Skip to main content
Top
Published in: Familial Cancer 1/2014

01-03-2014 | Original Article

Lynch Syndrome in high risk Ashkenazi Jews in Israel

Authors: Yael Goldberg, Inbal Kedar, Revital Kariiv, Naama Halpern, Morasha Plesser, Ayala Hubert, Luna Kaduri, Michal Sagi, Israela Lerer, Dvorah Abeliovich, Tamar Hamburger, Aviram Nissan, Hanoch Goldshmidt, Irit Solar, Ravit Geva, Hana Strul, Guy Rosner, Hagit Baris, Zohar Levi, Tamar Peretz

Published in: Familial Cancer | Issue 1/2014

Login to get access

Abstract

Lynch Syndrome is caused by mutations in DNA mismatch repair genes. Diagnosis is not always trivial and may be costly. Information regarding incidence, genotype-phenotype correlation, spectrum of mutations and genes involved in specific populations facilitate the diagnostic process and contribute to clinical work-up. To report gene distribution, mutations detected and co-occurrence of related syndromes in a cohort of Ashkenazi Jews in Israel. Patients were identified in dedicated high risk clinics in 3 medical centers in Israel. Diagnostic process followed a multi-step scheme. It included testing for founder mutations, tumor testing, gene sequencing and MLPA. Lynch Syndrome was defined either by positive mutation testing, or by clinical criteria and positive tumor analysis. We report a cohort of 75 Ashkenazi families suspected of Lynch Syndrome. Mutations were identified in 51/75 (68 %) families: 38 in MSH2, 9 in MSH6, and 4 in MLH1. 37/51 (73 %) of these families carried one of the 3 ‘Ashkenazi’ founder mutations in MSH2 or MSH6. Each of the other 14 families carried a private mutation. 3 (6 %) were large deletions. Only 20/51 (39 %) families were Amsterdam Criteria positive; 42 (82 %) were positive for the Bethesda guidelines and 9 (18 %) did not fulfill any Lynch Syndrome criteria. We report C-MMRD and co-occurrence of BRCA and Lynch Syndrome in our cohort. Mutation spectra and gene distribution among Ashkenazi Jews are unique. Three founder Lynch Syndrome mutations are found in 73 % families with known mutations. Among the three, MSH2 and MSH6 are the most common. These features affect the phenotype, the diagnostic process, risk estimation, and genetic counseling.
Literature
1.
go back to reference Cunningham JM, Kim CY, Christensen ER et al (2001) The frequency of hereditary defective mismatch repair in a prospective series of unselected colorectal carcinomas. Am J Hum Genet 69:780–790PubMedCentralPubMedCrossRef Cunningham JM, Kim CY, Christensen ER et al (2001) The frequency of hereditary defective mismatch repair in a prospective series of unselected colorectal carcinomas. Am J Hum Genet 69:780–790PubMedCentralPubMedCrossRef
2.
go back to reference Hampel H, Frankel WL, Martin E et al (2008) Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol 26:5783–5788PubMedCrossRef Hampel H, Frankel WL, Martin E et al (2008) Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol 26:5783–5788PubMedCrossRef
3.
4.
go back to reference Berends MJ, Wu Y, Sijmons RH et al (2002) Molecular and clinical characteristics of MSH6 variants: an analysis of 25 index carriers of a germline variant. Am J Hum Genet 70:26–37PubMedCentralPubMedCrossRef Berends MJ, Wu Y, Sijmons RH et al (2002) Molecular and clinical characteristics of MSH6 variants: an analysis of 25 index carriers of a germline variant. Am J Hum Genet 70:26–37PubMedCentralPubMedCrossRef
5.
6.
go back to reference Truninger K, Menigatti M, Luz J et al (2005) Immunohistochemical analysis reveals high frequency of PMS2 defects in colorectal cancer. Gastroenterology 128:1160–1171PubMedCrossRef Truninger K, Menigatti M, Luz J et al (2005) Immunohistochemical analysis reveals high frequency of PMS2 defects in colorectal cancer. Gastroenterology 128:1160–1171PubMedCrossRef
7.
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
8.
go back to reference Rumilla K, Schowalter KV, Lindor NM et al (2011) Frequency of deletions of EPCAM (TACSTD1) in MSH2-associated Lynch syndrome cases. J Mol Diagn 13:93–99PubMedCentralPubMedCrossRef Rumilla K, Schowalter KV, Lindor NM et al (2011) Frequency of deletions of EPCAM (TACSTD1) in MSH2-associated Lynch syndrome cases. J Mol Diagn 13:93–99PubMedCentralPubMedCrossRef
9.
go back to reference Charbonnier F, Raux G, Wang Q et al (2000) Detection of exon deletions and duplications of the mismatch repair genes in hereditary nonpolyposis colorectal cancer families using multiplex polymerase chain reaction of short fluorescent fragments. Cancer Res 60:2760–2763PubMed Charbonnier F, Raux G, Wang Q et al (2000) Detection of exon deletions and duplications of the mismatch repair genes in hereditary nonpolyposis colorectal cancer families using multiplex polymerase chain reaction of short fluorescent fragments. Cancer Res 60:2760–2763PubMed
10.
go back to reference Plaschke J, Kruger S, Dietmaier W et al (2004) Eight novel MSH6 germline mutations in patients with familial and nonfamilial colorectal cancer selected by loss of protein expression in tumor tissue. Hum Mutat 23:285PubMedCrossRef Plaschke J, Kruger S, Dietmaier W et al (2004) Eight novel MSH6 germline mutations in patients with familial and nonfamilial colorectal cancer selected by loss of protein expression in tumor tissue. Hum Mutat 23:285PubMedCrossRef
11.
go back to reference Senter L, Clendenning M, Sotamaa K et al (2008) The clinical phenotype of Lynch syndrome due to germ-line PMS2 mutations. Gastroenterology 135:419–428PubMedCentralPubMedCrossRef Senter L, Clendenning M, Sotamaa K et al (2008) The clinical phenotype of Lynch syndrome due to germ-line PMS2 mutations. Gastroenterology 135:419–428PubMedCentralPubMedCrossRef
12.
go back to reference Wagner A, Barrows A, Wijnen JT et al (2003) Molecular analysis of hereditary nonpolyposis colorectal cancer in the United States: high mutation detection rate among clinically selected families and characterization of an American founder genomic deletion of the MSH2 gene. Am J Hum Genet 72:1088–1100PubMedCentralPubMedCrossRef Wagner A, Barrows A, Wijnen JT et al (2003) Molecular analysis of hereditary nonpolyposis colorectal cancer in the United States: high mutation detection rate among clinically selected families and characterization of an American founder genomic deletion of the MSH2 gene. Am J Hum Genet 72:1088–1100PubMedCentralPubMedCrossRef
13.
go back to reference Wijnen J, van der Klift H, Vasen H et al (1998) MSH2 genomic deletions are a frequent cause of HNPCC. Nat Genet 20:326–328PubMedCrossRef Wijnen J, van der Klift H, Vasen H et al (1998) MSH2 genomic deletions are a frequent cause of HNPCC. Nat Genet 20:326–328PubMedCrossRef
14.
go back to reference Baudhuin LM, Ferber MJ, Winters JL et al (2005) Characterization of hMLH1 and hMSH2 gene dosage alterations in Lynch syndrome patients. Gastroenterology 129:846–854PubMedCrossRef Baudhuin LM, Ferber MJ, Winters JL et al (2005) Characterization of hMLH1 and hMSH2 gene dosage alterations in Lynch syndrome patients. Gastroenterology 129:846–854PubMedCrossRef
15.
go back to reference Dove-Edwin I, Sasieni P, Adams J et al (2005) Prevention of colorectal cancer by colonoscopic surveillance in individuals with a family history of colorectal cancer: 16 year, prospective, follow-up study. BMJ 331:1047PubMedCrossRef Dove-Edwin I, Sasieni P, Adams J et al (2005) Prevention of colorectal cancer by colonoscopic surveillance in individuals with a family history of colorectal cancer: 16 year, prospective, follow-up study. BMJ 331:1047PubMedCrossRef
16.
go back to reference Jarvinen HJ, Aarnio M, Mustonen H et al (2000) Controlled 15-year trial on screening for colorectal cancer in families with hereditary nonpolyposis colorectal cancer. Gastroenterology 118:829–834PubMedCrossRef Jarvinen HJ, Aarnio M, Mustonen H et al (2000) Controlled 15-year trial on screening for colorectal cancer in families with hereditary nonpolyposis colorectal cancer. Gastroenterology 118:829–834PubMedCrossRef
17.
go back to reference Vasen HF, Abdirahman M, Brohet R et al (2010) One to 2-year surveillance intervals reduce risk of colorectal cancer in families with Lynch syndrome. Gastroenterology 138:2300–2306PubMedCrossRef Vasen HF, Abdirahman M, Brohet R et al (2010) One to 2-year surveillance intervals reduce risk of colorectal cancer in families with Lynch syndrome. Gastroenterology 138:2300–2306PubMedCrossRef
18.
go back to reference Vasen HF, de Vos tot Nederveen Cappel WH (2011) Cancer: Lynch syndrome–how should colorectal cancer be managed? Nat Rev Gastroenterol Hepatol 8:184–186PubMedCrossRef Vasen HF, de Vos tot Nederveen Cappel WH (2011) Cancer: Lynch syndrome–how should colorectal cancer be managed? Nat Rev Gastroenterol Hepatol 8:184–186PubMedCrossRef
19.
go back to reference Baglietto L, Lindor NM, Dowty JG et al (2010) Risks of Lynch syndrome cancers for MSH6 mutation carriers. J Natl Cancer Inst 102:193–201PubMedCrossRef Baglietto L, Lindor NM, Dowty JG et al (2010) Risks of Lynch syndrome cancers for MSH6 mutation carriers. J Natl Cancer Inst 102:193–201PubMedCrossRef
20.
go back to reference Hendriks YM, Wagner A, Morreau H et al (2004) Cancer risk in hereditary nonpolyposis colorectal cancer due to MSH6 mutations: impact on counseling and surveillance. Gastroenterology 127:17–25PubMedCrossRef Hendriks YM, Wagner A, Morreau H et al (2004) Cancer risk in hereditary nonpolyposis colorectal cancer due to MSH6 mutations: impact on counseling and surveillance. Gastroenterology 127:17–25PubMedCrossRef
21.
go back to reference Bonadona V, Bonaiti B, Olschwang S et al (2011) Cancer risks associated with germline mutations in MLH1, MSH2, and MSH6 genes in Lynch syndrome. JAMA 305:2304–2310PubMedCrossRef Bonadona V, Bonaiti B, Olschwang S et al (2011) Cancer risks associated with germline mutations in MLH1, MSH2, and MSH6 genes in Lynch syndrome. JAMA 305:2304–2310PubMedCrossRef
22.
go back to reference Moisio AL, Sistonen P, Weissenbach J et al (1996) Age and origin of two common MLH1 mutations predisposing to hereditary colon cancer. Am J Hum Genet 59:1243–1251PubMedCentralPubMed Moisio AL, Sistonen P, Weissenbach J et al (1996) Age and origin of two common MLH1 mutations predisposing to hereditary colon cancer. Am J Hum Genet 59:1243–1251PubMedCentralPubMed
23.
go back to reference Lynch HT, Coronel SM, Okimoto R et al (2004) A founder mutation of the MSH2 gene and hereditary nonpolyposis colorectal cancer in the United States. JAMA 291:718–724PubMedCrossRef Lynch HT, Coronel SM, Okimoto R et al (2004) A founder mutation of the MSH2 gene and hereditary nonpolyposis colorectal cancer in the United States. JAMA 291:718–724PubMedCrossRef
24.
go back to reference Foulkes WD, Thiffault I, Gruber SB et al (2002) The founder mutation MSH2*1906G–> C is an important cause of hereditary nonpolyposis colorectal cancer in the Ashkenazi Jewish population. Am J Hum Genet 71:1395–1412PubMedCentralPubMedCrossRef Foulkes WD, Thiffault I, Gruber SB et al (2002) The founder mutation MSH2*1906G–> C is an important cause of hereditary nonpolyposis colorectal cancer in the Ashkenazi Jewish population. Am J Hum Genet 71:1395–1412PubMedCentralPubMedCrossRef
25.
go back to reference Sun S, Greenwood CM, Thiffault I et al (2005) The HNPCC associated MSH2*1906G–> C founder mutation probably originated between 1,440 CE and 1,715 CE in the Ashkenazi Jewish population. J Med Genet 42:766–768PubMedCrossRef Sun S, Greenwood CM, Thiffault I et al (2005) The HNPCC associated MSH2*1906G–> C founder mutation probably originated between 1,440 CE and 1,715 CE in the Ashkenazi Jewish population. J Med Genet 42:766–768PubMedCrossRef
26.
go back to reference Goldberg Y, Porat RM, Kedar I et al (2010) An Ashkenazi founder mutation in the MSH6 gene leading to HNPCC. Fam Cancer 9:141–150PubMedCrossRef Goldberg Y, Porat RM, Kedar I et al (2010) An Ashkenazi founder mutation in the MSH6 gene leading to HNPCC. Fam Cancer 9:141–150PubMedCrossRef
27.
go back to reference Raskin L, Schwenter F, Freytsis M et al (2011) Characterization of two Ashkenazi Jewish founder mutations in MSH6 gene causing Lynch syndrome. Clin Genet 79:512–522PubMedCrossRef Raskin L, Schwenter F, Freytsis M et al (2011) Characterization of two Ashkenazi Jewish founder mutations in MSH6 gene causing Lynch syndrome. Clin Genet 79:512–522PubMedCrossRef
28.
go back to reference Goodfellow PJ, Buttin BM, Herzog TJ et al (2003) Prevalence of defective DNA mismatch repair and MSH6 mutation in an unselected series of endometrial cancers. Proc Natl Acad Sci USA 100:5908–5913PubMedCrossRef Goodfellow PJ, Buttin BM, Herzog TJ et al (2003) Prevalence of defective DNA mismatch repair and MSH6 mutation in an unselected series of endometrial cancers. Proc Natl Acad Sci USA 100:5908–5913PubMedCrossRef
30.
go back to reference Rodriguez-Bigas MA, Boland CR, Hamilton SR et al (1997) A National Cancer Institute workshop on hereditary nonpolyposis colorectal cancer syndrome: meeting highlights and Bethesda guidelines. J Natl Cancer Inst 89:1758–1762PubMedCrossRef Rodriguez-Bigas MA, Boland CR, Hamilton SR et al (1997) A National Cancer Institute workshop on hereditary nonpolyposis colorectal cancer syndrome: meeting highlights and Bethesda guidelines. J Natl Cancer Inst 89:1758–1762PubMedCrossRef
31.
go back to reference Umar A, Boland CR, Terdiman JP et al (2004) Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst 96:261–268PubMedCentralPubMedCrossRef Umar A, Boland CR, Terdiman JP et al (2004) Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst 96:261–268PubMedCentralPubMedCrossRef
32.
go back to reference Moreira L, Balaguer F, Lindor N et al (2012) Identification of Lynch syndrome among patients with colorectal cancer. JAMA 308:1555–1565PubMedCrossRef Moreira L, Balaguer F, Lindor N et al (2012) Identification of Lynch syndrome among patients with colorectal cancer. JAMA 308:1555–1565PubMedCrossRef
33.
go back to reference Wimmer K, Kratz CP (2010) Constitutional mismatch repair-deficiency syndrome. Haematologica 95:699–701PubMedCrossRef Wimmer K, Kratz CP (2010) Constitutional mismatch repair-deficiency syndrome. Haematologica 95:699–701PubMedCrossRef
34.
go back to reference Toledano H, Goldberg Y, Kedar-Barnes I et al (2009) Homozygosity of MSH2 c.1906G–> C germline mutation is associated with childhood colon cancer, astrocytoma and signs of Neurofibromatosis type I. Fam Cancer 8:187–194PubMedCrossRef Toledano H, Goldberg Y, Kedar-Barnes I et al (2009) Homozygosity of MSH2 c.1906G–> C germline mutation is associated with childhood colon cancer, astrocytoma and signs of Neurofibromatosis type I. Fam Cancer 8:187–194PubMedCrossRef
35.
go back to reference Banerjee SK, Makdisi WF, Weston AP et al (1995) Microwave-based DNA extraction from paraffin-embedded tissue for PCR amplification. Biotechniques 18:768–770, 772–773 Banerjee SK, Makdisi WF, Weston AP et al (1995) Microwave-based DNA extraction from paraffin-embedded tissue for PCR amplification. Biotechniques 18:768–770, 772–773
36.
go back to reference Goldberg Y, Porat RM, Kedar I et al (2008) Mutation spectrum in HNPCC in the Israeli population. Fam Cancer 7:309–317PubMedCrossRef Goldberg Y, Porat RM, Kedar I et al (2008) Mutation spectrum in HNPCC in the Israeli population. Fam Cancer 7:309–317PubMedCrossRef
37.
go back to reference Shiri-Sverdlov R, Oefner P, Green L et al (2000) Mutational analyses of BRCA1 and BRCA2 in Ashkenazi and non-Ashkenazi Jewish women with familial breast and ovarian cancer. Hum Mutat 16:491–501PubMedCrossRef Shiri-Sverdlov R, Oefner P, Green L et al (2000) Mutational analyses of BRCA1 and BRCA2 in Ashkenazi and non-Ashkenazi Jewish women with familial breast and ovarian cancer. Hum Mutat 16:491–501PubMedCrossRef
38.
go back to reference Struewing JP, Hartge P, Wacholder S et al (1997) The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 336:1401–1408PubMedCrossRef Struewing JP, Hartge P, Wacholder S et al (1997) The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 336:1401–1408PubMedCrossRef
39.
go back to reference Palomaki GE, McClain MR, Melillo S et al (2009) EGAPP supplementary evidence review: DNA testing strategies aimed at reducing morbidity and mortality from Lynch syndrome. Genet Med 11:42–65PubMedCentralPubMedCrossRef Palomaki GE, McClain MR, Melillo S et al (2009) EGAPP supplementary evidence review: DNA testing strategies aimed at reducing morbidity and mortality from Lynch syndrome. Genet Med 11:42–65PubMedCentralPubMedCrossRef
40.
go back to reference Shia J (2008) Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part I. The utility of immunohistochemistry. J Mol Diagn 10:293–300PubMedCentralPubMedCrossRef Shia J (2008) Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part I. The utility of immunohistochemistry. J Mol Diagn 10:293–300PubMedCentralPubMedCrossRef
41.
go back to reference Shia J, Holck S, Depetris G et al (2013) Lynch syndrome-associated neoplasms: a discussion on histopathology and immunohistochemistry. Fam Cancer 12(2):241–260 Shia J, Holck S, Depetris G et al (2013) Lynch syndrome-associated neoplasms: a discussion on histopathology and immunohistochemistry. Fam Cancer 12(2):241–260
42.
go back to reference Radu OM, Nikiforova MN, Farkas LM et al (2011) Challenging cases encountered in colorectal cancer screening for Lynch syndrome reveal novel findings: nucleolar MSH6 staining and impact of prior chemoradiation therapy. Hum Pathol 42:1247–1258PubMedCrossRef Radu OM, Nikiforova MN, Farkas LM et al (2011) Challenging cases encountered in colorectal cancer screening for Lynch syndrome reveal novel findings: nucleolar MSH6 staining and impact of prior chemoradiation therapy. Hum Pathol 42:1247–1258PubMedCrossRef
43.
go back to reference Nilbert M, Wikman FP, Hansen TV et al (2009) Major contribution from recurrent alterations and MSH6 mutations in the Danish Lynch syndrome population. Fam Cancer 8:75–83PubMedCrossRef Nilbert M, Wikman FP, Hansen TV et al (2009) Major contribution from recurrent alterations and MSH6 mutations in the Danish Lynch syndrome population. Fam Cancer 8:75–83PubMedCrossRef
44.
go back to reference Kast K, Neuhann TM, Gorgens H et al (2012) Germline truncating-mutations in BRCA1 and MSH6 in a patient with early onset endometrial cancer. BMC Cancer 12:531PubMedCentralPubMedCrossRef Kast K, Neuhann TM, Gorgens H et al (2012) Germline truncating-mutations in BRCA1 and MSH6 in a patient with early onset endometrial cancer. BMC Cancer 12:531PubMedCentralPubMedCrossRef
45.
46.
go back to reference Raevaara TE, Korhonen MK, Lohi H et al (2005) Functional significance and clinical phenotype of nontruncating mismatch repair variants of MLH1. Gastroenterology 129:537–549 Raevaara TE, Korhonen MK, Lohi H et al (2005) Functional significance and clinical phenotype of nontruncating mismatch repair variants of MLH1. Gastroenterology 129:537–549
47.
go back to reference Liu B, Parsons R, Papadopoulos N et al (1996) Analysis of mismatch repair genes in hereditary non-polyposis colorectal cancer patients. Nat Med 2:169–174 Liu B, Parsons R, Papadopoulos N et al (1996) Analysis of mismatch repair genes in hereditary non-polyposis colorectal cancer patients. Nat Med 2:169–174
48.
go back to reference Miyaki M, Konishi M, Muraoka M et al (1995) Germ line mutations of hMSH2 and hMLH1 genes in Japanese families with hereditary nonpolyposis colorectal cancer (HNPCC): usefulness of DNA analysis for screening and diagnosis of HNPCC patients. J Mol Med (Berl) 73:515–520 Miyaki M, Konishi M, Muraoka M et al (1995) Germ line mutations of hMSH2 and hMLH1 genes in Japanese families with hereditary nonpolyposis colorectal cancer (HNPCC): usefulness of DNA analysis for screening and diagnosis of HNPCC patients. J Mol Med (Berl) 73:515–520
49.
go back to reference Taylor CF, Charlton RS, Burn J et al (2003) Genomic deletions in MSH2 or MLH1 are a frequent cause of hereditary non-polyposis colorectal cancer: identification of novel and recurrent deletions by MLPA. Hum Mutat 22:428–433 Taylor CF, Charlton RS, Burn J et al (2003) Genomic deletions in MSH2 or MLH1 are a frequent cause of hereditary non-polyposis colorectal cancer: identification of novel and recurrent deletions by MLPA. Hum Mutat 22:428–433
50.
go back to reference Pritchard CC, Smith C, Salipante SJ et al (2012) ColoSeq provides comprehensive lynch and polyposis syndrome mutational analysis using massively parallel sequencing. J Mol Diagn 14:357–366 Pritchard CC, Smith C, Salipante SJ et al (2012) ColoSeq provides comprehensive lynch and polyposis syndrome mutational analysis using massively parallel sequencing. J Mol Diagn 14:357–366
51.
go back to reference Stella A, Surdo NC, Lastella P et al (2007) Germline novel MSH2 deletions and a founder MSH2 deletion associated with anticipation effects in HNPCC. Clin Genet 71:130–139 Stella A, Surdo NC, Lastella P et al (2007) Germline novel MSH2 deletions and a founder MSH2 deletion associated with anticipation effects in HNPCC. Clin Genet 71:130–139
52.
go back to reference Wahlberg SS, Nystrom-Lahti M, Kane MF et al (1997) Low frequency of hMSH2 mutations in Swedish HNPCC families. Int J Cancer 74:134–137 Wahlberg SS, Nystrom-Lahti M, Kane MF et al (1997) Low frequency of hMSH2 mutations in Swedish HNPCC families. Int J Cancer 74:134–137
53.
go back to reference Glasl S, Papatheodorou L, Baretton G et al (2000) Novel germline mutation (300-305delAGTTGA) in the human MSH2 gene in hereditary non-polyposis colorectal cancer (HNPCC). Hum Mutat 16:91–92 Glasl S, Papatheodorou L, Baretton G et al (2000) Novel germline mutation (300-305delAGTTGA) in the human MSH2 gene in hereditary non-polyposis colorectal cancer (HNPCC). Hum Mutat 16:91–92
54.
go back to reference Buerstedde JM, Alday P, Torhorst J et al (1995) Detection of new mutations in six out of 10 Swiss HNPCC families by genomic sequencing of the hMSH2 and hMLH1 genes. J Med Genet 32:909–912 Buerstedde JM, Alday P, Torhorst J et al (1995) Detection of new mutations in six out of 10 Swiss HNPCC families by genomic sequencing of the hMSH2 and hMLH1 genes. J Med Genet 32:909–912
55.
go back to reference Zhou JN, Wang DQ, Song L et al (2010) [Association of IVS10+12G>A polymorphism in hMSH2 gene with colorectal cancer in Chinese]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 27:579–583 Zhou JN, Wang DQ, Song L et al (2010) [Association of IVS10+12G>A polymorphism in hMSH2 gene with colorectal cancer in Chinese]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 27:579–583
56.
go back to reference Wijnen J, Khan PM, Vasen H et al (1997) Hereditary nonpolyposis colorectal cancer families not complying with the Amsterdam criteria show extremely low frequency of mismatch-repair-gene mutations. Am J Hum Genet 61:329–335 Wijnen J, Khan PM, Vasen H et al (1997) Hereditary nonpolyposis colorectal cancer families not complying with the Amsterdam criteria show extremely low frequency of mismatch-repair-gene mutations. Am J Hum Genet 61:329–335
57.
go back to reference Gammie AE, Erdeniz N, Beaver J et al (2007) Functional characterization of pathogenic human MSH2 missense mutations in Saccharomyces cerevisiae. Genetics 177:707–721 Gammie AE, Erdeniz N, Beaver J et al (2007) Functional characterization of pathogenic human MSH2 missense mutations in Saccharomyces cerevisiae. Genetics 177:707–721
58.
go back to reference Thompson BA, Greenblatt MS, Vallee MP et al (2013) Calibration of multiple in silico tools for predicting pathogenicity of mismatch repair gene missense substitutions. Hum Mutat 34:255–265PubMedCrossRef Thompson BA, Greenblatt MS, Vallee MP et al (2013) Calibration of multiple in silico tools for predicting pathogenicity of mismatch repair gene missense substitutions. Hum Mutat 34:255–265PubMedCrossRef
Metadata
Title
Lynch Syndrome in high risk Ashkenazi Jews in Israel
Authors
Yael Goldberg
Inbal Kedar
Revital Kariiv
Naama Halpern
Morasha Plesser
Ayala Hubert
Luna Kaduri
Michal Sagi
Israela Lerer
Dvorah Abeliovich
Tamar Hamburger
Aviram Nissan
Hanoch Goldshmidt
Irit Solar
Ravit Geva
Hana Strul
Guy Rosner
Hagit Baris
Zohar Levi
Tamar Peretz
Publication date
01-03-2014
Publisher
Springer Netherlands
Published in
Familial Cancer / Issue 1/2014
Print ISSN: 1389-9600
Electronic ISSN: 1573-7292
DOI
https://doi.org/10.1007/s10689-013-9675-2

Other articles of this Issue 1/2014

Familial Cancer 1/2014 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