Skip to main content
Erschienen in: Current Treatment Options in Oncology 3/2014

01.09.2014 | Lower Gastrointestinal Cancers (AB Benson, Section Editor)

Genetics, Biomarkers, Hereditary Cancer Syndrome Diagnosis, Heterogeneity and Treatment: A Review

verfasst von: Henry T. Lynch, MD, Kristen Drescher, PhD, Joseph Knezetic, PhD, Stephen Lanspa, MD

Erschienen in: Current Treatment Options in Oncology | Ausgabe 3/2014

Einloggen, um Zugang zu erhalten

Opinion statement

Molecular genetic pathways that drive the phenotypic and genotypic heterogeneity of hereditary colorectal cancer also can affect response to chemotherapy and chemoprevention. These mutations also can alter patients’ response to therapy. Environmental differences can affect this highly complex conundrum. We will use Lynch syndrome as a model to explore this issue. However, to begin with, after more than a century of documentation, we must ask what is meant by the eponym “Lynch syndrome”. Germline mutations may act as drivers of chemoprevention and chemotherapy and therein may act positively or conversely they may have a negative effect in terms of inhibiting the inactivation of cancer-causing germline mutations. A relatively new field of hereditary cancer therapeutics has significantly impacted cancer care, from the standpoint of the sensitivity or resistance to a particular form of chemotherapy and/or chemoprevention. The question for the diagnostician and therapist must always concern what is the best possible management approach for the patient, particularly when he or she harbors a cancer-causing germline mutation, which, in this case, causes Lynch syndrome. Continued molecular genetic research might yield a more tailored effective treatment for Lynch syndrome. The ultimate goal of such hereditary oncologic research is to better understand the mutation’s therapeutic task, namely, its potential to benefit the patient in terms of its treatment goal, thereby fulfilling the essence of personalized medicine. However, this goal may be exceedingly complicated. For example, in the natural clinical and molecular genetic history of hereditary forms of cancer, there will be a predominance of early-onset cancers of multiple anatomic sites. In our Lynch syndrome model, these will be most commonly colorectal, endometrial, and ovarian cancer. Attention must initially be focused upon cancer’s early age of onset coupled with the tendency to multiple primary cancers so that, in the case of CRC, colonoscopic screening must be initiated by age 20–25 years and repeated every other year until age 40 years and then annually thereafter. However, screening will be of limited efficacy in the gynecologic cancers (endometrial and ovarian) so that once the family is completed, particularly by age 35–40 years, careful attention must be given to the option of prophylactic hysterectomy and bilateral salpingo-oophorectomy. Given issues of tumor heterogeneity, selected Lynch syndrome families may show an excess of urologic cancers or cancers of the small bowel, and highly targeted screening should be given serious consideration for these as well as cancers of other anatomic sites in such high-risk, cancer-prone patients.
Literatur
1.
Zurück zum Zitat Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM: GLOBOCAN 2008, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 10 [Internet]. Lyon, France: International Agency for Research on Cancer. Available at: http://www-dep.iarc.fr. Accessed August 30, 2012. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM: GLOBOCAN 2008, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 10 [Internet]. Lyon, France: International Agency for Research on Cancer. Available at: http://​www-dep.​iarc.​fr. Accessed August 30, 2012.
2.
3.
Zurück zum Zitat Lynch HT, Lynch PM, Lanspa SJ, et al. Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications. Clin Genet. 2009;76:1–18.PubMedCentralPubMedCrossRef Lynch HT, Lynch PM, Lanspa SJ, et al. Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications. Clin Genet. 2009;76:1–18.PubMedCentralPubMedCrossRef
4.
Zurück zum Zitat Hampel H, Frankel WL, Martin E, et al. Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol. 2008;26:5783–8.PubMedCentralPubMedCrossRef Hampel H, Frankel WL, Martin E, et al. Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol. 2008;26:5783–8.PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Working Group. Recommendations from the EGAPP Working Group: genetic testing strategies in newly diagnosed individuals with colorectal cancer aimed at reducing morbidity and mortality from Lynch syndrome in relatives. Genet Med. 2009;11:35–41.CrossRef Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Working Group. Recommendations from the EGAPP Working Group: genetic testing strategies in newly diagnosed individuals with colorectal cancer aimed at reducing morbidity and mortality from Lynch syndrome in relatives. Genet Med. 2009;11:35–41.CrossRef
6.
Zurück zum Zitat Davidson NO. Genetic testing in colorectal cancer: who, when, how and why. Keio J Med. 2007;56:14–20.PubMedCrossRef Davidson NO. Genetic testing in colorectal cancer: who, when, how and why. Keio J Med. 2007;56:14–20.PubMedCrossRef
7.
Zurück zum Zitat Lynch HT, Boland CR, Rodriguez-Bigas MA, et al. Who should be sent for genetic testing in hereditary colorectal cancer syndromes? J Clin Oncol. 2007;25:3534–42.PubMedCrossRef Lynch HT, Boland CR, Rodriguez-Bigas MA, et al. Who should be sent for genetic testing in hereditary colorectal cancer syndromes? J Clin Oncol. 2007;25:3534–42.PubMedCrossRef
9.•
Zurück zum Zitat Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet. 2013;45:1113–20. This paper demonstrates some of the important work being done by the Cancer Genome Atlas Research Network aimed at increasing the efficacy of cancer therapy.CrossRef Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet. 2013;45:1113–20. This paper demonstrates some of the important work being done by the Cancer Genome Atlas Research Network aimed at increasing the efficacy of cancer therapy.CrossRef
10.
Zurück zum Zitat Kamalakaran S, Varadan V, Janevski A, et al. Translating next generation sequencing to practice: opportunities and necessary steps. Mol Oncol. 2013;7:743–55.PubMedCrossRef Kamalakaran S, Varadan V, Janevski A, et al. Translating next generation sequencing to practice: opportunities and necessary steps. Mol Oncol. 2013;7:743–55.PubMedCrossRef
12.
Zurück zum Zitat Mensenkamp AR, Vogelaar IP, van Zelst-Stams WAG, et al. Somatic mutations in MLH1 and MSH2 are a frequent cause of mismatch-repair deficiency in Lynch syndrome-like tumors. Gastroenterology. 2014;146:643–6.PubMedCrossRef Mensenkamp AR, Vogelaar IP, van Zelst-Stams WAG, et al. Somatic mutations in MLH1 and MSH2 are a frequent cause of mismatch-repair deficiency in Lynch syndrome-like tumors. Gastroenterology. 2014;146:643–6.PubMedCrossRef
13.•
Zurück zum Zitat Carethers JM. Differentiating Lynch-like from Lynch syndrome. Gastroenterology. 2014;146:602–4. This commentary pulls together the current thinking on the newly identified “Lynch-like” colorectal cancer and how it differs from Lynch syndrome.PubMedCrossRef Carethers JM. Differentiating Lynch-like from Lynch syndrome. Gastroenterology. 2014;146:602–4. This commentary pulls together the current thinking on the newly identified “Lynch-like” colorectal cancer and how it differs from Lynch syndrome.PubMedCrossRef
14.
Zurück zum Zitat Hitchins MP, Ward RL. Constitutional (germline) MLH1 epimutation as an aetiological mechanism for hereditary non-polyposis colorectal cancer. J Med Genet. 2009;46:793–802.PubMedCrossRef Hitchins MP, Ward RL. Constitutional (germline) MLH1 epimutation as an aetiological mechanism for hereditary non-polyposis colorectal cancer. J Med Genet. 2009;46:793–802.PubMedCrossRef
16.
Zurück zum Zitat Watson P, Vasen HFA, Mecklin J-P, et al. The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. Int J Cancer. 2008;123:444–9.PubMedCentralPubMedCrossRef Watson P, Vasen HFA, Mecklin J-P, et al. The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. Int J Cancer. 2008;123:444–9.PubMedCentralPubMedCrossRef
17.
Zurück zum Zitat Ligtenberg MJL, Kuiper RP, Chan TL, et al. Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3' exons of TACSTD1. Nat Genet. 2009;41:112–7.PubMedCrossRef Ligtenberg MJL, Kuiper RP, Chan TL, et al. Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3' exons of TACSTD1. Nat Genet. 2009;41:112–7.PubMedCrossRef
18.
Zurück zum Zitat Lynch H, Riegert-Johnson D, Snyder C, et al. Lynch syndrome associated extracolonic tumors are rare in two extended families with the same EPCAM deletion. Am J Gastroenterol. 2011;106:1829–36.PubMedCentralPubMedCrossRef Lynch H, Riegert-Johnson D, Snyder C, et al. Lynch syndrome associated extracolonic tumors are rare in two extended families with the same EPCAM deletion. Am J Gastroenterol. 2011;106:1829–36.PubMedCentralPubMedCrossRef
19.
Zurück zum Zitat Kempers MJE, Kuiper RP, Ockeloen CW, et al. Risk of colorectal and endometrial cancers in EPCAM deletion-positive Lynch syndrome: a cohort study. Lancet Oncol. 2011;12:49–55.PubMedCentralPubMedCrossRef Kempers MJE, Kuiper RP, Ockeloen CW, et al. Risk of colorectal and endometrial cancers in EPCAM deletion-positive Lynch syndrome: a cohort study. Lancet Oncol. 2011;12:49–55.PubMedCentralPubMedCrossRef
20.
Zurück zum Zitat Lindor NM, Rabe K, Petersen GM, et al. Lower cancer incidence in Amsterdam-I criteria families without mismatch repair deficiency: familial colorectal cancer type X. JAMA. 2005;293:1979–85.PubMedCentralPubMedCrossRef Lindor NM, Rabe K, Petersen GM, et al. Lower cancer incidence in Amsterdam-I criteria families without mismatch repair deficiency: familial colorectal cancer type X. JAMA. 2005;293:1979–85.PubMedCentralPubMedCrossRef
21.
Zurück zum Zitat Ferreira AM, Westers H, Sousa S, et al. Mononucleotide precedes dinucleotide repeat instability during colorectal tumour development in Lynch syndrome patients. J Pathol. 2009;219:96–102.PubMedCrossRef Ferreira AM, Westers H, Sousa S, et al. Mononucleotide precedes dinucleotide repeat instability during colorectal tumour development in Lynch syndrome patients. J Pathol. 2009;219:96–102.PubMedCrossRef
22.
Zurück zum Zitat Bessa X, Balleste B, Andreu M, et al. A prospective, multicenter, population-based study of BRAF mutational analysis for Lynch syndrome screening. Clin Gastroenterol Hepatol. 2008;6:206–14.PubMedCrossRef Bessa X, Balleste B, Andreu M, et al. A prospective, multicenter, population-based study of BRAF mutational analysis for Lynch syndrome screening. Clin Gastroenterol Hepatol. 2008;6:206–14.PubMedCrossRef
24.
Zurück zum Zitat Hamilton SR, Aaltonen LA, editors. Tumours of the Digestive System. Lyon: IARC Press; 2000. Hamilton SR, Aaltonen LA, editors. Tumours of the Digestive System. Lyon: IARC Press; 2000.
25.
Zurück zum Zitat Miyakura Y, Sugano K, Konishi F, et al. Extensive methylation of hMLH1 promoter region predominates in proximal colon cancer with microsatellite instability. Gastroenterology. 2001;121:1300–9.PubMedCrossRef Miyakura Y, Sugano K, Konishi F, et al. Extensive methylation of hMLH1 promoter region predominates in proximal colon cancer with microsatellite instability. Gastroenterology. 2001;121:1300–9.PubMedCrossRef
26.
Zurück zum Zitat Nakagawa H, Nuovo GJ, Zervos EE, et al. Age-related hypermethylation of the 5' region of MLH1 in normal colonic mucousa is associated with microsatellite-unstable colorectal cancer development. Cancer Res. 2001;61:6991–5.PubMed Nakagawa H, Nuovo GJ, Zervos EE, et al. Age-related hypermethylation of the 5' region of MLH1 in normal colonic mucousa is associated with microsatellite-unstable colorectal cancer development. Cancer Res. 2001;61:6991–5.PubMed
27.
Zurück zum Zitat Veigl ML, Kasturi L, Olechnowicz J, et al. Biallelic inactivation of hMLH1 by epigenetic gene silencing, a novel mechanism causing human MSI cancers. Proc Natl Acad Sci. 1998;95:8698–702.PubMedCentralPubMedCrossRef Veigl ML, Kasturi L, Olechnowicz J, et al. Biallelic inactivation of hMLH1 by epigenetic gene silencing, a novel mechanism causing human MSI cancers. Proc Natl Acad Sci. 1998;95:8698–702.PubMedCentralPubMedCrossRef
28.
Zurück zum Zitat Herman JG, Umar A, Polyak K, et al. Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc Natl Acad Sci. 1998;95:6870–5.PubMedCentralPubMedCrossRef Herman JG, Umar A, Polyak K, et al. Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc Natl Acad Sci. 1998;95:6870–5.PubMedCentralPubMedCrossRef
30.
Zurück zum Zitat Hampel H, Frankel WL, Martin E, et al. Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med. 2005;352:1851–60.PubMedCrossRef Hampel H, Frankel WL, Martin E, et al. Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med. 2005;352:1851–60.PubMedCrossRef
31.
Zurück zum Zitat Carethers JM. DNA testing and molecular screening for colon cancer. Clin Gastroenterol Hepatol. 2014;12:377–81.PubMedCrossRef Carethers JM. DNA testing and molecular screening for colon cancer. Clin Gastroenterol Hepatol. 2014;12:377–81.PubMedCrossRef
32.•
Zurück zum Zitat Rodriguez-Soler M, Perez-Carbonell L, Guarinos C, et al. Risk of cancer in cases of suspected lynch syndrome without germline mutation. Gastroenterology. 2013;144:926–32. This is an up-to-date study of cancer risk in suspected Lynch syndrome without an identified germline mutation.PubMedCrossRef Rodriguez-Soler M, Perez-Carbonell L, Guarinos C, et al. Risk of cancer in cases of suspected lynch syndrome without germline mutation. Gastroenterology. 2013;144:926–32. This is an up-to-date study of cancer risk in suspected Lynch syndrome without an identified germline mutation.PubMedCrossRef
33.
Zurück zum Zitat Sourrouille I, Coulet F, Lefevre JH, et al. Somatic mosaicism and double somatic hits can lead to MSI colorectal tumors. Fam Cancer. 2013;12:27–33.PubMedCrossRef Sourrouille I, Coulet F, Lefevre JH, et al. Somatic mosaicism and double somatic hits can lead to MSI colorectal tumors. Fam Cancer. 2013;12:27–33.PubMedCrossRef
34.
Zurück zum Zitat Boland CR. The mystery of mismatch repair deficiency: Lynch or Lynch-like? Gastroenterology. 2013;144:869.CrossRef Boland CR. The mystery of mismatch repair deficiency: Lynch or Lynch-like? Gastroenterology. 2013;144:869.CrossRef
35.
Zurück zum Zitat Poynter JN, Siegmund KD, Weisenberger DJ, et al. Molecular characterization of MSI-H colorectal cancer by MLH1 promoter methylation, immunohistochemistry, and mismatch repair germline mutation screening. Cancer Epidemiol Biomarkers Prev. 2008;17:3208–15.PubMedCentralPubMedCrossRef Poynter JN, Siegmund KD, Weisenberger DJ, et al. Molecular characterization of MSI-H colorectal cancer by MLH1 promoter methylation, immunohistochemistry, and mismatch repair germline mutation screening. Cancer Epidemiol Biomarkers Prev. 2008;17:3208–15.PubMedCentralPubMedCrossRef
36.
Zurück zum Zitat Bettstetter M, Dechant S, Ruemmele P, et al. Distinction of hereditary nonpolyposis colorectal cancer and sporadic microsatellite-unstable colorectal cancer through quantification of MLH1 methylation by real-time PCR. Clin Cancer Res. 2007;13:3221–8.PubMedCrossRef Bettstetter M, Dechant S, Ruemmele P, et al. Distinction of hereditary nonpolyposis colorectal cancer and sporadic microsatellite-unstable colorectal cancer through quantification of MLH1 methylation by real-time PCR. Clin Cancer Res. 2007;13:3221–8.PubMedCrossRef
37.
Zurück zum Zitat Salahshor S, Koeble K, Rubio C, et al. Microsatellite instability and hMLH1 and hMSH2 expression analysis in familial and sporadic colorectal cancer. Lab Investig. 2001;81:535–41.PubMedCrossRef Salahshor S, Koeble K, Rubio C, et al. Microsatellite instability and hMLH1 and hMSH2 expression analysis in familial and sporadic colorectal cancer. Lab Investig. 2001;81:535–41.PubMedCrossRef
38.
Zurück zum Zitat Lindor NM, Burgart LJ, Leontovich O, et al. Immunohistochemistry versus microsatellite instability testing in phenotyping colorectal tumors. J Clin Oncol. 2002;20:1043–8.PubMedCrossRef Lindor NM, Burgart LJ, Leontovich O, et al. Immunohistochemistry versus microsatellite instability testing in phenotyping colorectal tumors. J Clin Oncol. 2002;20:1043–8.PubMedCrossRef
39.
Zurück zum Zitat Domingo E, Laiho P, Ollikainen M, et al. BRAF screening as a low-cost effective strategy for simplifying HNPCC genetic testing. J Med Genet. 2004;41:664–8.PubMedCentralPubMedCrossRef Domingo E, Laiho P, Ollikainen M, et al. BRAF screening as a low-cost effective strategy for simplifying HNPCC genetic testing. J Med Genet. 2004;41:664–8.PubMedCentralPubMedCrossRef
40.
Zurück zum Zitat Lynch HT, Silva E, Wirtzfeld D, et al. Hereditary diffuse gastric cancer: prophylactic surgical oncology implications. Surg Clin N Am. 2008;88:759–78.PubMedCentralPubMedCrossRef Lynch HT, Silva E, Wirtzfeld D, et al. Hereditary diffuse gastric cancer: prophylactic surgical oncology implications. Surg Clin N Am. 2008;88:759–78.PubMedCentralPubMedCrossRef
41.
Zurück zum Zitat Ionov YM, Peinado MA, Malkhosyan S, et al. Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature. 1993;363:558–61.PubMedCrossRef Ionov YM, Peinado MA, Malkhosyan S, et al. Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature. 1993;363:558–61.PubMedCrossRef
42.
Zurück zum Zitat Rodriguez-Bigas MA, Boland CR, Hamilton SR, et al. A National Cancer Institute workshop on hereditary nonpolyposis colorectal cancer syndrome: meeting highlights and Bethesda Guidelines. J Natl Cancer Inst. 1997;89:1758–62.PubMedCrossRef Rodriguez-Bigas MA, Boland CR, Hamilton SR, et al. A National Cancer Institute workshop on hereditary nonpolyposis colorectal cancer syndrome: meeting highlights and Bethesda Guidelines. J Natl Cancer Inst. 1997;89:1758–62.PubMedCrossRef
43.
Zurück zum Zitat Umar A, Boland CR, Terdiman JP, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004;96:261–8.PubMedCentralPubMedCrossRef Umar A, Boland CR, Terdiman JP, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004;96:261–8.PubMedCentralPubMedCrossRef
44.
Zurück zum Zitat Pérez-Carbonell L, Ruiz-Ponte C, Guarinos C, et al. Comparison between universal molecular screening for Lynch syndrome and revised Bethesda guidelines in a large population-based cohort of patients wtih colorectal cancer. Gut. 2012;61:865–72.PubMedCrossRef Pérez-Carbonell L, Ruiz-Ponte C, Guarinos C, et al. Comparison between universal molecular screening for Lynch syndrome and revised Bethesda guidelines in a large population-based cohort of patients wtih colorectal cancer. Gut. 2012;61:865–72.PubMedCrossRef
45.
Zurück zum Zitat Mvundura M, Grosse SD, Hampel H, et al. The cost-effectiveness of genetic testing strategies for Lynch syndrome among newly diagnosed patients with colorectal cancer. Genet Med. 2010;12:93–104.PubMedCrossRef Mvundura M, Grosse SD, Hampel H, et al. The cost-effectiveness of genetic testing strategies for Lynch syndrome among newly diagnosed patients with colorectal cancer. Genet Med. 2010;12:93–104.PubMedCrossRef
46.
Zurück zum Zitat Ladabaum U, Wang G, Terdiman J, et al. Strategies to identify the Lynch syndrome among patients with colorectal cancer: a cost-effectiveness analysis. Ann Intern Med. 2011;155:69–79.PubMedCentralPubMedCrossRef Ladabaum U, Wang G, Terdiman J, et al. Strategies to identify the Lynch syndrome among patients with colorectal cancer: a cost-effectiveness analysis. Ann Intern Med. 2011;155:69–79.PubMedCentralPubMedCrossRef
47.
Zurück zum Zitat Heald B, Plesec T, Liu X, et al. Implementation of universal microsatellite instability and immunohistochemistry screening for diagnosing Lynch syndrome in a large acadmic medical center. J Clin Oncol. 2013;31:1336–40.PubMedCrossRef Heald B, Plesec T, Liu X, et al. Implementation of universal microsatellite instability and immunohistochemistry screening for diagnosing Lynch syndrome in a large acadmic medical center. J Clin Oncol. 2013;31:1336–40.PubMedCrossRef
49.
Zurück zum Zitat Stupart DA, Goldberg PA, Algar U, et al. Surveillance colonoscopy improves survival in a cohort of subjects with a single mismatch repair gene mutation. Color Dis. 2009;11:126–30.CrossRef Stupart DA, Goldberg PA, Algar U, et al. Surveillance colonoscopy improves survival in a cohort of subjects with a single mismatch repair gene mutation. Color Dis. 2009;11:126–30.CrossRef
50.
Zurück zum Zitat Järvinen HJ, Aarnio M, Mustonen H, et al. Controlled 15-year trial on screening for colorectal cancer in families with hereditary nonpolyposis colorectal cancer. Gastroenterology. 2000;118:829–34.PubMedCrossRef Järvinen HJ, Aarnio M, Mustonen H, et al. Controlled 15-year trial on screening for colorectal cancer in families with hereditary nonpolyposis colorectal cancer. Gastroenterology. 2000;118:829–34.PubMedCrossRef
51.
Zurück zum Zitat de Jong AE, Hendriks YMC, Kleibeuker JH, et al. Decrease in mortality in Lynch syndrome families because of surveillance. Gastroenterology. 2006;130:665–71.PubMedCrossRef de Jong AE, Hendriks YMC, Kleibeuker JH, et al. Decrease in mortality in Lynch syndrome families because of surveillance. Gastroenterology. 2006;130:665–71.PubMedCrossRef
52.
Zurück zum Zitat Järvinen HJ, Renkonen-Sinisalo L, Aktán-Collán K, et al. Ten years after mutation testing for Lynch syndrome: cancer incidence and outcome in mutation-positive and mutation-negative family members. J Clin Oncol. 2009;27:4793–7.PubMedCrossRef Järvinen HJ, Renkonen-Sinisalo L, Aktán-Collán K, et al. Ten years after mutation testing for Lynch syndrome: cancer incidence and outcome in mutation-positive and mutation-negative family members. J Clin Oncol. 2009;27:4793–7.PubMedCrossRef
53.
Zurück zum Zitat Smyrk TC, Watson P, Kaul K, et al. Tumor-infiltrating lymphocytes are a marker for microsatellite instability in colorectal cancer. Cancer. 2001;91:2417–22.PubMedCrossRef Smyrk TC, Watson P, Kaul K, et al. Tumor-infiltrating lymphocytes are a marker for microsatellite instability in colorectal cancer. Cancer. 2001;91:2417–22.PubMedCrossRef
54.
Zurück zum Zitat Schwitalle Y, Kloor M, Eiermann S, et al. Immune response against frameshift-induced neopeptides in HNPCC patients and healthy HNPCC mutation carriers. Gastroenterology. 2008;134:988–97.PubMedCrossRef Schwitalle Y, Kloor M, Eiermann S, et al. Immune response against frameshift-induced neopeptides in HNPCC patients and healthy HNPCC mutation carriers. Gastroenterology. 2008;134:988–97.PubMedCrossRef
55.••
Zurück zum Zitat Huh JW, Lee HG, Kim HR. Prognostic significance of tumor-infiltrating lymphocytes for patients with colorectal cancer. Arch Surg. 2012;147:366–71. This is a recent investigation of prognostic significance of tumor-infiltrating lymphocytes in colorectal cancer patients.PubMedCrossRef Huh JW, Lee HG, Kim HR. Prognostic significance of tumor-infiltrating lymphocytes for patients with colorectal cancer. Arch Surg. 2012;147:366–71. This is a recent investigation of prognostic significance of tumor-infiltrating lymphocytes in colorectal cancer patients.PubMedCrossRef
56.
Zurück zum Zitat Loi S, Sirtaine N, Piette F, et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02–98. J Clin Oncol. 2013;31:860–7.PubMedCrossRef Loi S, Sirtaine N, Piette F, et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02–98. J Clin Oncol. 2013;31:860–7.PubMedCrossRef
57.••
Zurück zum Zitat Devaud N, Gallinger S. Chemotherapy of MMR-deficient colorectal cancer. Fam Cancer. 2013;12:301–6. This is an up-to-date overview of the effect of chemotherapy in mismatch repair-deficient colorectal cancer.PubMedCrossRef Devaud N, Gallinger S. Chemotherapy of MMR-deficient colorectal cancer. Fam Cancer. 2013;12:301–6. This is an up-to-date overview of the effect of chemotherapy in mismatch repair-deficient colorectal cancer.PubMedCrossRef
58.
Zurück zum Zitat Hong SP, Min BS, Kim TI, et al. The differential impact of microsatellite instability as a marker of prognosis and tumour response between colon cancer and rectal cancer. Eur J Cancer. 2012;48:1235–43.PubMedCrossRef Hong SP, Min BS, Kim TI, et al. The differential impact of microsatellite instability as a marker of prognosis and tumour response between colon cancer and rectal cancer. Eur J Cancer. 2012;48:1235–43.PubMedCrossRef
59.
Zurück zum Zitat Guastadisegni C, Colafranceschi M, Ottini L, et al. Microsatellite instability as a marker of prognosis and response to therapy: A meta-analysis of colorectal cancer survival data. Eur J Cancer. 2010;46:2788–98.PubMedCrossRef Guastadisegni C, Colafranceschi M, Ottini L, et al. Microsatellite instability as a marker of prognosis and response to therapy: A meta-analysis of colorectal cancer survival data. Eur J Cancer. 2010;46:2788–98.PubMedCrossRef
60.••
Zurück zum Zitat Sinicrope FA, Foster NR, Thibodeau SN, et al. DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst. 2011;103:863–75. This is an important study on 5-fluorouracil-based adjuvant therapy in relation to mismatch repair status. Although there has been some criticism of the study's selection of subjects, its results are still noteworthy as the only study to date that has investigated the difference in chemotherapy results between Lynch syndrome colorectal cancer and sporadic microsatellite instability-high colorectal cancer.PubMedCentralPubMedCrossRef Sinicrope FA, Foster NR, Thibodeau SN, et al. DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst. 2011;103:863–75. This is an important study on 5-fluorouracil-based adjuvant therapy in relation to mismatch repair status. Although there has been some criticism of the study's selection of subjects, its results are still noteworthy as the only study to date that has investigated the difference in chemotherapy results between Lynch syndrome colorectal cancer and sporadic microsatellite instability-high colorectal cancer.PubMedCentralPubMedCrossRef
61.
Zurück zum Zitat de Vos tot Nederveen Cappel WH, Meulenbeld HJ, Kleibeuker JH, et al. Survival after adjuvant 5-FU treatment for stage III colon cancer in hereditary nonpolyposis colorectal cancer. Int J Cancer. 2004;109:468–71.PubMedCrossRef de Vos tot Nederveen Cappel WH, Meulenbeld HJ, Kleibeuker JH, et al. Survival after adjuvant 5-FU treatment for stage III colon cancer in hereditary nonpolyposis colorectal cancer. Int J Cancer. 2004;109:468–71.PubMedCrossRef
62.
Zurück zum Zitat Kim ST, Lee J, Park SH, et al. Clinical impact of microsatellite instability in colon cancer following adjuvant FOLFOX therapy. Cancer Chemother Pharmacol. 2010;66:659–67.PubMedCrossRef Kim ST, Lee J, Park SH, et al. Clinical impact of microsatellite instability in colon cancer following adjuvant FOLFOX therapy. Cancer Chemother Pharmacol. 2010;66:659–67.PubMedCrossRef
63.
Zurück zum Zitat Zaanan A, Fléjou J-F, Emile J-F, et al. Defective mismatch repair status as a prognostic biomarker of disease-free survival in stage III colon cancer patients treated with adjuvant FOLFOX chemotherapy. Clin Cancer Res. 2011;17:7470–8.PubMedCrossRef Zaanan A, Fléjou J-F, Emile J-F, et al. Defective mismatch repair status as a prognostic biomarker of disease-free survival in stage III colon cancer patients treated with adjuvant FOLFOX chemotherapy. Clin Cancer Res. 2011;17:7470–8.PubMedCrossRef
64.
Zurück zum Zitat Sinicrope FA, Mahoney MR, Smyrk TC, et al. Prognostic impact of deficient DNA mismatch repair in patients with stage III colon cancer from a randomized trial of FOLFOX-based adjuvant chemotherapy. J Clin Oncol. 2013;31:3664–72.PubMedCrossRef Sinicrope FA, Mahoney MR, Smyrk TC, et al. Prognostic impact of deficient DNA mismatch repair in patients with stage III colon cancer from a randomized trial of FOLFOX-based adjuvant chemotherapy. J Clin Oncol. 2013;31:3664–72.PubMedCrossRef
65.
Zurück zum Zitat Fink D, Nebel S, Aebi S, et al. The role of DNA mismatch repair in platinum drug resistance. Cancer Res. 1996;56:4881–6.PubMed Fink D, Nebel S, Aebi S, et al. The role of DNA mismatch repair in platinum drug resistance. Cancer Res. 1996;56:4881–6.PubMed
66.
Zurück zum Zitat Gavin PG, Colangelo LH, Fumagalli D, et al. Mutation profiling and microsatellite instability in stage II and stage III colon cancer: an assessment of their prognostic and oxaliplatin predictive value. Clin Cancer Res. 2012;18:6531–41.PubMedCrossRef Gavin PG, Colangelo LH, Fumagalli D, et al. Mutation profiling and microsatellite instability in stage II and stage III colon cancer: an assessment of their prognostic and oxaliplatin predictive value. Clin Cancer Res. 2012;18:6531–41.PubMedCrossRef
67.
Zurück zum Zitat Burn J, Bishop T, Mecklin J-P, et al. Effect of aspirin or resistant starch on colorectal neoplasia in the Lynch syndrome. N Engl J Med. 2008;359:2567–78.PubMedCrossRef Burn J, Bishop T, Mecklin J-P, et al. Effect of aspirin or resistant starch on colorectal neoplasia in the Lynch syndrome. N Engl J Med. 2008;359:2567–78.PubMedCrossRef
68.••
Zurück zum Zitat Burn J, Gerdes A-M, Macrae F, et al. Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet. 2011;378:2081–7. This study demonstrates the importance of investigating long-term effects of chemoprevention—in this case, use of aspirin, which was not found to have a beneficial short-term effect on carriers of hereditary colorectal cancer mutations but was found to carry long-term benefits.PubMedCentralPubMedCrossRef Burn J, Gerdes A-M, Macrae F, et al. Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet. 2011;378:2081–7. This study demonstrates the importance of investigating long-term effects of chemoprevention—in this case, use of aspirin, which was not found to have a beneficial short-term effect on carriers of hereditary colorectal cancer mutations but was found to carry long-term benefits.PubMedCentralPubMedCrossRef
69.•
Zurück zum Zitat Fuchs CS, Ogino S. Aspirin therapy for colorectal cancer with PIK3CA mutation: simply complex! J Clin Oncol. 2013;31:4358–61. This paper elucidates what is known about aspirin therapy in colorectal cancer based on PIK3CA mutation status.PubMedCrossRef Fuchs CS, Ogino S. Aspirin therapy for colorectal cancer with PIK3CA mutation: simply complex! J Clin Oncol. 2013;31:4358–61. This paper elucidates what is known about aspirin therapy in colorectal cancer based on PIK3CA mutation status.PubMedCrossRef
70.
Zurück zum Zitat Baron JA, Cole BF, Sandler RS, et al. A randomized trial of aspirin to prevent colorectal adenomas. N Engl J Med. 2003;348:891–9.PubMedCrossRef Baron JA, Cole BF, Sandler RS, et al. A randomized trial of aspirin to prevent colorectal adenomas. N Engl J Med. 2003;348:891–9.PubMedCrossRef
71.
Zurück zum Zitat Baron JA, Sandler RS, Bresalier RS, et al. A randomized trial of rofecoxib for the chemoprevention of colorectal adenomas. Gastroenterology. 2006;131:1674–82.PubMedCrossRef Baron JA, Sandler RS, Bresalier RS, et al. A randomized trial of rofecoxib for the chemoprevention of colorectal adenomas. Gastroenterology. 2006;131:1674–82.PubMedCrossRef
72.
Zurück zum Zitat Logan RF, Grainge MJ, Shepherd VC, et al. Aspirin and folic acid for the prevention of recurrent colorectal adenomas. Gastroenterology. 2008;134:29–38.PubMedCrossRef Logan RF, Grainge MJ, Shepherd VC, et al. Aspirin and folic acid for the prevention of recurrent colorectal adenomas. Gastroenterology. 2008;134:29–38.PubMedCrossRef
73.
Zurück zum Zitat Domingo E, Church DN, Sieber O, et al. Evaluation of PIK3CA mutation as a predictor of benefit from nonsteroidal anti-inflammatory drug therapy in colorectal cancer. J Clin Oncol. 2013;31:4297–305.PubMedCrossRef Domingo E, Church DN, Sieber O, et al. Evaluation of PIK3CA mutation as a predictor of benefit from nonsteroidal anti-inflammatory drug therapy in colorectal cancer. J Clin Oncol. 2013;31:4297–305.PubMedCrossRef
74.•
Zurück zum Zitat Liao X, Lochhead P, Nishihara R, et al. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med. 2012;367:1596–606. This study looks at the effect of aspirin use on colorectal cancer survival in relation to a number of molecular genetic variations with important results regarding PIK3CA mutation status.PubMedCentralPubMedCrossRef Liao X, Lochhead P, Nishihara R, et al. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med. 2012;367:1596–606. This study looks at the effect of aspirin use on colorectal cancer survival in relation to a number of molecular genetic variations with important results regarding PIK3CA mutation status.PubMedCentralPubMedCrossRef
75.
Zurück zum Zitat Amatu A, Bencardino K, Sartore-Bianchi A, et al. Aspirin for colorectal cancer with PIK3CA mutations: the rising of the oldest targeted therapy? Ann Transl Med. 2013;1:12–3. Amatu A, Bencardino K, Sartore-Bianchi A, et al. Aspirin for colorectal cancer with PIK3CA mutations: the rising of the oldest targeted therapy? Ann Transl Med. 2013;1:12–3.
76.
Zurück zum Zitat Mathers JC, Movahedi M, Macrae F, et al. Long-term effect of resistant starch on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet Oncol. 2012;13:1242–9.PubMedCrossRef Mathers JC, Movahedi M, Macrae F, et al. Long-term effect of resistant starch on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet Oncol. 2012;13:1242–9.PubMedCrossRef
77.••
Zurück zum Zitat Rothwell PM, Price JF, Fowkes FGR, et al. Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials. Lancet. 2012;379:1602–12. This study analyzes the results of 51 randomized, controlled trials of aspirin.PubMedCrossRef Rothwell PM, Price JF, Fowkes FGR, et al. Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials. Lancet. 2012;379:1602–12. This study analyzes the results of 51 randomized, controlled trials of aspirin.PubMedCrossRef
78.
Zurück zum Zitat Berger JS, Lala A, Krantz MJ, et al. Aspirin for the prevention of cardiovascular events in patients without clinical cardiovascular disease: a meta-analysis of randomized trials. Am Heart J. 2011;162:115–124.e2.PubMedCrossRef Berger JS, Lala A, Krantz MJ, et al. Aspirin for the prevention of cardiovascular events in patients without clinical cardiovascular disease: a meta-analysis of randomized trials. Am Heart J. 2011;162:115–124.e2.PubMedCrossRef
79.••
Zurück zum Zitat Offit K. Personalized medicine: new genomics, old lessons. Hum Genet. 2011;130:3–14. This paper by a world-renowned hereditary cancer researcher provides an excellent, thought-provoking look at personalized medicine in relation to genomics.PubMedCentralPubMedCrossRef Offit K. Personalized medicine: new genomics, old lessons. Hum Genet. 2011;130:3–14. This paper by a world-renowned hereditary cancer researcher provides an excellent, thought-provoking look at personalized medicine in relation to genomics.PubMedCentralPubMedCrossRef
80.•
Zurück zum Zitat Morris V, Kopetz S. Clinical biomarkers in colorectal cancer. Clin Adv Hematol Oncol. 2013;11:768–76. This gives an up-to-date overview of the clinical use of biomarkers in colorectal cancer. Emphasis is given to the use of biomarkers in targeted chemotherapy.PubMed Morris V, Kopetz S. Clinical biomarkers in colorectal cancer. Clin Adv Hematol Oncol. 2013;11:768–76. This gives an up-to-date overview of the clinical use of biomarkers in colorectal cancer. Emphasis is given to the use of biomarkers in targeted chemotherapy.PubMed
81.
Zurück zum Zitat Wu TT, Rezai B, Rashid A, et al. Genetic alterations and epithelial dysplasia in juvenile polyposis syndrome and sporadic juvenile polyps. Am J Pathol. 1997;150:939–47.PubMedCentralPubMed Wu TT, Rezai B, Rashid A, et al. Genetic alterations and epithelial dysplasia in juvenile polyposis syndrome and sporadic juvenile polyps. Am J Pathol. 1997;150:939–47.PubMedCentralPubMed
82.
Zurück zum Zitat Howe JR, Mitros FA, Summers RW. The risk of gastrointestinal carcinoma in familial juvenile polyposis. Ann Surg Oncol. 1998;5:751–6.PubMedCrossRef Howe JR, Mitros FA, Summers RW. The risk of gastrointestinal carcinoma in familial juvenile polyposis. Ann Surg Oncol. 1998;5:751–6.PubMedCrossRef
83.
Zurück zum Zitat Giardiello FM, Welsh SB, Hamilton SR, et al. Increased risk of cancer in the Peutz-Jeghers syndrome. N Engl J Med. 1987;316:1511–4.PubMedCrossRef Giardiello FM, Welsh SB, Hamilton SR, et al. Increased risk of cancer in the Peutz-Jeghers syndrome. N Engl J Med. 1987;316:1511–4.PubMedCrossRef
Metadaten
Titel
Genetics, Biomarkers, Hereditary Cancer Syndrome Diagnosis, Heterogeneity and Treatment: A Review
verfasst von
Henry T. Lynch, MD
Kristen Drescher, PhD
Joseph Knezetic, PhD
Stephen Lanspa, MD
Publikationsdatum
01.09.2014
Verlag
Springer US
Erschienen in
Current Treatment Options in Oncology / Ausgabe 3/2014
Print ISSN: 1527-2729
Elektronische ISSN: 1534-6277
DOI
https://doi.org/10.1007/s11864-014-0293-5

Weitere Artikel der Ausgabe 3/2014

Current Treatment Options in Oncology 3/2014 Zur Ausgabe

Upper Gastrointestinal Cancers (L Rajdev, Section Editor)

HER2 Directed Therapy for Gastric/Esophageal Cancers

Upper Gastrointestinal Cancers (L Rajdev, Section Editor)

Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors

Lower Gastrointestinal Cancers (AB Benson, Section Editor)

Optimal Treatment Strategies for Anal Cancer

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Update Onkologie

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