Skip to main content
Top
Published in: Familial Cancer 2/2013

01-06-2013 | Review

EPCAM deletion carriers constitute a unique subgroup of Lynch syndrome patients

Authors: Marjolijn J. L. Ligtenberg, Roland P. Kuiper, Ad Geurts van Kessel, Nicoline Hoogerbrugge

Published in: Familial Cancer | Issue 2/2013

Login to get access

Abstract

Lynch syndrome, one of the most common cancer susceptibility syndromes, is caused by germline mutations of genes affecting the mismatch repair proteins MLH1, MSH2, MSH6 or PMS2. Most of these mutations disrupt the open reading frame of the genes involved and, as such, lead to constitutive inactivation of the mutated allele. In a subset of Lynch syndrome patients MSH2 was found to be specifically inactivated in cell lineages exhibiting EPCAM expression. These patients carry deletions of the 3′ end of the EPCAM gene, including its polyadenylation signal. Due to concomitant transcriptional read-through of EPCAM, the promoter of MSH2 15 kb further downstream becomes inactivated through hypermethylation. As these 3′ EPCAM deletions occur in the germline, this MSH2 promoter methylation (‘epimutation’) is heritable. Worldwide, numerous EPCAM 3′ end deletions that differ in size and location have been detected. The risk of colorectal cancer in carriers of such EPCAM deletions is comparable to that of MSH2 mutation carriers, and is in accordance with a high expression of EPCAM in colorectal cancer stem cells. The risk of endometrial cancer in the entire group of EPCAM deletion carriers is significantly lower than that in MSH2 mutation carriers, but the actual risk appears to be dependent on the size and location of the EPCAM deletion. These observations may have important implications for the surveillance of EPCAM deletion carriers and, thus, calls for an in-depth assessment of clinically relevant genotype-phenotype correlations and its underlying molecular mechanism(s).
Literature
1.
go back to reference Lynch HT, Lynch PM, Lanspa SJ et al (2009) Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications. Clin Genet 76:1–18PubMedCrossRef Lynch HT, Lynch PM, Lanspa SJ et al (2009) Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications. Clin Genet 76:1–18PubMedCrossRef
2.
go back to reference Charbonnier F, Baert-Desurmont S, Liang P et al (2005) The 5′ region of the MSH2 gene involved in hereditary non-polyposis colorectal cancer contains a high density of recombinogenic sequences. Hum Mutat 26:255–261PubMedCrossRef Charbonnier F, Baert-Desurmont S, Liang P et al (2005) The 5′ region of the MSH2 gene involved in hereditary non-polyposis colorectal cancer contains a high density of recombinogenic sequences. Hum Mutat 26:255–261PubMedCrossRef
3.
go back to reference van der Klift H, Wijnen J, Wagner A et al (2005) Molecular characterization of the spectrum of genomic deletions in the mismatch repair genes MSH2, MLH1, MSH6, and PMS2 responsible for hereditary nonpolyposis colorectal cancer (HNPCC). Genes Chromosom Cancer 44:123–138PubMedCrossRef van der Klift H, Wijnen J, Wagner A et al (2005) Molecular characterization of the spectrum of genomic deletions in the mismatch repair genes MSH2, MLH1, MSH6, and PMS2 responsible for hereditary nonpolyposis colorectal cancer (HNPCC). Genes Chromosom Cancer 44:123–138PubMedCrossRef
4.
go back to reference Kovacs ME, Papp J, Szentirmay Z et al (2009) Deletions removing the last exon of TACSTD1 constitute a distinct class of mutations predisposing to Lynch syndrome. Hum Mutat 30:197–203PubMedCrossRef Kovacs ME, Papp J, Szentirmay Z et al (2009) Deletions removing the last exon of TACSTD1 constitute a distinct class of mutations predisposing to Lynch syndrome. Hum Mutat 30:197–203PubMedCrossRef
5.
go back to reference Ligtenberg MJL, Kuiper RP, Chan TL et al (2009) Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nat Genet 41:112–117PubMedCrossRef Ligtenberg MJL, Kuiper RP, Chan TL et al (2009) Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nat Genet 41:112–117PubMedCrossRef
6.
go back to reference Chan TL, Yuen ST, Kong CK et al (2006) Heritable germline epimutation of MSH2 in a family with hereditary nonpolyposis colorectal cancer. Nat Genet 38:1178–1183PubMedCrossRef Chan TL, Yuen ST, Kong CK et al (2006) Heritable germline epimutation of MSH2 in a family with hereditary nonpolyposis colorectal cancer. Nat Genet 38:1178–1183PubMedCrossRef
7.
go back to reference Tufarelli C, Stanley JA, Garrick D et al (2003) Transcription of antisense RNA leading to gene silencing and methylation as a novel cause of human genetic disease. Nat Genet 34:157–165PubMedCrossRef Tufarelli C, Stanley JA, Garrick D et al (2003) Transcription of antisense RNA leading to gene silencing and methylation as a novel cause of human genetic disease. Nat Genet 34:157–165PubMedCrossRef
8.
go back to reference Yu W, Gius D, Onyango P et al (2008) Epigenetic silencing of tumour suppressor gene p15 by its antisense RNA. Nature 451:202–206PubMedCrossRef Yu W, Gius D, Onyango P et al (2008) Epigenetic silencing of tumour suppressor gene p15 by its antisense RNA. Nature 451:202–206PubMedCrossRef
9.
go back to reference Venkatachalam R, Ligtenberg MJL, Hoogerbrugge N et al (2010) Germline epigenetic silencing of the tumor suppressor gene PTPRJ in early onset familial colorectal cancer. Gastroenterology 139:2221–2224PubMedCrossRef Venkatachalam R, Ligtenberg MJL, Hoogerbrugge N et al (2010) Germline epigenetic silencing of the tumor suppressor gene PTPRJ in early onset familial colorectal cancer. Gastroenterology 139:2221–2224PubMedCrossRef
10.
go back to reference Nagasaka T, Rhees J (2010) Kloor M et al Somatic hypermethylation of MSH2 is a frequent event in Lynch Syndrome colorectal cancers. Cancer Res 70:3098–3108PubMedCrossRef Nagasaka T, Rhees J (2010) Kloor M et al Somatic hypermethylation of MSH2 is a frequent event in Lynch Syndrome colorectal cancers. Cancer Res 70:3098–3108PubMedCrossRef
11.
go back to reference Niessen RC, Hofstra RMW, Westers H et al (2009) Germline hypermethylation of MLH1 and EPCAM deletions are a frequent cause of Lynch syndrome. Genes Chromosom Cancer 48:737–744PubMedCrossRef Niessen RC, Hofstra RMW, Westers H et al (2009) Germline hypermethylation of MLH1 and EPCAM deletions are a frequent cause of Lynch syndrome. Genes Chromosom Cancer 48:737–744PubMedCrossRef
12.
go back to reference Rumilla K, Schowalter KV, Kloor M et al (2011) Frequency of deletions of EPCAM (TACSTD1) in MSH2-associated Lynch syndrome cases. J Mol Diagn 13:93–99PubMedCrossRef Rumilla K, Schowalter KV, Kloor M et al (2011) Frequency of deletions of EPCAM (TACSTD1) in MSH2-associated Lynch syndrome cases. J Mol Diagn 13:93–99PubMedCrossRef
13.
go back to reference Hesson LB, Hitchins MP, Ward RL (2010) Epimutations and cancer predisposition: importance and mechanisms. Curr Opin Genet Dev 20:290–298PubMedCrossRef Hesson LB, Hitchins MP, Ward RL (2010) Epimutations and cancer predisposition: importance and mechanisms. Curr Opin Genet Dev 20:290–298PubMedCrossRef
14.
go back to reference Guarinos C, Castillejo A, Barberá VM et al (2010) EPCAM germ line deletions as causes of Lynch syndrome in Spanish patients. J Mol Diagn 12:765–770PubMedCrossRef Guarinos C, Castillejo A, Barberá VM et al (2010) EPCAM germ line deletions as causes of Lynch syndrome in Spanish patients. J Mol Diagn 12:765–770PubMedCrossRef
15.
go back to reference Kuiper RP, Vissers LE, Venkatachalam R et al (2011) Recurrence and variability of germline EPCAM deletions in Lynch syndrome. Hum Mutat 32:407–414PubMedCrossRef Kuiper RP, Vissers LE, Venkatachalam R et al (2011) Recurrence and variability of germline EPCAM deletions in Lynch syndrome. Hum Mutat 32:407–414PubMedCrossRef
16.
go back to reference Lynch HT, Riegert-Johnson DL, Snyder C et al (2011) Lynch syndrome-associated extracolonic tumors are rare in two extended families with the same EPCAM deletion. Am J Gastroenterol 106:1829–1836PubMedCrossRef Lynch HT, Riegert-Johnson DL, Snyder C et al (2011) Lynch syndrome-associated extracolonic tumors are rare in two extended families with the same EPCAM deletion. Am J Gastroenterol 106:1829–1836PubMedCrossRef
17.
go back to reference Grandval P, Baert-Desurmont S, Bonnet F et al (2012) Colon-specific phenotype in Lynch syndrome associated with EPCAM deletion. Clin Genet 82:97–99PubMedCrossRef Grandval P, Baert-Desurmont S, Bonnet F et al (2012) Colon-specific phenotype in Lynch syndrome associated with EPCAM deletion. Clin Genet 82:97–99PubMedCrossRef
18.
go back to reference Kloor M, Voigt AY, Schackert HK et al (2011) Analysis of EPCAM protein expression in diagnostics of Lynch syndrome. J Clin Oncol 29:223–227PubMedCrossRef Kloor M, Voigt AY, Schackert HK et al (2011) Analysis of EPCAM protein expression in diagnostics of Lynch syndrome. J Clin Oncol 29:223–227PubMedCrossRef
19.
go back to reference Huth C, Kloor M, Voigt AY et al (2012) The molecular basis of EPCAM expression loss in Lynch syndrome-associated tumors. Mod Pathol 25:911–916PubMedCrossRef Huth C, Kloor M, Voigt AY et al (2012) The molecular basis of EPCAM expression loss in Lynch syndrome-associated tumors. Mod Pathol 25:911–916PubMedCrossRef
20.
go back to reference Kloor M, Huth C, Voigt AY et al (2012) Prevalence of mismatch repair-deficient crypt foci in Lynch syndrome: a pathological study. Lancet Oncol 13:598–606PubMedCrossRef Kloor M, Huth C, Voigt AY et al (2012) Prevalence of mismatch repair-deficient crypt foci in Lynch syndrome: a pathological study. Lancet Oncol 13:598–606PubMedCrossRef
21.
go back to reference Munz M, Baeuerle PA, Gires O (2009) The emerging role of EpCAM in cancer and stem cell signaling. Cancer Res 69:5627–5629PubMedCrossRef Munz M, Baeuerle PA, Gires O (2009) The emerging role of EpCAM in cancer and stem cell signaling. Cancer Res 69:5627–5629PubMedCrossRef
22.
go back to reference Patriarca C, Macchi RM, Marschner AK et al (2012) Epithelial cell adhesion molecule expression (CD326) in cancer: a short review. Cancer Treat Rev 38:68–75PubMedCrossRef Patriarca C, Macchi RM, Marschner AK et al (2012) Epithelial cell adhesion molecule expression (CD326) in cancer: a short review. Cancer Treat Rev 38:68–75PubMedCrossRef
23.
go back to reference Gires O (2011) Lessons from common markers of tumor-initiating cells in solid cancers. Cell Mol Life Sci 68:4009–4022PubMedCrossRef Gires O (2011) Lessons from common markers of tumor-initiating cells in solid cancers. Cell Mol Life Sci 68:4009–4022PubMedCrossRef
24.
go back to reference Kempers MJ, Kuiper RP, Ockeloen CW et al (2011) Risk of colorectal and endometrial cancers in EPCAM deletion-positive Lynch syndrome: a cohort study. Lancet Oncol 12:49–55PubMedCrossRef Kempers MJ, Kuiper RP, Ockeloen CW et al (2011) Risk of colorectal and endometrial cancers in EPCAM deletion-positive Lynch syndrome: a cohort study. Lancet Oncol 12:49–55PubMedCrossRef
25.
go back to reference Sivagnanam M, Mueller JL, Lee H et al (2008) Identification of EpCAM as the gene for congenital tufting enteropathy. Gastroenterology 135:429–437PubMedCrossRef Sivagnanam M, Mueller JL, Lee H et al (2008) Identification of EpCAM as the gene for congenital tufting enteropathy. Gastroenterology 135:429–437PubMedCrossRef
26.
go back to reference Sivagnanam M, Schaible T, Szigeti R et al (2010) Further evidence for EpCAM as the gene for congenital tufting enteropathy. Am J Med Genet A 152A:222–224PubMedCrossRef Sivagnanam M, Schaible T, Szigeti R et al (2010) Further evidence for EpCAM as the gene for congenital tufting enteropathy. Am J Med Genet A 152A:222–224PubMedCrossRef
27.
go back to reference Salomon J, Espinosa-Parrilla Y, Goulet O et al (2011) A founder effect at the EPCAM locus in Congenital Tufting Enteropathy in the Arabic Gulf. Eur J Med Genet 54:319–322PubMedCrossRef Salomon J, Espinosa-Parrilla Y, Goulet O et al (2011) A founder effect at the EPCAM locus in Congenital Tufting Enteropathy in the Arabic Gulf. Eur J Med Genet 54:319–322PubMedCrossRef
Metadata
Title
EPCAM deletion carriers constitute a unique subgroup of Lynch syndrome patients
Authors
Marjolijn J. L. Ligtenberg
Roland P. Kuiper
Ad Geurts van Kessel
Nicoline Hoogerbrugge
Publication date
01-06-2013
Publisher
Springer Netherlands
Published in
Familial Cancer / Issue 2/2013
Print ISSN: 1389-9600
Electronic ISSN: 1573-7292
DOI
https://doi.org/10.1007/s10689-012-9591-x

Other articles of this Issue 2/2013

Familial Cancer 2/2013 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine