Skip to main content
Top
Published in: Familial Cancer 2/2011

01-06-2011

Identification and surveillance of 19 Lynch syndrome families in southern Italy: report of six novel germline mutations and a common founder mutation

Authors: Patrizia Lastella, Margherita Patruno, Giovanna Forte, Alba Montanaro, Carmela Di Gregorio, Carlo Sabbà, Patrizia Suppressa, Adalgisa Piepoli, Anna Panza, Angelo Andriulli, Nicoletta Resta, Alessandro Stella

Published in: Familial Cancer | Issue 2/2011

Login to get access

Abstract

Lynch syndrome (LS), or hereditary non-polyposis colorectal cancer (HNPCC), is an autosomal dominant condition responsible for early onset cancer mostly in the colonrectum and endometrium as well as in other organ sites. Lynch syndrome is caused by germline mutations in mismatch repair genes, prevalently in hMSH2, hMLH1, and less frequently in hMSH6 and hPMS2. Twenty-nine non-related index cases with colorectal cancer (CRC) were collected from a region in southeast Italy (Apulia). Among this set of patients, fifteen fulfilled the Amsterdam criteria II. The presence of tumor microsatellite instability (MSI) was assessed in all index cases and 19 (15 AC+/4 AC−) were classified as MSI-H. Mutation analysis performed on all patients, identified 15 pathogenic mutations in hMLH1 and 4 in hMSH2. 4/15 mutations in hMLH1 and 2/4 hMSH2 mutations have not been previously reported. Three previously reported mutations were further investigated for the possibility of a common founder effect. Genetic counseling was offered to all probands and extended to 183 relatives after molecular testing and 85 (46%) mutation carriers were identified. Eighty mutation carriers underwent an accurate clinical and instrumental surveillance protocol. Our results confirm that the identification of LS patients based exclusively on family history may miss patients carrying germline mutations in the MMR genes. Moreover, our results demonstrated that molecular screening and subsequent instrumental surveillance are very effective in identifying CRCs at earlier stages and reducing the number of deaths from secondary cancers in HNPCC patients.
Literature
1.
go back to reference Hampel H, Frankel Wl, Martin E et al (2005) Screening for the lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med 352:1851–1860PubMedCrossRef Hampel H, Frankel Wl, Martin E et al (2005) Screening for the lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med 352:1851–1860PubMedCrossRef
2.
3.
go back to reference Peltomäki P, Vasen HF (1997) Mutations predisposing to hereditary nonpolyposis colorectal cancer: database and results of a collaborative study. The International Collaborative Group on hereditary nonpolyposis colorectal cancer. Gastroenterology 11:1146–1158CrossRef Peltomäki P, Vasen HF (1997) Mutations predisposing to hereditary nonpolyposis colorectal cancer: database and results of a collaborative study. The International Collaborative Group on hereditary nonpolyposis colorectal cancer. Gastroenterology 11:1146–1158CrossRef
4.
go back to reference Lagerstedt Robinson K, Liu T, Vandrovcova J et al (2007) Lynch syndrome (hereditary nonpolyposis colorectal cancer) diagnostics. J Natl Cancer Inst 99(4):291–299PubMedCrossRef Lagerstedt Robinson K, Liu T, Vandrovcova J et al (2007) Lynch syndrome (hereditary nonpolyposis colorectal cancer) diagnostics. J Natl Cancer Inst 99(4):291–299PubMedCrossRef
5.
go back to reference Mueller J, Gazzoli I, Bandipalliam P, Garber JE, Syngal S, Kolodner RD (2009) Comprehensive molecular analysis of mismatch repair gene defects in suspected Lynch syndrome (hereditary nonpolyposis colorectal cancer) cases. Cancer Res 69:7053–7061PubMedCrossRef Mueller J, Gazzoli I, Bandipalliam P, Garber JE, Syngal S, Kolodner RD (2009) Comprehensive molecular analysis of mismatch repair gene defects in suspected Lynch syndrome (hereditary nonpolyposis colorectal cancer) cases. Cancer Res 69:7053–7061PubMedCrossRef
6.
go back to reference n Cappel WH, Nagengast FM, Griffioen G (2002) Surveillance for hereditary nonpolyposis colorectal cancer: a long-term study on 114 families. Dis Colon Rectum 45(12):1588–1594CrossRef n Cappel WH, Nagengast FM, Griffioen G (2002) Surveillance for hereditary nonpolyposis colorectal cancer: a long-term study on 114 families. Dis Colon Rectum 45(12):1588–1594CrossRef
7.
go back to reference Lynch HT (1996) Is there a role for prophylactic subtotal colectomy among hereditary nonpolyposis colorectal cancer germline mutation carriers? Dis Colon Rectum 39(1):109–110PubMedCrossRef Lynch HT (1996) Is there a role for prophylactic subtotal colectomy among hereditary nonpolyposis colorectal cancer germline mutation carriers? Dis Colon Rectum 39(1):109–110PubMedCrossRef
8.
go back to reference Perea J, Justo I, Alvaro M (2009) Surgical management of hereditary colorectal cancer surgery based on molecular analysis and family history. Rev Esp Enferm Dig 101(8):536–540PubMedCrossRef Perea J, Justo I, Alvaro M (2009) Surgical management of hereditary colorectal cancer surgery based on molecular analysis and family history. Rev Esp Enferm Dig 101(8):536–540PubMedCrossRef
9.
go back to reference Lindor NM, Petersen GM, Hadley DW et al (2006) Recommendations for the care of individuals with an inherited predisposition to Lynch syndrome: a systematic review. JAMA 296(12):1507–1517PubMedCrossRef Lindor NM, Petersen GM, Hadley DW et al (2006) Recommendations for the care of individuals with an inherited predisposition to Lynch syndrome: a systematic review. JAMA 296(12):1507–1517PubMedCrossRef
10.
go back to reference Jarvinen HJ, Aarnio M, Mustonen H (2000) Controlled 15-years trial on screening for colorectal cancer in families with hereditary nonpolyposis colorectal cancer. Gastroenterology 118(5):829–834PubMedCrossRef Jarvinen HJ, Aarnio M, Mustonen H (2000) Controlled 15-years trial on screening for colorectal cancer in families with hereditary nonpolyposis colorectal cancer. Gastroenterology 118(5):829–834PubMedCrossRef
11.
go back to reference Vasen HF, Mecklin JP, Khan PM, Lynch HT (1991) The International Collaborative Group on hereditary nonpolyposis colorectal cancer (ICG-HNPCC). Dis Col Rectum 34(5):424–425CrossRef Vasen HF, Mecklin JP, Khan PM, Lynch HT (1991) The International Collaborative Group on hereditary nonpolyposis colorectal cancer (ICG-HNPCC). Dis Col Rectum 34(5):424–425CrossRef
12.
go back to reference Rodriguez-Bigas MA, Boland CR, Hamilton SR (1997) A National Cancer Institute workshop on hereditary non polyposis colorectal cancer syndrome: meeting highlights and Bethesda guidelines. J Natl Cancer Inst 89(22):1758–1762PubMed Rodriguez-Bigas MA, Boland CR, Hamilton SR (1997) A National Cancer Institute workshop on hereditary non polyposis colorectal cancer syndrome: meeting highlights and Bethesda guidelines. J Natl Cancer Inst 89(22):1758–1762PubMed
13.
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(4):261–268PubMedCrossRef 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(4):261–268PubMedCrossRef
14.
go back to reference Hampel H, Frankel WL, Martin E (2008) Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol 26(35):5783–5788PubMedCrossRef Hampel H, Frankel WL, Martin E (2008) Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol 26(35):5783–5788PubMedCrossRef
15.
go back to reference Barnetson RA, Tenesa A, Farrington S (2006) Identification and survival of carriers of mutations in DNA Mismatch-repair genes in colon cancer. New Engl J Med 354(26):2751–2761PubMedCrossRef Barnetson RA, Tenesa A, Farrington S (2006) Identification and survival of carriers of mutations in DNA Mismatch-repair genes in colon cancer. New Engl J Med 354(26):2751–2761PubMedCrossRef
16.
go back to reference Kastrinos F, Steyerberg EW, Mercado R et al. (2010) The PREMM 1,2,6 Model predicts risk of MLH1, MSH2, and MSH6 Germline Mutations based on cancer history. Gastroenterology Aug 18. [Epub ahead of print] Kastrinos F, Steyerberg EW, Mercado R et al. (2010) The PREMM 1,2,6 Model predicts risk of MLH1, MSH2, and MSH6 Germline Mutations based on cancer history. Gastroenterology Aug 18. [Epub ahead of print]
17.
go back to reference Boland CR, Thibodeau SN, Hamilton SR (1998) A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58(22):5248–5257PubMed Boland CR, Thibodeau SN, Hamilton SR (1998) A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58(22):5248–5257PubMed
18.
go back to reference Pedroni M, Sala E, Scarselli A et al (2001) Microsatellite instability and mismatch-repair protein expression in hereditary and sporadic colorectal carcinogenesis. Cancer Res 61:896–899PubMed Pedroni M, Sala E, Scarselli A et al (2001) Microsatellite instability and mismatch-repair protein expression in hereditary and sporadic colorectal carcinogenesis. Cancer Res 61:896–899PubMed
19.
go back to reference Holinski-Feder E, Muller-Koch Y, Friedl W (2001) DHPLC mutation analysis of the hereditary nonpolyposis colon cancer (HNPCC) genes hMLH1 and hMSH2. J Biochem Biophys Methods 47(1–2):21–32PubMedCrossRef Holinski-Feder E, Muller-Koch Y, Friedl W (2001) DHPLC mutation analysis of the hereditary nonpolyposis colon cancer (HNPCC) genes hMLH1 and hMSH2. J Biochem Biophys Methods 47(1–2):21–32PubMedCrossRef
20.
go back to reference Lastella P, Surdo NC, Resta N, Guanti G, Stella A (2006) In silico and in vivo splicing analysis of MLH1 and MSH2 missense mutations shows exon- and tissue-specific effects. BMC Genomics 7:243PubMedCrossRef Lastella P, Surdo NC, Resta N, Guanti G, Stella A (2006) In silico and in vivo splicing analysis of MLH1 and MSH2 missense mutations shows exon- and tissue-specific effects. BMC Genomics 7:243PubMedCrossRef
21.
go back to reference den Dunnen JT, Antonarakis SE (2000) Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat 15(1):7–12CrossRef den Dunnen JT, Antonarakis SE (2000) Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat 15(1):7–12CrossRef
22.
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(2):130–139PubMedCrossRef 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(2):130–139PubMedCrossRef
23.
go back to reference Tournier I, Vezain M, Martins A et al (2008) A large fraction of unclassified variants of the mismatch repair genes MLH1 and MSH2 is associated with splicing defects. Hum Mutat 29:1412–1424PubMedCrossRef Tournier I, Vezain M, Martins A et al (2008) A large fraction of unclassified variants of the mismatch repair genes MLH1 and MSH2 is associated with splicing defects. Hum Mutat 29:1412–1424PubMedCrossRef
24.
go back to reference Piepoli A, Santoro R, Cristofaro G et al (1996) Linkage analysis identifies gene carriers among members of families with hereditary nonpolyposis colorectal cancer. Gastroenterology 110(5):1404–1409PubMedCrossRef Piepoli A, Santoro R, Cristofaro G et al (1996) Linkage analysis identifies gene carriers among members of families with hereditary nonpolyposis colorectal cancer. Gastroenterology 110(5):1404–1409PubMedCrossRef
25.
go back to reference Viel A, Genuardi M, Lucci-Cordisco E et al (1998) Hereditary nonpolyposis colorectal cancer: an approach to the selection of candidates to genetic testing based on clinical and molecular characteristics. Community Genet 1(4):229–236PubMedCrossRef Viel A, Genuardi M, Lucci-Cordisco E et al (1998) Hereditary nonpolyposis colorectal cancer: an approach to the selection of candidates to genetic testing based on clinical and molecular characteristics. Community Genet 1(4):229–236PubMedCrossRef
26.
go back to reference Benatti P, Roncucci L, Percesepe A et al (1998) Small bowel carcinoma in hereditary nonpolyposis colorectal cancer. Am J Gastroenterol 93(11):2219–2222PubMedCrossRef Benatti P, Roncucci L, Percesepe A et al (1998) Small bowel carcinoma in hereditary nonpolyposis colorectal cancer. Am J Gastroenterol 93(11):2219–2222PubMedCrossRef
27.
go back to reference Wagner A, Tops C, Wijnen JT et al (2002) Genetic testing in hereditary non-polyposis colorectal cancer families with a MSH2, MLH1, or MSH6 mutation. J Med Genet 39:833–837PubMedCrossRef Wagner A, Tops C, Wijnen JT et al (2002) Genetic testing in hereditary non-polyposis colorectal cancer families with a MSH2, MLH1, or MSH6 mutation. J Med Genet 39:833–837PubMedCrossRef
28.
go back to reference Julié C, Trésallet C, Brouquet A et al (2008) Identification in daily practice of patients with Lynch syndrome (hereditary nonpolyposis colorectal cancer): revised Bethesda guidelines-based approach versus molecular screening. Am J Gastroenterol 103:2825–2835PubMedCrossRef Julié C, Trésallet C, Brouquet A et al (2008) Identification in daily practice of patients with Lynch syndrome (hereditary nonpolyposis colorectal cancer): revised Bethesda guidelines-based approach versus molecular screening. Am J Gastroenterol 103:2825–2835PubMedCrossRef
29.
go back to reference Wijnen J, Vasen H, Khan PM et al (1995) Seven new mutations in hMSH2, an HNPCC gene, identified by denaturing gradient-gel electrophoresis. Am J Hum Genet 56(5):1060–1066PubMed Wijnen J, Vasen H, Khan PM et al (1995) Seven new mutations in hMSH2, an HNPCC gene, identified by denaturing gradient-gel electrophoresis. Am J Hum Genet 56(5):1060–1066PubMed
30.
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(5):1088–1100PubMedCrossRef 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(5):1088–1100PubMedCrossRef
31.
go back to reference Desai DC, Lockman JC, Chadwick RB et al (2000) Recurrent germline mutation in MSH2 arises frequently de novo. J Med Genet 37(9):646–652PubMedCrossRef Desai DC, Lockman JC, Chadwick RB et al (2000) Recurrent germline mutation in MSH2 arises frequently de novo. J Med Genet 37(9):646–652PubMedCrossRef
32.
go back to reference Foulkes WD, Thiffault I, Gruber SB et al (2002) The founder mutation MSH2*1906G>C in an important cause of hereditary nonpolyposis colorectal cancer in the Ashkenazi Jewish population. Am J Hum Genet 71(6):1395–1412PubMedCrossRef Foulkes WD, Thiffault I, Gruber SB et al (2002) The founder mutation MSH2*1906G>C in an important cause of hereditary nonpolyposis colorectal cancer in the Ashkenazi Jewish population. Am J Hum Genet 71(6):1395–1412PubMedCrossRef
33.
go back to reference Caluseriu O, Di Gregorio C, Lucci-Cordisco E et al (2004) A founder MLH1 mutation in families from the districts of Modena and Reggio-Emilia in northern Italy with hereditary non-polyposis colorectal cancer associated with protein elongation and instability. J Med Genet 41(3):e34PubMedCrossRef Caluseriu O, Di Gregorio C, Lucci-Cordisco E et al (2004) A founder MLH1 mutation in families from the districts of Modena and Reggio-Emilia in northern Italy with hereditary non-polyposis colorectal cancer associated with protein elongation and instability. J Med Genet 41(3):e34PubMedCrossRef
34.
go back to reference Chan TL, Chan YW, Ho JW et al (2004) MSH2 c.1452–1455delAATG is a founder mutation and an important cause of hereditary nonpolyposis colorectal cancer in the southern Chinese population. Am J Hum Genet 74(5):1035–1042PubMedCrossRef Chan TL, Chan YW, Ho JW et al (2004) MSH2 c.1452–1455delAATG is a founder mutation and an important cause of hereditary nonpolyposis colorectal cancer in the southern Chinese population. Am J Hum Genet 74(5):1035–1042PubMedCrossRef
35.
go back to reference Pensotti V, Radice P, Presciutini S et al (1997) Mean age of tumor onset in hereditary nonpolyposis colorectal cancer (HNPCC) families correlates with the presence of mutations in DNA mismatch repair genes. Genes Chromosomes Cancer 19(3):135–142PubMedCrossRef Pensotti V, Radice P, Presciutini S et al (1997) Mean age of tumor onset in hereditary nonpolyposis colorectal cancer (HNPCC) families correlates with the presence of mutations in DNA mismatch repair genes. Genes Chromosomes Cancer 19(3):135–142PubMedCrossRef
36.
go back to reference Lynch HT, Lynch PM, Lanspa SJ, Snyder CL, Lynch JF, Boland CR (2009) Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis and medico legal ramifications. Clin Genet 76(1):1–18PubMedCrossRef Lynch HT, Lynch PM, Lanspa SJ, Snyder CL, Lynch JF, Boland CR (2009) Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis and medico legal ramifications. Clin Genet 76(1):1–18PubMedCrossRef
37.
go back to reference Kastrinos F, Stoffel EM, Balmaña J, Steyerberg EW, Mercado R, Syngal S (2008) Phenotype comparison of MLH1 and MSH2 mutation carriers in a cohort of 1914 individuals undergoing clinical genetic testing in the United States. Cancer Epidemiol Biomarkers Prev 17(8):2044–2051PubMedCrossRef Kastrinos F, Stoffel EM, Balmaña J, Steyerberg EW, Mercado R, Syngal S (2008) Phenotype comparison of MLH1 and MSH2 mutation carriers in a cohort of 1914 individuals undergoing clinical genetic testing in the United States. Cancer Epidemiol Biomarkers Prev 17(8):2044–2051PubMedCrossRef
38.
go back to reference n Cappel WH, Nagengast FM, Griffioen G et al (2002) Surveillance for hereditary nonpolyposis colorectal cancer: a long-term study on 114 families. Dis Colon Rectum 45(12):1558–1594 n Cappel WH, Nagengast FM, Griffioen G et al (2002) Surveillance for hereditary nonpolyposis colorectal cancer: a long-term study on 114 families. Dis Colon Rectum 45(12):1558–1594
39.
go back to reference Pylvanainen K, Kairaluoma M, Mecklin JP (2006) Compliance and satisfaction with long-term surveillance in Finnish HNPCC families. Fam Cancer 5(2):175–178PubMedCrossRef Pylvanainen K, Kairaluoma M, Mecklin JP (2006) Compliance and satisfaction with long-term surveillance in Finnish HNPCC families. Fam Cancer 5(2):175–178PubMedCrossRef
40.
go back to reference Järvinen HJ, Renkonen-Sinisalo L, Aktán-Collán K, Peltomäki P, Aaltonen LA, Mecklin JP (2009) Ten years after mutation testing for Lynch syndrome: cancer incidence and outcome in mutation-positive and mutation-negative family members. J Clin Oncol 27:4793–4797PubMedCrossRef Järvinen HJ, Renkonen-Sinisalo L, Aktán-Collán K, Peltomäki P, Aaltonen LA, Mecklin JP (2009) Ten years after mutation testing for Lynch syndrome: cancer incidence and outcome in mutation-positive and mutation-negative family members. J Clin Oncol 27:4793–4797PubMedCrossRef
41.
go back to reference De Jong AE, Morreau H, Van Puijenbroek M et al (2004) The role of mismatch repair gene defects in the development of adenomas in patients with HNPCC. Gastroenterology 126:42–48PubMedCrossRef De Jong AE, Morreau H, Van Puijenbroek M et al (2004) The role of mismatch repair gene defects in the development of adenomas in patients with HNPCC. Gastroenterology 126:42–48PubMedCrossRef
42.
go back to reference German HNPCC Consortium, Müller A, Beckmann C, Westphal G et al (2006) Prevalence of the mismatch-repair-deficient phenotype in colonic adenomas arising in HNPCC patients: results of a 5-year follow-up study. Int J Colorectal Dis 21:632–641PubMedCrossRef German HNPCC Consortium, Müller A, Beckmann C, Westphal G et al (2006) Prevalence of the mismatch-repair-deficient phenotype in colonic adenomas arising in HNPCC patients: results of a 5-year follow-up study. Int J Colorectal Dis 21:632–641PubMedCrossRef
43.
go back to reference Hurlstone DP, Karajeh M, Cross SS et al (2005) The role of high-magnification-chromoscopic colonoscopy in hereditary nonpolyposis colorectal cancer screening: a prospective “back-to-back” endoscopy study. Am J Gastroenterology 100:2167–2173CrossRef Hurlstone DP, Karajeh M, Cross SS et al (2005) The role of high-magnification-chromoscopic colonoscopy in hereditary nonpolyposis colorectal cancer screening: a prospective “back-to-back” endoscopy study. Am J Gastroenterology 100:2167–2173CrossRef
44.
go back to reference Koornstra JJ, Mourits MJ, Sijmons RH et al (2009) Management of extracolonic tumours in patients with Lynch syndrome. Lancet 10(4):400–408CrossRef Koornstra JJ, Mourits MJ, Sijmons RH et al (2009) Management of extracolonic tumours in patients with Lynch syndrome. Lancet 10(4):400–408CrossRef
45.
go back to reference Hüneburg R, Lammert F, Rabe C et al (2009) Chromocolonoscopy detects more adenomas than white light colonoscopy or narrow band imaging colonoscopy in hereditary nonpolyposis colorectal cancer screening. Endoscopy 41(4):316–322PubMedCrossRef Hüneburg R, Lammert F, Rabe C et al (2009) Chromocolonoscopy detects more adenomas than white light colonoscopy or narrow band imaging colonoscopy in hereditary nonpolyposis colorectal cancer screening. Endoscopy 41(4):316–322PubMedCrossRef
46.
go back to reference Rex DK, Cutler CS, Lemmel GT et al (1997) Colonoscopic miss rates of adenomas determined by back-to-back colonoscopies. Gastroenterology 112:24–28PubMedCrossRef Rex DK, Cutler CS, Lemmel GT et al (1997) Colonoscopic miss rates of adenomas determined by back-to-back colonoscopies. Gastroenterology 112:24–28PubMedCrossRef
47.
go back to reference Trojan J, Zeuzem S, Randolph A et al (2002) Functional analysis of hMLH1 variants and HNPCC-related mutations using a human expression system. Gastroenterology 122(1):211–219PubMedCrossRef Trojan J, Zeuzem S, Randolph A et al (2002) Functional analysis of hMLH1 variants and HNPCC-related mutations using a human expression system. Gastroenterology 122(1):211–219PubMedCrossRef
48.
go back to reference Takahashi M, Shimodaira H, Andreutti-Zaugg C, Iggo R, Kolodner RD, Ishioka C (2007) Functional analysis of human MLH1 variants using yeast and in vitro mismatch repair assays. Cancer Res 67(10):4595–4604PubMedCrossRef Takahashi M, Shimodaira H, Andreutti-Zaugg C, Iggo R, Kolodner RD, Ishioka C (2007) Functional analysis of human MLH1 variants using yeast and in vitro mismatch repair assays. Cancer Res 67(10):4595–4604PubMedCrossRef
49.
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(2):537–549PubMed Raevaara TE, Korhonen MK, Lohi H et al (2005) Functional significance and clinical phenotype of nontruncating mismatch repair variants of MLH1. Gastroenterology 129(2):537–549PubMed
50.
go back to reference Pagenstecher C, Wehner M, Friedl W et al (2006) Aberrant splicing in MLH1 and MSH2 due to exonic and intronic variants. Hum Genet 119(1–2):9–22PubMedCrossRef Pagenstecher C, Wehner M, Friedl W et al (2006) Aberrant splicing in MLH1 and MSH2 due to exonic and intronic variants. Hum Genet 119(1–2):9–22PubMedCrossRef
51.
go back to reference Auclair J, Busine MP, Navarro C et al (2006) Systematic mRNA analysis for the effect of MLH1 and MSH2 missense and silent mutations on aberrant splicing. Hum Mutat 27(2):145–154PubMedCrossRef Auclair J, Busine MP, Navarro C et al (2006) Systematic mRNA analysis for the effect of MLH1 and MSH2 missense and silent mutations on aberrant splicing. Hum Mutat 27(2):145–154PubMedCrossRef
52.
go back to reference Kondo E, Suzuki H, Horii A, Fukushige S (2003) A yeast two-hybrid assay provides a simple way to evaluate the vast majority of hMLH1 germline mutations. Cancer Res 63(12):3302–3308PubMed Kondo E, Suzuki H, Horii A, Fukushige S (2003) A yeast two-hybrid assay provides a simple way to evaluate the vast majority of hMLH1 germline mutations. Cancer Res 63(12):3302–3308PubMed
Metadata
Title
Identification and surveillance of 19 Lynch syndrome families in southern Italy: report of six novel germline mutations and a common founder mutation
Authors
Patrizia Lastella
Margherita Patruno
Giovanna Forte
Alba Montanaro
Carmela Di Gregorio
Carlo Sabbà
Patrizia Suppressa
Adalgisa Piepoli
Anna Panza
Angelo Andriulli
Nicoletta Resta
Alessandro Stella
Publication date
01-06-2011
Publisher
Springer Netherlands
Published in
Familial Cancer / Issue 2/2011
Print ISSN: 1389-9600
Electronic ISSN: 1573-7292
DOI
https://doi.org/10.1007/s10689-011-9419-0

Other articles of this Issue 2/2011

Familial Cancer 2/2011 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