colorectal cancer: clinical and pathologic features. Cancer Genet Cytogenet. 2006; 169: 338. [PubMed]
Lynch HT, Smyrk TC, Watson P, Lanspa SJ, Lynch PM, Jenkins JX, Rouse J, Cavalieri J, Howard L, Lynch J. Hereditary flat adenoma syndrome: a variant of familial adenomatous polyposis? Dis Colon Rectum. 1992; 35: 41121. [PubMed]
Marchesa P, Fazio VW, Church JM, McGannon E. Adrenal masses in patients with familial adenomatous polyposis. Dis Colon Rectum. 1997; 40: 10238. [PubMed]
Matsumine A, Ogai A, Senda T, Okumura N, Satoh K, Baeg GH, Kawahara T, Kobayashi S, Okada M, Toyoshima K, Akiyama T. Binding of APC to the human homolog of the Drosophila discs large tumor suppressor protein. Science. 1996; 272: 10203. [PubMed]
Michie S, Collins V, Halliday J, Marteau TM. Likelihood of attending bowel screening after a negative genetic test result: the possible influence of health professionals. Genet Test. 2002; 6: 30711. [PubMed]
Michie S, Smith JA, Senior V, Marteau TM. Understanding why negative genetic test results sometimes fail to reassure. Am J Med Genet. 2003; 119A: 3407.
Michils G, Tejpar S, Fryns JP, Legius E, Van Cutsem E, Cassiman JJ, Matthijs G. Pathogenic mutations and rare variants of the APC gene identified in 75 Belgian patients with familial adenomatous polyposis by fluorescent enzymatic mutation detection (EMD). Eur J Hum Genet. 2002; 10: 50510. [PubMed]
Michils G, Tejpar S, Thoelen R, van Cutsem E, Vermeesch JR, Fryns JP, Legius E, Matthijs G. Large deletions of the APC gene in 15% of mutation-negative patients with classical polyposis (FAP): a Belgian study. Hum Mutat. 2005; 25: 12534. [PubMed]
Middleton SB, Clark SK, Matravers P, Katz D, Reznek R, Phillips RK. Stepwise progression of familial adenomatous polyposis-associated desmoid precursor lesions demonstrated by a novel CT scoring system. Dis Colon Rectum. 2003; 46: 4815. [PubMed]
Miyoshi Y, Nagase H, Ando H, Horii A, Ichii S, Nakatsuru S, Aoki T, Miki Y, Mori T, Nakamura Y. Somatic mutations of the APC gene in colorectal tumors: mutation cluster region in the APC gene. Hum Mol Genet. 1992; 1: 22933. [PubMed]
Morin PJ, Sparks AB, Korinek V, Barker N, Clevers H, Vogelstein B, Kinzler KW. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science. 1997; 275: 178790. [PubMed]
Moutou C, Gardes N, Nicod JC, Viville S. Strategies and outcomes of PGD of familial adenomatous polyposis. Mol Hum Reprod. 2007; 13: 95101. [PubMed]
Munemitsu S, Souza B, Muller O, Albert I, Rubinfeld B, Polakis P. The APC gene product associates with microtubules in vivo and promotes their assembly in vitro. Cancer Res. 1994; 54: 367681. [PubMed]
Nagase H, Miyoshi Y, Horii A, Aoki T, Ogawa M, Utsunomiya J, Baba S, Sasazuki T, Nakamura Y. Correlation between the location of germ-line mutations in the APC gene and the number of colorectal polyps in familial adenomatous polyposis patients. Cancer Res. 1992; 52: 40557. [PubMed]
Nakamura Y. Cleaning up on beta-catenin. Nat Med. 1997; 3: 499500. [PubMed]
Nathke IS, Adams CL, Polakis P, Sellin JH, Nelson WJ. The adenomatous polyposis coli tumor suppressor protein localizes to plasma membrane sites involved in active cell migration. J Cell Biol. 1996; 134: 16579. [PubMed]
Neklason DW, Stevens J, Boucher KM, Kerber RA, Matsunami N, Barlow J, Mineau G, Leppert MF, Burt RW. American founder mutation for attenuated familial adenomatous polyposis. Clin Gastroenterol Hepatol. 2008; 6: 4652. [PubMed]
Neufeld KL, White RL. Nuclear and cytoplasmic localizations of the adenomatous polyposis coli protein. Proc Natl Acad Sci USA. 1997; 94: 30349. [PubMed]
Nielsen M, Bik E, Hes FJ, Breuning MH, Vasen HF, Bakker E, Tops CM, Weiss MM. Genotype-phenotype correlations in 19 Dutch cases with APC gene deletions and a literature review. Eur J Hum Genet. 2007a; 15: 103442. [PubMed]
Nielsen M, Hes FJ, Nagengast FM, Weiss MM, Mathus-Vliegen EM, Morreau H, Breuning MH, Wijnen JT, Tops CM, Vasen HF. Germline mutations in APC and MUTYH are responsible for the majority of families with attenuated familial adenomatous polyposis. Clin Genet. 2007b; 71: 42733. [PubMed]
Nieuwenhuis MH, Vasen HF. Correlations between mutation site in APC and phenotype of familial adenomatous polyposis (FAP): a review of the literature. Crit Rev Oncol Hematol. 2007; 61: 15361. [PubMed]
Offerhaus GJ, Entius MM, Giardiello FM. Upper gastrointestinal polyps in familial adenomatous polyposis. Hepatogastroenterology. 1999; 46: 6679. [PubMed]
Olsen KO, Juul S, Bülow S, Jarvinen HJ, Bakka A, Bjork J, Oresland T, Laurberg S. Female fecundity before and after operation for familial adenomatous polyposis. Br J Surg. 2003; 90: 22731. [PubMed]
Ooi BS, Remzi FH, Gramlich T, Church JM, Preen M, Fazio VW. Anal transition zone cancer after restorative proctocolectomy and ileoanal anastomosis in familial adenomatous polyposis: report of two cases. Dis Colon Rectum. 2003; 46: 141823. [PubMed]
Paraf F, Jothy S, Van Meir EG. Brain tumor-polyposis syndrome: two genetic diseases? J Clin Oncol. 1997; 15: 274458. [PubMed]
Peifer M. Beta-catenin as oncogene: the smoking gun. Science. 1997; 275: 17523. [PubMed]
Petersen GM, Slack J, Nakamura Y. Screening guidelines and premorbid diagnosis of familial adenomatous polyposis using linkage. Gastroenterology. 1991; 100: 165864. [PubMed]
Pilarski RT, Brothman AR, Benn P, Shulman Rosengren S. Attenuated familial adenomatous polyposis in a man with an interstitial deletion of chromosome arm 5q. Am J Med Genet. 1999; 86: 3214. [PubMed]
Popat S, Stone J, Coleman G, Marshall G, Peto J, Frayling I, Houlston R. Prevalence of the APC E1317Q variant in colorectal cancer patients. Cancer Lett. 2000; 149: 2036. [PubMed]
Powell SM, Petersen GM, Krush AJ, Booker S, Jen J, Giardiello FM, Hamilton SR, Vogelstein B, Kinzler KW. Molecular diagnosis of familial adenomatous polyposis. N Engl J Med. 1993; 329: 19827. [PubMed]
Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW. APC mutations occur early during colorectal tumorigenesis. Nature. 1992; 359: 2357. [PubMed]
Pujol RM, Casanova JM, Egido R, Pujol J, de Moragas JM. Multiple familial pilomatricomas: a cutaneous marker for Gardner syndrome? Pediatr Dermatol. 1995; 12: 3315. [PubMed]
Rechitsky S, Verlinsky O, Chistokhina A, Sharapova T, Ozen S, Masciangelo C, Kuliev A, Verlinsky Y. Preimplantation genetic diagnosis for cancer predisposition. Reprod Biomed Online. 2002; 5: 14855. [PubMed]
Rennert G, Almog R, Tomsho LP, Low M, Pinchev M, Chaiter Y, Bonner JD, Rennert HS, Greenson JK, Gruber SB. Colorectal polyps in carriers of the APC I1307K polymorphism. Dis Colon Rectum. 2005; 48: 231721. [PubMed]
Ripa R, Bisgaard ML, Bülow S, Nielsen FC. De novo mutations in familial adenomatous polyposis (FAP). Eur J Hum Genet. 2002; 10: 6317. [PubMed]
Rozek LS, Rennert G, Gruber SB. APC E1317Q is not associated with Colorectal Cancer in a population-based case-control study in Northern Israel. Cancer Epidemiol Biomarkers Prev. 2006; 15: 23257. [PubMed]
Rozen P, Samuel Z, Shomrat R, Legum C. Notable intrafamilial phenotypic variability in a kindred with familial adenomatous polyposis and an APC mutation in exon 9. Gut. 1999; 45: 82933. [PubMed]
Rubinfeld B, Albert I, Porfiri E, Fiol C, Munemitsu S, Polakis P. Binding of GSK3beta to the APC-beta-catenin complex and regulation of complex assembly. Science. 1996; 272: 10236. [PubMed]
Rubinfeld B, Robbins P, El-Gamil M, Albert I, Porfiri E, Polakis P. Stabilization of beta-catenin by genetic defects in melanoma cell lines. Science. 1997; 275: 17902. [PubMed]
Rubinfeld B, Souza B, Albert I, Muller O, Chamberlain SH, Masiarz FR, Munemitsu S, Polakis P. Association of the APC gene product with beta-catenin. Science. 1993; 262: 17314. [PubMed]
Rubinfeld B, Souza B, Albert I, Munemitsu S, Polakis P. The APC protein and E-cadherin form similar but independent complexes with alpha-catenin, beta-catenin, and plakoglobin. J Biol Chem. 1995; 270: 554955. [PubMed]
Saurin JC, Chayvialle JA, Ponchon T. Management of duodenal adenomas in familial adenomatous polyposis. Endoscopy. 1999; 31: 4728. [PubMed]
Schwab AL, Tuohy TM, Condie M, Neklason DW, Burt RW (2007) Gonadal mosaicism and familial adenomatous polyposis. Fam Cancer published online Nov 18.
Scott RJ, Froggatt NJ, Trembath RC, Evans DG, Hodgson SV, Maher ER. Familial infiltrative fibromatosis (desmoid tumours) (MIM135290) caused by a recurrent 3' APC gene mutation. Hum Mol Genet. 1996; 5: 19214. [PubMed]
Sieber OM, Lamlum H, Crabtree MD, Rowan AJ, Barclay E, Lipton L, Hodgson S, Thomas HJW, Neale K, Phillips RKS, Farrington SM, Dunlop MG, Mueller HJ, Bisgaard ML, Bülow S, Fidalgo P, Albuquerque C, Scarano MI, Bodmer W, Tomlinson IPM, Heinimann K. Whole-gene APC deletions cause classical familial adnomatous polyposis, but not attenuated polyposis or "multiple" colorectal adenomas. PNAS. 2002; 99: 29548. [PubMed]
Sieber OM, Lipton L, Crabtree M, Heinimann K, Fidalgo P, Phillips RK, Bisgaard ML, Orntoft TF, Aaltonen LA, Hodgson SV, Thomas HJ, Tomlinson IP. Multiple colorectal adenomas, classic adenomatous polyposis, and germ- line mutations in MYH. N Engl J Med. 2003; 348: 7919. [PubMed]
Sieber OM, Segditsas S, Knudsen AL, Zhang J, Luz J, Rowan AJ, Spain SL, Thirlwell C, Howarth KM, Jaeger EE, Robinson J, Volikos E, Silver A, Kelly G, Aretz S, Frayling I, Hutter P, Dunlop M, Guenther T, Neale K, Phillips R, Heinimann K, Tomlinson IP. Disease severity and genetic pathways in attenuated familial adenomatous polyposis vary greatly but depend on the site of the germline mutation. Gut. 2006; 55: 14408. [PubMed]
Slattery ML, Samowitz W, Ballard L, Schaffer D, Leppert M, Potter JD. A molecular variant of the APC gene at codon 1822: its association with diet, lifestyle, and risk of colon cancer. Cancer Res. 2001; 61: 3222.
Smith AJ, Lewis JJ, Merchant NB, Leung DH, Woodruff JM, Brennan MF. Surgical management of intra-abdominal desmoid tumours. Br J Surg. 2000a; 87: 60813. [PubMed]
Smith KJ, Johnson KA, Bryan TM, Hill DE, Markowitz S, Willson JK, Paraskeva C, Petersen GM, Hamilton SR, Vogelstein B. et al. The APC gene product in normal and tumor cells. Proc Natl Acad Sci USA. 1993; 90: 284650. [PubMed]
Smith KJ, Levy DB, Maupin P, Pollard TD, Vogelstein B, Kinzler KW. Wild-type but not mutant APC associates with the microtubule cytoskeleton. Cancer Res. 1994; 54: 36725. [PubMed]
Smith TG, Clark SK, Katz DE, Reznek RH, Phillips RK. Adrenal masses are associated with familial adenomatous polyposis. Dis Colon Rectum. 2000b; 43: 173942. [PubMed]
Soravia C, Berk T, Madlensky L, Mitri A, Cheng H, Gallinger S, Cohen Z, Bapat B. Genotype-phenotype correlations in attenuated adenomatous polyposis coli. Am J Hum Genet. 1998; 62: 1290301. [PubMed]
Spigelman AD, Williams CB, Talbot IC, Domizio P, Phillips RK. Upper gastrointestinal cancer in patients with familial adenomatous polyposis. Lancet. 1989; 2: 7835. [PubMed]
Spirio L, Olschwang S, Groden J, Robertson M, Samowitz W, Joslyn G, Gelbert L, Thliveris A, Carlson M, Otterud B. et al. Alleles of the APC gene: an attenuated form of familial polyposis. Cell. 1993; 75: 9517. [PubMed]
Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB, Wakabayashi N, Saunders B, Shen Y, Fujimura T, Su LK, Levin B. The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med. 2000; 342: 194652. [PubMed]
Strul H, Barenboim E, Leshno M, Gartner M, Kariv R, Aljadeff E, Aljadeff Y, Kazanov D, Strier L, Keidar A, Knaani Y, Degani Y, Alon-Baron L, Sobol-Dvory H, Halpern Z, Arber N. The I1307K adenomatous polyposis coli gene variant does not contribute in the assessment of the risk for colorectal cancer in Ashkenazi Jews. Cancer Epidemiol Biomarkers Prev. 2003; 12: 10125. [PubMed]
Su LK, Burrell M, Hill DE, Gyuris J, Brent R, Wiltshire R, Trent J, Vogelstein B, Kinzler KW. APC binds to the novel protein EB1. Cancer Res. 1995; 55: 29727. [PubMed]
Su LK, Vogelstein B, Kinzler KW. Association of the APC tumor suppressor protein with catenins. Science. 1993; 262: 17347. [PubMed]
Suraweera N, Latchford A, McCart A, Rogers P, Spain S, Sieber O, Phillips R, Tomlinson I, Silver A. Pregnancy does not influence colonic polyp multiplicity but may modulate upper gastrointestinal disease in patients with FAP. Med Genet. 2007; 44: 5414.
Tan TY, Amor DJ. Tumour surveillance in Beckwith-Wiedemann syndrome and hemihyperplasia: a critical review of the evidence and suggested guidelines for local practice. J Paediatr Child Health. 2006; 42: 48690. [PubMed]
Tonelli F, Ficari F, Valanzano R, Brandi ML. Treatment of desmoids and mesenteric fibromatosis in familial adenomatous polyposis with raloxifene. Tumori. 2003; 89: 3916. [PubMed]
Truta B, Allen BA, Conrad PG, Kim YS, Berk T, Gallinger S, Bapat B, Terdiman JP, Sleisenger MH. Genotype and phenotype of patients with both familial adenomatous polyposis and thyroid carcinoma. Fam Cancer. 2003; 2: 959. [PubMed]
Truta B, Allen BA, Conrad PG, Weinberg V, Miller GA, Pomponio R, Lipton LR, Guerra G, Tomlinson IP, Sleisenger MH, Kim YS, Terdiman JP. A comparison of the phenotype and genotype in adenomatous polyposis patients with and without a family history. Fam Cancer. 2005; 4: 12733. [PubMed]
van der Luijt RB, Meera Khan P, Vasen HF, Breukel C, Tops CM, Scott RJ, Fodde R. Germline mutations in the 3' part of APC exon 15 do not result in truncated proteins and are associated with attenuated adenomatous polyposis coli. Hum Genet. 1996; 98: 72734. [PubMed]
van der Luijt RB, Vasen HF, Tops CM, Breukel C, Fodde R, Meera Khan P. APC mutation in the alternatively spliced region of exon 9 associated with late onset familial adenomatous polyposis. Hum Genet. 1995; 96: 70510. [PubMed]
Vasen HF, van der Luijt RB, Slors JF, Buskens E, de Ruiter P, Baeten CG, Schouten WR, Oostvogel HJ, Kuijpers JH, Tops CM, Meera Khan P. Molecular genetic tests as a guide to surgical management of familial adenomatous polyposis. Lancet. 1996; 348: 4335. [PubMed]
Wallace MH, Phillips RK. Upper gastrointestinal disease in patients with familial adenomatous polyposis. Br J Surg. 1998; 85: 74250. [PubMed]
Wallis YL, Morton DG, McKeown CM, Macdonald F. Molecular analysis of the APC gene in 205 families: extended genotype- phenotype correlations in FAP and evidence for the role of APC amino acid changes in colorectal cancer predisposition. J Med Genet. 1999; 36: 1420. [PubMed]
Walon C, Kartheuser A, Michils G, Smaers M, Lannoy N, Ngounou P, Mertens G, Verellen-Dumoulin C. Novel germline mutations in the APC gene and their phenotypic spectrum in familial adenomatous polyposis kindreds. Hum Genet. 1997; 100: 6015. [PubMed]
Wang L, Baudhuin LM, Boardman LA, Steenblock KJ, Petersen GM, Halling KC, French AJ, Johnson RA, Burgart LJ, Rabe K, Lindor NM, Thibodeau SN. MYH mutations in patients with attenuated and classic polyposis and with young-onset colorectal cancer without polyps. Gastroenterology. 2004; 127: 916. [PubMed]
Wehrli BM, Weiss SW, Yandow S, Coffin CM. Gardner-associated fibromas (GAF) in young patients: a distinct fibrous lesion that identifies unsuspected Gardner syndrome and risk for fibromatosis. Am J Surg Pathol. 2001; 25: 64551. [PubMed]
Wong RF, Tuteja AK, Haslem DS, Pappas L, Szabo A, Ogara MM, DiSario JA. Video capsule endoscopy compared with standard endoscopy for the evaluation of small-bowel polyps in persons with familial adenomatous polyposis (with video). Gastrointest Endosc. 2006; 64: 5307. [PubMed]
Zwick A, Munir M, Ryan CK, Gian J, Burt RW, Leppert M, Spirio L, Chey WY. Gastric adenocarcinoma and dysplasia in fundic gland polyps of a patient with attenuated adenomatous polyposis coli. Gastroenterology. 1997; 113: 65963. [PubMed]

Published Statements and Policies Regarding Genetic Testing

American Society of Clinical Oncology (2003) Policy statement update: genetic testing for cancer susceptibility.
American Society of Colon and Rectal Surgeons (2003) Practice parameters for the treatment of patients with dominantly inherited colorectal cancer (FAP and HNPCC).
American Society of Human Genetics/American College of Medical Genetics (1995) Points to consider: ethical, legal, and psychosocial implications of genetic testing in children and adolescents (Genetic Testing; pdf).
American Gastroenterological Association (2001) Medical position statement: hereditary colorectal cancer and genetic testing.
Giardiello FM, Brensinger JD, Petersen GM (2001) American Gastroenterological Association technical review on hereditary colorectal cancer and genetic testing.
American College of Medical Genetics/American Society of Human Genetics (2000) Joint statement on genetic testing for colon cancer (pdf).
Winawer S, Fletcher R, Rex D, Bond J, Burt R, Ferrucii J, Ganiats T, Levin T, Woolf S, Johnson D, Kirk L, Litin S, Simmang C for the U.S. Multisociety Task Force on Colorectal Cancer (2003) Colorectal cancer screening and surveillance: clinical guidelines and rationale - Update based on new evidence.

Suggested Reading

Bunz F, Kinzler KW, Vogelstein B. Colorectal tumors. In: Scriver CR, Beaudet AL, Sly WS, Valle D, Vogelstein B (eds) The Metabolic and Molecular Bases of Inherited Disease (OMMBID), McGraw-Hill, New York, Chap 48. Available at www.ommbid.com. Accessed 7-17-08.
Olschwang S (1998) Familial adenomatous polyposis (FAP). Atlas of Genetics and Cytogenetics Oncology and Haematology. atlasgeneticsoncology.org.

Chapter Notes

Author History

Randall W Burt, MD (1998-present)
Kory W Jasperson, MS (2008-present)
Cindy Solomon, MS; Myriad Genetic Laboratories (1998-2008)

Revision History

  • 24 July 2008 (me) Comprehensive update posted live

  • 21 October 2005 (me) Comprehensive update posted to live Web site

  • 20 September 2004 (chs) Revision: new clinical method

  • 27 May 2004 (chs) Revision: Genetic Counseling - genetic cancer subsection added

  • 15 March 2004 (me) Comprehensive update posted to live Web site

  • 23 June 2003 (cd) Revision: terminology

  • 18 January 2002 (me) Comprehensive update posted to live Web site

  • 18 December 1998 (pb) Review posted to live Web site

  • 11 September 1998 (ch) 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