Skip to main content
Erschienen in: Hereditary Cancer in Clinical Practice 1/2017

Open Access 01.12.2017 | Review

The genetic basis of colonic adenomatous polyposis syndromes

verfasst von: Bente A. Talseth-Palmer

Erschienen in: Hereditary Cancer in Clinical Practice | Ausgabe 1/2017

Abstract

Colorectal cancer (CRC) is one of the most common forms of cancer worldwide and familial adenomatous polyposis (FAP) accounts for approximately 1% of all CRCs. Adenomatous polyposis syndromes can be divided into; familial adenomatous polyposis (FAP) – classic FAP and attenuated familial adenomatous polyposis (AFAP), MUTYH-associated polyposis (MAP), NTHL1-associated polyposis (NAP) and polymerase proofreading-associated polyposis (PPAP). The polyposis syndromes genetics and clinical manifestation of disease varies and cases with clinical diagnosis of FAP might molecularly show a different diagnosis.
This review examines different aspects of the adenomatous polyposis syndromes genetics and clinical manifestation of disease; in addition the genotype-phenotype and modifier alleles of FAP will be discussed. New technology has made it possible to diagnose some of the APC mutation negative patients into their respective syndromes. There still remain many molecularly undiagnosed adenomatous polyposis patients indicating that there remain causative genes to be discovered and with today’s technology these are expected to be identified in the near future. The knowledge about the role of modifier alleles in FAP will contribute to improved pre-symptomatic diagnosis and treatment.
New novel mutations will continually be discovered in genes already associated with disease and new genes will be discovered that are associated with adenomatous polyposis. The search for modifier alleles in FAP should be made a priority.

Background

Colorectal cancer (CRC) is one of the most common form of cancer worldwide [1]. CRC development is considered to be a result of a combination of genetic and environmental factors and it is estimated that up to 35% of all CRCs are associated with a genetic predisposition [2]. Familial adenomatous polyposis (FAP) accounts for approximately 1% of all CRCs [3] and is an autosomal dominantly inherited condition where affected individuals develop hundreds to thousands of adenomas (polyposis) throughout the colon and rectum at unusually young ages. The disease is due to mutations in the adenomatous polyposis coli (APC) gene. If left untreated (if the colon is not removed), one or more of these adenomas invariably become malignant with almost 100% penetrance. Although prophylactic surgery significantly reduces the mortality associated with FAP, extra-colonic manifestations of the disease are now more clinically relevant, most notably desmoid tumours, which are hard to treat and a major cause of death [4, 5]. Attenuated FAP (AFAP) is a milder form of classic FAP with less polyps (<100) and a later age of polyp/cancer onset.
Herein an update on the genetic basis of FAP and other adenomatous polyposis syndromes (MAP, NAP and PPAP) is discussed. This review focuses on the genetics of FAP, the genetic phenotypes of the disease (genotype-phenotype correlations) and studies on modifier alleles. It also gives an update on other adenomatous polyposis syndromes. Recent findings are highlighted and gaps are identified in current literature, and consideration is give as to how these may be addressed through genomic approaches. A summary of the different adenomatous polyposis syndromes is shown in Table 1.
Table 1
Summary of adenomatous polyposis syndrome genetics, inheritance and clinical manifestation
Name
Abbreviation
Genetics
Inheritance
Clinical manifestation
Classical familial adenomatous polyposis
FAP
Germline APC mutations
Autosomal dominant
100–1000s colorectal polyps which manifests at age; early childhood-mid 30s (typically 16) and rapidly increasing. Almost 100% risk of CRC if left untreated.
Treatment recommendations, colectomy after adenomas emerges.
Associated with adenomatous polyps colon, CRC, fundic gland polyps, adenomatous polyps in the duodenum and periampullary region, extra intestinal lesions (fibromas, lipomas, sebaceous and epidermoid cysts = Gardner syndrome), desmoid tumours (benign soft-tissue tumours), congenital hypertrophy of the retinal pigment epithelium (CHRPE), and cancers of the brain (medulloblastoma = Turcot syndrome), pancreas, thyroid, gall bladder, bile duct and adrenal gland.
Attenuated familial adenomatous polyposis
AFAP
Germline APC mutations
Autosomal dominant
<100 colorectal polyps (typically 30) at age typically between 40 and 70 years (average 55). Estimated 70% CRC risk by age 80 years.
Treatment recommendations, colectomy may be necessary but for some polyps are limited enough in number that surveillance of colon is sufficient.
Associated with adenomatous polyps of the colon, CRC, upper gastrointestinal polyps, duodenal and gastric adenomas and fundic glad polyps. In addition, hepatoblastoma, gastric and breast adenocarcinoma have been documented.
MUTYH associated polyposis
MAP
Germline biallelic MUTYH mutations
Autosomal recessive
Usually < 100 polyps at average age of mid-50s and give a high risk of CRC.
Associated with malignancies of the duodenum, ovary, bladder and skin.
NTHL1 associated polyposis
NAP
Germline homozygous or compound heterozygous NTHL1 mutations
Autosomal recessive
Polyp number unknown as it is a recently discovered association but an extended spectrum of cancer diagnosis has been observed (CRC, endometrium, duodenum, skin, breast, pancreatic and others).
Multiple primary tumours in all patients.
Polymerase proofreading associated polyposis
PPAP
Germline POLE or POLD1 mutations
Autosomal dominant
Polyp number unknown, also recently discovered.
Associated with multi-tumour phenotypes like colon/pancreas/ovaries/small intestine and colon/ovarian/endometrial/brain.

Familial Adenomatous Polyposis (FAP) genetics

FAP is a result of germline mutations in APC [6, 7]. APC is a tumour suppressor gene that plays a central role in the Wnt signalling pathway. A detailed review of APC structure and function has been published by Half et al. [8]. In brief, APC is located on locus 5q21-22, consists of 15 coding exons (number of exons have increased to 18 after the identification of two promoter regions of APC [9]) and is 8532 bp in size which translates to a protein comprising 2843 amino acids [10]. Somatic mutations in APC is also a key molecular event in sporadic colorectal cancer present in about 80% of patients [10]. Two codons (1061 and 1309) are mutational hot-spots and account for 11 and 17% of all germline mutations, respectively [11] and are common sites of somatic change in sporadic CRC. But in a number of patients no underlying germline mutation can be identified [12, 13].
APC plays a central role in the Wnt-signalling pathway, especially in regards to the degradation of β-catenin within the cell cytoplasm. If APC is mutated, the β-catenin-Tcf complex is not suppressed and leads to constitutive activation of several genes and oncogenes controlling cell growth and division [10]. Mutations in APC affect the ability of the cell to maintain normal growth and function, which results in cell overgrowth/adenoma formation.
About 25% of people with FAP do not have any family history of disease and harbour a de novo mutation in APC without any clinical or genetic evidence of FAP in the family [1416]. One study suggests that a 5 bp deletion of codon 1309 (c.3927_3931del) is over-represented in patients with a suspected de novo mutations (29%) and in proven de novo mutation carriers (45%) [17], supporting the view of codon 1309 as a hot-spot for mutations.
New methods that can screen genomic loci at great depths are revealing that patients that were thought to be APC mutation negative have pathogenic germline heterozygous APC mutations [18], APC promoter mutations [9], deep intronic mutations [19], complex genomic rearrangements [20], somatic mutations or APC mutation mosaicism [12, 2123].

Classic FAP

Classic FAP (OMIM #175100) refers to patients who are diagnosed with FAP due to the development of more than 100 adenomatous colorectal polyps from early childhood (typically at age 16) who harbour an APC germline mutation. On average, cancer develops a decade after the appearance of adenomas and if the colon is left untreated most patients develop CRC by 40 years of age [8]. Other gastrointestinal manifestations include fundic gland polyps (which occurs in approximately 90% of FAP patients and are mostly benign [24]), adenomatous polyps in the duodenum and periampullary region (lifetime risk has been reported to reach 100% [25, 26]), and small bowel adenomas [8]. Extra-colonic manifestations are common but rarely malignant and include [8]; desmoid tumours (benign soft-tissue tumours that can be fatal due to progressive invasion into surrounding tissues [5, 27]), cutaneous lesions such as fibromas, lipomas, sebaceous and epidermoid cysts (present in Gardner syndrome [28], a phenotypic variant of FAP), congenital hypertrophy of the retinal pigment epithelium (CHRPE, which is a lesion causing discoloration in the ocular fundus – low-grade adenocarcinoma has been described in these lesions [29]), brain tumours (mainly medulloblastoma, described in Turcot’s syndrome, another phenotypic variant of FAP), hepatoblastoma, dental abnormalities, cancer of the pancreas, thyroid, gallbladder, bile duct and adrenal glands [8, 3032].

Attenuated familial adenomatous polyposis (AFAP)

AFAP is a phenotypic variant of FAP; patients develop less than 100 polyps, delayed polyp growth and later age of cancer onset. Germline APC mutations are also present in these patients, which are mainly observed in three sections of the gene (first 5 exons, exon 9 and in the distal 3′end of APC) [33]. Mean age of polyp diagnosis in AFAP patients is variable but on average in fourth to fifth decade of life, with cancer developing 10–15 years later [8, 34]. Screening is suggested to start late teens to mid-20s [34]. As with FAP the most common extra-colonic manifestations are upper gastrointestinal polyps, duodenal and gastric adenomas and fundic gland polyps [33]. Extra-colonic manifestations in AFAP are rare but hepatoblastoma, gastric and breast adenocarcinoma have been documented [8, 33].

FAP genotype-phenotype

In families with FAP, considerable variability in disease expression is observed within and between families harbouring identical APC mutations [13, 3537] and it has been shown that the greater the number of colorectal adenomas, the greater the risk of CRC [38]. It has been demonstrated that there is significant variation with respect to age of onset of intestinal symptoms and the development of CRC, even in patients with the same mutations [13]. Haplotype reconstruction from pedigrees have revealed there is no evidence for a specific APC haplotype associated with disease severity [39]. Genotype-phenotype correlations have been associated with the location of germline mutations within APC that are related to disease severity and the expression of extra-colonic disease [4042], see figures in Half et al. [8] and Macrae [10]. Patients with mutations in the mutation cluster region (MCR), located between codons 1286 and 1513 [43], have generally a worse prognosis with earlier disease onset than those with mutations outside this region [44]. Germline mutations at codon 1309 is associated with most severe disease [45], while milder forms with less than 100 adenomas and later ages of onset (AFAP) are associated with codons <157, 312–412 and >1595 [33, 41]. CHRPE has been associated with mutations between codons 457 and 1444 and susceptibility to desmoid tumours is correlated with mutations between codons 1395 and 2000, with slight variability in codon ranges between reports [8, 10, 46]. There is evidence of large phenotypic variation among patients with identical germline mutations [13], strongly suggesting the existence of FAP modifier alleles.

FAP modifier alleles

The phenotypic variation of APC Min mouse model of FAP reveal among different inbred strains the importance of modifier alleles [47]. There is evidence to suggest that these phenotypic differences are caused by segregating modifier alleles that impact adenoma number [47]. Several have been found in the min mouse model. The best known modifier is possibly Mom1 (modifier of Min 1), which is semi-dominant - each copy affects tumour multiplicity by a factor of approximately 2 [47]. Pla2g2a (found at the same region as Mom1) has also been shown to affect the net growth rate of adjacent tumours [48]. The exact mechanism by which it influences tumorigenesis remains unresolved [47] and the effort of linking PLA2G2A to FAP in humans has failed [49, 50], illustrating the difficult task of searching for modifier alleles in FAP. Many different genetic modifiers of the Apc knockout mouse models have been found, affecting karyotypic stability, DNA mutation rate, recombination rates, differentiation, DNA methylation, stromal regulation, cell growth and proliferation (reviewed in [47, 51]). There are claims that mouse models are essential in identifying modifiers of human disease and by using an Apc (Min/+) model have identified seven genes that are the most likely candidates for the Mom5 modifier [52]. Recently it has been reported that a new Xenopus tumour model might be especially useful for identifying or characterising modifier genes associated with APC mediated tumour formation [53].
Genome wide association studies (GWASs) have identified approximately 40 CRC susceptibility loci, where each loci gives a small increased risk of CRC [54]. The risk associated with each variant is too small on their own for translation to testing in clinical practice but the development of algorithms estimating cumulative risk are expected to lead to clinical application [54]. Two of these SNPs have been associated with Lynch syndrome [55, 56] and recently the same SNPs (rs16892766 and rs3802842) have been associated with adenoma number in APC mutation carriers causing a more sever FAP phenotype [57].
Several studies suggest that low-penetrant susceptibility genes may play an important role in the development of sporadic CRC [5863]. There is evidence to show that the variation in FAP severity (which have been shown to be independent of APC mutations and most likely the action of modifier alleles), is expected to result in different rates of adenoma number rather than differences in tumour progression [64]. Modifier genes can influence individual susceptibility to cancer by enhancing or suppressing disease initiation, growth and/or progression. The pattern of intra-familial variation in colonic FAP severity is consistent with the action of modifier genes [39, 6466]. As described above there is plenty of evidence from animal models for the existence of FAP modifiers and knowledge of modifier genes will contribute to better prophylactic measures for FAP patients [67]. It is important that the search for modifier genes/alleles continues.

Other adenomatous polyposis syndromes

Some of the recently discovered adenomatous polyposis syndromes are recessively inherited and present a diagnostic challenge. Individually, the other polyposis syndromes are very rare and may show overlapping phenotypes.

MUTYH-associated polyposis (MAP)

MAP (OMIM #608456) is an autosomal recessive disease caused by biallelic mutations in the base excision repair gene MUTYH. MUTYH is involved in base excision repair and is necessary in the amelioration of reactive oxygen species DNA damage prior to cell division [68]. In a recent study, 23% of APC mutation negative samples (FAP samples screened for APC mutations) were found to harbour pathogenic mutations in MUTYH [69]. Patients usually present with <100 colorectal polyps at an average age of disease diagnosis at around 50 years of age (which is similar to AFAP) and a high risk of CRC [69, 70]. The age of onset of polyposis has been shown to be significantly delayed for biallelic MUTYH carriers compared to APC mutation carriers [69]. MAP has been associated with malignancies of the duodenum, ovaries, urinary bladder and skin—occasionally resembling the phenotype of LS [71]. In a recent report describing extra-colonic disease, biallelic carriers are at high risk of urinary bladder and ovarian cancer, while there is some evidence that monoallelic carriers are at risk of gastric, hepatobiliary, endometrial and breast cancer [72]. No increased risk of other extra-colonic cancers associated with FAP was observed in this study [72].

NTHL1-associated polyposis (NAP)

A recently described autosomal recessive polyposis condition has been named NAP (OMIM #616415). Patients have germline homozygous or compound heterozygous mutations in the base excision repair gene NTHL1 [73]. Due to its recent discovery the clinical manifestation is not set, but it points towards an extended spectrum of cancer diagnosis in these patients; endometrial, duodenal, skin (basal cell carcinoma) and others [74]. Given such disease heterogeneity, Dutch researchers suggest it is a novel cancer syndrome. This is supported by Canadian researchers who also identified biallelic NTHL1 mutations in a woman with multiple primary tumours [75].

Polymerase proofreading-associated polyposis (PPAP)

PPAP is associated with mono- and biallelic mutations in the genes POLE and POLD1 [76], both genes a part of the mismatch repair (MMR) pathway. PPAP is in an autosomal dominantly inherited CRC predisposition [77]. Variants in POLE and POLD1 are known to increase the somatic mutation rate in tumours [78], thereby increasing the risk of tumour development. The somatic mutation landscape can display great diversity [79], which could be a reason for the differences observed in the location of primary tumours between patients. Both POLE and POLD1 have been associated with an increased risk of endometrial cancer [76, 80]. POLD1 has been associated with breast and brain tumours in addition to CRC and endometrial cancer [81]. Multi-tumour phenotypes such as colon/pancreas/ovaries/small intestine [82] and colon/ovarian/endometrial/brain [80] have been seen in POLE mutation carriers. In addition, POLE has been linked to an early onset cancer case raising the question whether this specific POLE mutation may confer a more severe phenotype than previously reported POLE/POLD1 mutations [83].

Conclusions

Genetic testing has rapidly grown in the last few years with the advancement of next-generation sequencing technologies. Targeted testing of all polyposis patients with a gene-panel can now be performed at reasonable cost such that targeted screening or prophylactic surgery can be offered to patients with a molecular diagnosis of polyposis.
In 2009 Half et al. [8] identified unresolved questions regarding FAP, one being that there are many FAP patients who do not get a molecular diagnosis. Since 2009, three additional genes have been associated with adenomatous polyposis, one being NTHL1 which has been classified as NAP, and POLE and POLD1 which has been classified as PPAP.
New novel mutations will continually be discovered in genes already associated with disease and new genes will be discovered that are associated with adenomatous polyposis. Exome sequencing has already been used to identify new candidate genes; PIEZ01 and ZSWiM7 [84], which are currently subject to further investigation.
A recent study has found that gene expression of APC was reduced in FAP patients without germline APC mutations [85]. An explanation may lie in differential epigenetic factors that contribute to the lack of gene expression in these patients, maybe more focus should be placed on understanding the role of epigenetics in polyposis syndromes.
There remain a high proportion of APC mutation negative patients even after extensive searches for new causative genes. The question remains, have we just missed them, or is it that these patients harbour rare alleles that await discovery. Diagnostics laboratories around the world are spending considerable amounts of time designing gene-panels to test for all adenomatous polyposis syndromes using new sequencing technology such that in the near future the number of APC mutation negative patients is expected to significantly decrease. Until then, re-testing “old” APC mutation negative patients for additional genes that have already been identified should be of special interest.

Acknowledgment

Thank you to Professor Rodney J. Scott for helping with the editing of the final draft of the manuscript.

Funding

This work was supported by the Liaison Committee between the Central Norway Regional Health Authority (RHA) and the Norwegian University of Science and Technology (NTNU), Norway; Møre og Romsdal Hospital Trust, Norway; and the Cancer Institute NSW, Australia.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

Author’s contributions

BTP has reviewed the literature and written the review.

Competing interests

The author declares that she has no competing interest.
Not applicable.
Not applicable.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Davidson NO. Genetic testing in colorectal cancer: who, when, how and why. Keio J Med. 2007;56(1):14–20.CrossRefPubMed Davidson NO. Genetic testing in colorectal cancer: who, when, how and why. Keio J Med. 2007;56(1):14–20.CrossRefPubMed
3.
4.
Zurück zum Zitat Clark SK, Phillips RK. Desmoids in familial adenomatous polyposis. Br J Surg. 1996;83(11):1494–504.CrossRefPubMed Clark SK, Phillips RK. Desmoids in familial adenomatous polyposis. Br J Surg. 1996;83(11):1494–504.CrossRefPubMed
6.
Zurück zum Zitat Groden J, Thliveris A, Samowitz W, Carlson M, Gelbert L, Albertsen H, et al. Identification and characterization of the familial adenomatous polyposis coli gene. Cell. 1991;66(3):589–600.CrossRefPubMed Groden J, Thliveris A, Samowitz W, Carlson M, Gelbert L, Albertsen H, et al. Identification and characterization of the familial adenomatous polyposis coli gene. Cell. 1991;66(3):589–600.CrossRefPubMed
7.
Zurück zum Zitat Kinzler KW, Nilbert MC, Su LK, Vogelstein B, Bryan TM, Levy DB, et al. Identification of FAP locus genes from chromosome 5q21. Science. 1991;253(5020):661–5.CrossRefPubMed Kinzler KW, Nilbert MC, Su LK, Vogelstein B, Bryan TM, Levy DB, et al. Identification of FAP locus genes from chromosome 5q21. Science. 1991;253(5020):661–5.CrossRefPubMed
9.
Zurück zum Zitat Rohlin A, Engwall Y, Fritzell K, Goransson K, Bergsten A, Einbeigi Z, et al. Inactivation of promoter 1B of APC causes partial gene silencing: evidence for a significant role of the promoter in regulation and causative of familial adenomatous polyposis. Oncogene. 2011;30(50):4977–89. doi:10.1038/onc.2011.201.CrossRefPubMedPubMedCentral Rohlin A, Engwall Y, Fritzell K, Goransson K, Bergsten A, Einbeigi Z, et al. Inactivation of promoter 1B of APC causes partial gene silencing: evidence for a significant role of the promoter in regulation and causative of familial adenomatous polyposis. Oncogene. 2011;30(50):4977–89. doi:10.​1038/​onc.​2011.​201.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Spier I, Drichel D, Kerick M, Kirfel J, Horpaopan S, Laner A et al. Low-level APC mutational mosaicism is the underlying cause in a substantial fraction of unexplained colorectal adenomatous polyposis cases. J Med Genet. 2015. doi:10.1136/jmedgenet-2015-103468. Spier I, Drichel D, Kerick M, Kirfel J, Horpaopan S, Laner A et al. Low-level APC mutational mosaicism is the underlying cause in a substantial fraction of unexplained colorectal adenomatous polyposis cases. J Med Genet. 2015. doi:10.​1136/​jmedgenet-2015-103468.
13.
Zurück zum Zitat Friedl W, Caspari R, Sengteller M, Uhlhaas S, Lamberti C, Jungck M, et al. Can APC mutation analysis contribute to therapeutic decisions in familial adenomatous polyposis? Experience from 680 FAP families. Gut. 2001;48(4):515–21.CrossRefPubMedPubMedCentral Friedl W, Caspari R, Sengteller M, Uhlhaas S, Lamberti C, Jungck M, et al. Can APC mutation analysis contribute to therapeutic decisions in familial adenomatous polyposis? Experience from 680 FAP families. Gut. 2001;48(4):515–21.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Gayther SA, Wells D, SenGupta SB, Chapman P, Neale K, Tsioupra K, et al. Regionally clustered APC mutations are associated with a severe phenotype and occur at a high frequency in new mutation cases of adenomatous polyposis coli. Hum Mol Genet. 1994;3(1):53–6.CrossRefPubMed Gayther SA, Wells D, SenGupta SB, Chapman P, Neale K, Tsioupra K, et al. Regionally clustered APC mutations are associated with a severe phenotype and occur at a high frequency in new mutation cases of adenomatous polyposis coli. Hum Mol Genet. 1994;3(1):53–6.CrossRefPubMed
16.
17.
18.
Zurück zum Zitat Out AA, van Minderhout IJ, van der Stoep N, van Bommel LS, Kluijt I, Aalfs C, et al. High-resolution melting (HRM) re-analysis of a polyposis patients cohort reveals previously undetected heterozygous and mosaic APC gene mutations. Fam Cancer. 2015;14(2):247–57. doi:10.1007/s10689-015-9780-5.CrossRefPubMedPubMedCentral Out AA, van Minderhout IJ, van der Stoep N, van Bommel LS, Kluijt I, Aalfs C, et al. High-resolution melting (HRM) re-analysis of a polyposis patients cohort reveals previously undetected heterozygous and mosaic APC gene mutations. Fam Cancer. 2015;14(2):247–57. doi:10.​1007/​s10689-015-9780-5.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Spier I, Horpaopan S, Vogt S, Uhlhaas S, Morak M, Stienen D, et al. Deep intronic APC mutations explain a substantial proportion of patients with familial or early-onset adenomatous polyposis. Hum Mutat. 2012;33(7):1045–50. doi:10.1002/humu.22082.CrossRefPubMed Spier I, Horpaopan S, Vogt S, Uhlhaas S, Morak M, Stienen D, et al. Deep intronic APC mutations explain a substantial proportion of patients with familial or early-onset adenomatous polyposis. Hum Mutat. 2012;33(7):1045–50. doi:10.​1002/​humu.​22082.CrossRefPubMed
20.
Zurück zum Zitat Charames GS, Ramyar L, Mitri A, Berk T, Cheng H, Jung J, et al. A large novel deletion in the APC promoter region causes gene silencing and leads to classical familial adenomatous polyposis in a Manitoba Mennonite kindred. Hum Genet. 2008;124(5):535–41. doi:10.1007/s00439-008-0579-4.CrossRefPubMed Charames GS, Ramyar L, Mitri A, Berk T, Cheng H, Jung J, et al. A large novel deletion in the APC promoter region causes gene silencing and leads to classical familial adenomatous polyposis in a Manitoba Mennonite kindred. Hum Genet. 2008;124(5):535–41. doi:10.​1007/​s00439-008-0579-4.CrossRefPubMed
22.
Zurück zum Zitat Aretz S, Stienen D, Friedrichs N, Stemmler S, Uhlhaas S, Rahner N, et al. Somatic APC mosaicism: a frequent cause of familial adenomatous polyposis (FAP). Hum Mutat. 2007;28(10):985–92. doi:10.1002/humu.20549.CrossRefPubMed Aretz S, Stienen D, Friedrichs N, Stemmler S, Uhlhaas S, Rahner N, et al. Somatic APC mosaicism: a frequent cause of familial adenomatous polyposis (FAP). Hum Mutat. 2007;28(10):985–92. doi:10.​1002/​humu.​20549.CrossRefPubMed
23.
25.
Zurück zum Zitat Bulow S, Bjork J, Christensen IJ, Fausa O, Jarvinen H, Moesgaard F, et al. Duodenal adenomatosis in familial adenomatous polyposis. Gut. 2004;53(3):381–6.CrossRefPubMedPubMedCentral Bulow S, Bjork J, Christensen IJ, Fausa O, Jarvinen H, Moesgaard F, et al. Duodenal adenomatosis in familial adenomatous polyposis. Gut. 2004;53(3):381–6.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Soravia C, Berk T, McLeod RS, Cohen Z. Desmoid disease in patients with familial adenomatous polyposis. Dis Colon Rectum. 2000;43(3):363–9.CrossRefPubMed Soravia C, Berk T, McLeod RS, Cohen Z. Desmoid disease in patients with familial adenomatous polyposis. Dis Colon Rectum. 2000;43(3):363–9.CrossRefPubMed
29.
Zurück zum Zitat Shields JA, Shields CL, Eagle Jr RC, Singh AD. Adenocarcinoma arising from congenital hypertrophy of retinal pigment epithelium. Arch Ophthalmol. 2001;119(4):597–602.CrossRefPubMed Shields JA, Shields CL, Eagle Jr RC, Singh AD. Adenocarcinoma arising from congenital hypertrophy of retinal pigment epithelium. Arch Ophthalmol. 2001;119(4):597–602.CrossRefPubMed
30.
Zurück zum Zitat Nilbert M, Kristoffersson U, Ericsson M, Johannsson O, Rambech E, Mangell P. Broad phenotypic spectrum in familial adenomatous polyposis; from early onset and severe phenotypes to late onset of attenuated polyposis with the first manifestation at age 72. BMC Med Genet. 2008;9:101. doi:10.1186/1471-2350-9-101.CrossRefPubMedPubMedCentral Nilbert M, Kristoffersson U, Ericsson M, Johannsson O, Rambech E, Mangell P. Broad phenotypic spectrum in familial adenomatous polyposis; from early onset and severe phenotypes to late onset of attenuated polyposis with the first manifestation at age 72. BMC Med Genet. 2008;9:101. doi:10.​1186/​1471-2350-9-101.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Knudsen AL, Bisgaard ML, Bulow S. Attenuated familial adenomatous polyposis (AFAP). A review of the literature. Fam Cancer. 2003;2(1):43–55.CrossRefPubMed Knudsen AL, Bisgaard ML, Bulow S. Attenuated familial adenomatous polyposis (AFAP). A review of the literature. Fam Cancer. 2003;2(1):43–55.CrossRefPubMed
35.
Zurück zum Zitat Giardiello FM, Krush AJ, Petersen GM, Booker SV, Kerr M, Tong LL, et al. Phenotypic variability of familial adenomatous polyposis in 11 unrelated families with identical APC gene mutation. Gastroenterology. 1994;106(6):1542–7.CrossRefPubMed Giardiello FM, Krush AJ, Petersen GM, Booker SV, Kerr M, Tong LL, et al. Phenotypic variability of familial adenomatous polyposis in 11 unrelated families with identical APC gene mutation. Gastroenterology. 1994;106(6):1542–7.CrossRefPubMed
36.
Zurück zum Zitat Brensinger JD, Laken SJ, Luce MC, Powell SM, Vance GH, Ahnen DJ, et al. Variable phenotype of familial adenomatous polyposis in pedigrees with 3′ mutation in the APC gene. Gut. 1998;43(4):548–52.CrossRefPubMedPubMedCentral Brensinger JD, Laken SJ, Luce MC, Powell SM, Vance GH, Ahnen DJ, et al. Variable phenotype of familial adenomatous polyposis in pedigrees with 3′ mutation in the APC gene. Gut. 1998;43(4):548–52.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Scott RJ, van der Luijt R, Spycher M, Mary JL, Muller A, Hoppeler T, et al. Novel germline APC gene mutation in a large familial adenomatous polyposis kindred displaying variable phenotypes. Gut. 1995;36(5):731–6.CrossRefPubMedPubMedCentral Scott RJ, van der Luijt R, Spycher M, Mary JL, Muller A, Hoppeler T, et al. Novel germline APC gene mutation in a large familial adenomatous polyposis kindred displaying variable phenotypes. Gut. 1995;36(5):731–6.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Debinski HS, Love S, Spigelman AD, Phillips RK. Colorectal polyp counts and cancer risk in familial adenomatous polyposis. Gastroenterology. 1996;110(4):1028–30.CrossRefPubMed Debinski HS, Love S, Spigelman AD, Phillips RK. Colorectal polyp counts and cancer risk in familial adenomatous polyposis. Gastroenterology. 1996;110(4):1028–30.CrossRefPubMed
39.
Zurück zum Zitat Crabtree MD, Fletcher C, Churchman M, Hodgson SV, Neale K, Phillips RK, et al. Analysis of candidate modifier loci for the severity of colonic familial adenomatous polyposis, with evidence for the importance of the N-acetyl transferases. Gut. 2004;53(2):271–6.CrossRefPubMedPubMedCentral Crabtree MD, Fletcher C, Churchman M, Hodgson SV, Neale K, Phillips RK, et al. Analysis of candidate modifier loci for the severity of colonic familial adenomatous polyposis, with evidence for the importance of the N-acetyl transferases. Gut. 2004;53(2):271–6.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Wachsmannova-Matelova L, Stevurkova V, Adamcikova Z, Holec V, Zajac V. Different phenotype manifestation of familial adenomatous polyposis in families with APC mutation at codon 1309. Neoplasma. 2009;56(6):486–9.CrossRefPubMed Wachsmannova-Matelova L, Stevurkova V, Adamcikova Z, Holec V, Zajac V. Different phenotype manifestation of familial adenomatous polyposis in families with APC mutation at codon 1309. Neoplasma. 2009;56(6):486–9.CrossRefPubMed
45.
Zurück zum Zitat Houlston R, Crabtree M, Phillips R, Tomlinson I. Explaining differences in the severity of familial adenomatous polyposis and the search for modifier genes. Gut. 2001;48(1):1–5.CrossRefPubMedPubMedCentral Houlston R, Crabtree M, Phillips R, Tomlinson I. Explaining differences in the severity of familial adenomatous polyposis and the search for modifier genes. Gut. 2001;48(1):1–5.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Bertario L, Russo A, Sala P, Varesco L, Giarola M, Mondini P, et al. Multiple approach to the exploration of genotype-phenotype correlations in familial adenomatous polyposis. J Clin Oncol. 2003;21(9):1698–707. doi:10.1200/JCO.2003.09.118.CrossRefPubMed Bertario L, Russo A, Sala P, Varesco L, Giarola M, Mondini P, et al. Multiple approach to the exploration of genotype-phenotype correlations in familial adenomatous polyposis. J Clin Oncol. 2003;21(9):1698–707. doi:10.​1200/​JCO.​2003.​09.​118.CrossRefPubMed
48.
Zurück zum Zitat Cormier RT, Dove WF. Dnmt1N/+ reduces the net growth rate and multiplicity of intestinal adenomas in C57BL/6-multiple intestinal neoplasia (Min)/+ mice independently of p53 but demonstrates strong synergy with the modifier of Min 1(AKR) resistance allele. Cancer Res. 2000;60(14):3965–70.PubMed Cormier RT, Dove WF. Dnmt1N/+ reduces the net growth rate and multiplicity of intestinal adenomas in C57BL/6-multiple intestinal neoplasia (Min)/+ mice independently of p53 but demonstrates strong synergy with the modifier of Min 1(AKR) resistance allele. Cancer Res. 2000;60(14):3965–70.PubMed
49.
Zurück zum Zitat Nimmrich I, Friedl W, Kruse R, Pietsch S, Hentsch S, Deuter R, et al. Loss of the PLA2G2A gene in a sporadic colorectal tumor of a patient with a PLA2G2A germline mutation and absence of PLA2G2A germline alterations in patients with FAP. Hum Genet. 1997;100(3–4):345–9.CrossRefPubMed Nimmrich I, Friedl W, Kruse R, Pietsch S, Hentsch S, Deuter R, et al. Loss of the PLA2G2A gene in a sporadic colorectal tumor of a patient with a PLA2G2A germline mutation and absence of PLA2G2A germline alterations in patients with FAP. Hum Genet. 1997;100(3–4):345–9.CrossRefPubMed
50.
Zurück zum Zitat Tomlinson IP, Beck NE, Neale K, Bodmer WF. Variants at the secretory phospholipase A2 (PLA2G2A) locus: analysis of associations with familial adenomatous polyposis and sporadic colorectal tumours. Ann Hum Genet. 1996;60(Pt 5):369–76.CrossRefPubMed Tomlinson IP, Beck NE, Neale K, Bodmer WF. Variants at the secretory phospholipase A2 (PLA2G2A) locus: analysis of associations with familial adenomatous polyposis and sporadic colorectal tumours. Ann Hum Genet. 1996;60(Pt 5):369–76.CrossRefPubMed
53.
Zurück zum Zitat Van Nieuwenhuysen T, Naert T, Tran HT, Van Imschoot G, Geurs S, Sanders E, et al. TALEN-mediated apc mutation in Xenopus tropicalis phenocopies familial adenomatous polyposis. Oncoscience. 2015;2(5):555–66.CrossRefPubMedPubMedCentral Van Nieuwenhuysen T, Naert T, Tran HT, Van Imschoot G, Geurs S, Sanders E, et al. TALEN-mediated apc mutation in Xenopus tropicalis phenocopies familial adenomatous polyposis. Oncoscience. 2015;2(5):555–66.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Talseth-Palmer BA, Brenne IS, Ashton KA, Evans TJ, McPhillips M, Groombridge C, et al. Colorectal cancer susceptibility loci on chromosome 8q23.3 and 11q23.1 as modifiers for disease expression in lynch syndrome. J Med Genet. 2011;48(4):279–84.CrossRefPubMed Talseth-Palmer BA, Brenne IS, Ashton KA, Evans TJ, McPhillips M, Groombridge C, et al. Colorectal cancer susceptibility loci on chromosome 8q23.3 and 11q23.1 as modifiers for disease expression in lynch syndrome. J Med Genet. 2011;48(4):279–84.CrossRefPubMed
56.
Zurück zum Zitat Wijnen JT, Brohet RM, van Eijk R, Jagmohan-Changur S, Middeldorp A, Tops CM, et al. Chromosome 8q23.3 and 11q23.1 variants modify colorectal cancer risk in Lynch syndrome. Gastroenterology. 2009;136(1):131–7.CrossRefPubMed Wijnen JT, Brohet RM, van Eijk R, Jagmohan-Changur S, Middeldorp A, Tops CM, et al. Chromosome 8q23.3 and 11q23.1 variants modify colorectal cancer risk in Lynch syndrome. Gastroenterology. 2009;136(1):131–7.CrossRefPubMed
57.
Zurück zum Zitat Ghorbanoghli Z, Nieuwenhuis MH, Houwing-Duistermaat JJ, Jagmohan-Changur S, Hes FJ, Tops CM et al. Colorectal cancer risk variants at 8q23.3 and 11q23.1 are associated with disease phenotype in APC mutation carriers. Fam Cancer. 2016. doi:10.1007/s10689-016-9877-5. Ghorbanoghli Z, Nieuwenhuis MH, Houwing-Duistermaat JJ, Jagmohan-Changur S, Hes FJ, Tops CM et al. Colorectal cancer risk variants at 8q23.3 and 11q23.1 are associated with disease phenotype in APC mutation carriers. Fam Cancer. 2016. doi:10.​1007/​s10689-016-9877-5.
58.
Zurück zum Zitat Jaeger E, Webb E, Howarth K, Carvajal-Carmona L, Rowan A, Broderick P, et al. Common genetic variants at the CRAC1 (HMPS) locus on chromosome 15q13.3 influence colorectal cancer risk. Nat Genet. 2008;40(1):26–8.CrossRefPubMed Jaeger E, Webb E, Howarth K, Carvajal-Carmona L, Rowan A, Broderick P, et al. Common genetic variants at the CRAC1 (HMPS) locus on chromosome 15q13.3 influence colorectal cancer risk. Nat Genet. 2008;40(1):26–8.CrossRefPubMed
59.
Zurück zum Zitat Tenesa A, Farrington SM, Prendergast JG, Porteous ME, Walker M, Haq N, et al. Genome-wide association scan identifies a colorectal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21. Nat Genet. 2008;40(5):631–7.CrossRefPubMedPubMedCentral Tenesa A, Farrington SM, Prendergast JG, Porteous ME, Walker M, Haq N, et al. Genome-wide association scan identifies a colorectal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21. Nat Genet. 2008;40(5):631–7.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Tomlinson IP, Webb E, Carvajal-Carmona L, Broderick P, Howarth K, Pittman AM, et al. A genome-wide association study identifies colorectal cancer susceptibility loci on chromosomes 10p14 and 8q23.3. Nat Genet. 2008;40(5):623–30.CrossRefPubMed Tomlinson IP, Webb E, Carvajal-Carmona L, Broderick P, Howarth K, Pittman AM, et al. A genome-wide association study identifies colorectal cancer susceptibility loci on chromosomes 10p14 and 8q23.3. Nat Genet. 2008;40(5):623–30.CrossRefPubMed
61.
Zurück zum Zitat Broderick P, Carvajal-Carmona L, Pittman AM, Webb E, Howarth K, Rowan A, et al. A genome-wide association study shows that common alleles of SMAD7 influence colorectal cancer risk. Nat Genet. 2007;39(11):1315–7.CrossRefPubMed Broderick P, Carvajal-Carmona L, Pittman AM, Webb E, Howarth K, Rowan A, et al. A genome-wide association study shows that common alleles of SMAD7 influence colorectal cancer risk. Nat Genet. 2007;39(11):1315–7.CrossRefPubMed
62.
Zurück zum Zitat Houlston RS, Cheadle J, Dobbins SE, Tenesa A, Jones AM, Howarth K, et al. Meta-analysis of three genome-wide association studies identifies susceptibility loci for colorectal cancer at 1q41, 3q26.2, 12q13.13 and 20q13.33. Nat Genet. 2010;42(11):973–7. doi:10.1038/ng.670.CrossRefPubMedPubMedCentral Houlston RS, Cheadle J, Dobbins SE, Tenesa A, Jones AM, Howarth K, et al. Meta-analysis of three genome-wide association studies identifies susceptibility loci for colorectal cancer at 1q41, 3q26.2, 12q13.13 and 20q13.33. Nat Genet. 2010;42(11):973–7. doi:10.​1038/​ng.​670.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Houlston RS, Webb E, Broderick P, Pittman AM, Di Bernardo MC, Lubbe S, et al. Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat Genet. 2008;40(12):1426–35. doi:10.1038/ng.262.CrossRefPubMed Houlston RS, Webb E, Broderick P, Pittman AM, Di Bernardo MC, Lubbe S, et al. Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat Genet. 2008;40(12):1426–35. doi:10.​1038/​ng.​262.CrossRefPubMed
64.
Zurück zum Zitat Crabtree MD, Tomlinson IP, Talbot IC, Phillips RK. Variability in the severity of colonic disease in familial adenomatous polyposis results from differences in tumour initiation rather than progression and depends relatively little on patient age. Gut. 2001;49(4):540–3.CrossRefPubMedPubMedCentral Crabtree MD, Tomlinson IP, Talbot IC, Phillips RK. Variability in the severity of colonic disease in familial adenomatous polyposis results from differences in tumour initiation rather than progression and depends relatively little on patient age. Gut. 2001;49(4):540–3.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Crabtree MD, Tomlinson IP, Hodgson SV, Neale K, Phillips RK, Houlston RS. Explaining variation in familial adenomatous polyposis: relationship between genotype and phenotype and evidence for modifier genes. Gut. 2002;51(3):420–3.CrossRefPubMedPubMedCentral Crabtree MD, Tomlinson IP, Hodgson SV, Neale K, Phillips RK, Houlston RS. Explaining variation in familial adenomatous polyposis: relationship between genotype and phenotype and evidence for modifier genes. Gut. 2002;51(3):420–3.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Dietrich WF, Lander ES, Smith JS, Moser AR, Gould KA, Luongo C, et al. Genetic identification of Mom-1, a major modifier locus affecting Min-induced intestinal neoplasia in the mouse. Cell. 1993;75(4):631–9.CrossRefPubMed Dietrich WF, Lander ES, Smith JS, Moser AR, Gould KA, Luongo C, et al. Genetic identification of Mom-1, a major modifier locus affecting Min-induced intestinal neoplasia in the mouse. Cell. 1993;75(4):631–9.CrossRefPubMed
67.
Zurück zum Zitat Ballhausen WG. Genetic testing for familial adenomatous polyposis. Ann N Y Acad Sci. 2000;910:36–47. discussion −9.CrossRefPubMed Ballhausen WG. Genetic testing for familial adenomatous polyposis. Ann N Y Acad Sci. 2000;910:36–47. discussion −9.CrossRefPubMed
69.
Zurück zum Zitat Papp J, Kovacs ME, Matrai Z, Orosz E, Kasler M, Borresen-Dale AL, et al. Contribution of APC and MUTYH mutations to familial adenomatous polyposis susceptibility in Hungary. Fam Cancer. 2016;15(1):85–97. doi:10.1007/s10689-015-9845-5.CrossRefPubMed Papp J, Kovacs ME, Matrai Z, Orosz E, Kasler M, Borresen-Dale AL, et al. Contribution of APC and MUTYH mutations to familial adenomatous polyposis susceptibility in Hungary. Fam Cancer. 2016;15(1):85–97. doi:10.​1007/​s10689-015-9845-5.CrossRefPubMed
72.
Zurück zum Zitat Win AK, Reece JC, Dowty JG, Buchanan DD, Clendenning M, Rosty C, et al. Risk of extracolonic cancers for people with biallelic and monoallelic mutations in MUTYH. Int J Cancer. 2016;139(7):1557–63. doi:10.1002/ijc.30197.CrossRefPubMed Win AK, Reece JC, Dowty JG, Buchanan DD, Clendenning M, Rosty C, et al. Risk of extracolonic cancers for people with biallelic and monoallelic mutations in MUTYH. Int J Cancer. 2016;139(7):1557–63. doi:10.​1002/​ijc.​30197.CrossRefPubMed
73.
Zurück zum Zitat Weren RD, Ligtenberg MJ, Kets CM, de Voer RM, Verwiel ET, Spruijt L, et al. A germline homozygous mutation in the base-excision repair gene NTHL1 causes adenomatous polyposis and colorectal cancer. Nat Genet. 2015;47(6):668–71. doi:10.1038/ng.3287.CrossRefPubMed Weren RD, Ligtenberg MJ, Kets CM, de Voer RM, Verwiel ET, Spruijt L, et al. A germline homozygous mutation in the base-excision repair gene NTHL1 causes adenomatous polyposis and colorectal cancer. Nat Genet. 2015;47(6):668–71. doi:10.​1038/​ng.​3287.CrossRefPubMed
74.
Zurück zum Zitat Kuiper RP, Hoogerbrugge N. NTHL1 defines novel cancer syndrome. Oncotarget. 2015;6(33):34069–70. 10.18632/oncotarget.5864.PubMedPubMedCentral Kuiper RP, Hoogerbrugge N. NTHL1 defines novel cancer syndrome. Oncotarget. 2015;6(33):34069–70. 10.18632/oncotarget.5864.PubMedPubMedCentral
76.
Zurück zum Zitat Palles C, Cazier JB, Howarth KM, Domingo E, Jones AM, Broderick P, et al. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat Genet. 2013;45(2):136–44. doi:10.1038/ng.2503.CrossRefPubMed Palles C, Cazier JB, Howarth KM, Domingo E, Jones AM, Broderick P, et al. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat Genet. 2013;45(2):136–44. doi:10.​1038/​ng.​2503.CrossRefPubMed
79.
Zurück zum Zitat Rashid M, Fischer A, Wilson CH, Tiffen J, Rust AG, Stevens P, et al. Adenoma development in familial adenomatous polyposis and MUTYH-associated polyposis: somatic landscape and driver genes. J Pathol. 2016;238(1):98–108. doi:10.1002/path.4643.CrossRefPubMed Rashid M, Fischer A, Wilson CH, Tiffen J, Rust AG, Stevens P, et al. Adenoma development in familial adenomatous polyposis and MUTYH-associated polyposis: somatic landscape and driver genes. J Pathol. 2016;238(1):98–108. doi:10.​1002/​path.​4643.CrossRefPubMed
83.
Zurück zum Zitat Wimmer K, Beilken A, Nustede R, Ripperger T, Lamottke B, Ure B, et al. A novel germline POLE mutation causes an early onset cancer prone syndrome mimicking constitutional mismatch repair deficiency. Fam Cancer. 2017;16(1):67–71. doi:10.1007/s10689-016-9925-1.CrossRefPubMed Wimmer K, Beilken A, Nustede R, Ripperger T, Lamottke B, Ure B, et al. A novel germline POLE mutation causes an early onset cancer prone syndrome mimicking constitutional mismatch repair deficiency. Fam Cancer. 2017;16(1):67–71. doi:10.​1007/​s10689-016-9925-1.CrossRefPubMed
84.
Zurück zum Zitat Spier I, Kerick M, Drichel D, Horpaopan S, Altmuller J, Laner A et al. Exome sequencing identifies potential novel candidate genes in patients with unexplained colorectal adenomatous polyposis. Fam Cancer. 2016. doi:10.1007/s10689-016-9870-z. Spier I, Kerick M, Drichel D, Horpaopan S, Altmuller J, Laner A et al. Exome sequencing identifies potential novel candidate genes in patients with unexplained colorectal adenomatous polyposis. Fam Cancer. 2016. doi:10.​1007/​s10689-016-9870-z.
85.
Metadaten
Titel
The genetic basis of colonic adenomatous polyposis syndromes
verfasst von
Bente A. Talseth-Palmer
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Hereditary Cancer in Clinical Practice / Ausgabe 1/2017
Elektronische ISSN: 1897-4287
DOI
https://doi.org/10.1186/s13053-017-0065-x

Weitere Artikel der Ausgabe 1/2017

Hereditary Cancer in Clinical Practice 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.