I. Facteurs-de-risque-de-prédisposition-génétique-au-cancer and .. , 40 A. Facteurs de risque présents dans, p.40

T. Sjöblom, S. Jones, L. Wood, D. Parsons, J. Lin et al., The Consensus Coding Sequences of Human Breast and Colorectal Cancers, Science, vol.314, issue.5797, pp.268-74, 2006.
DOI : 10.1126/science.1133427

L. Wood, D. Parsons, S. Jones, J. Lin, T. Sjöblom et al., The genomic landscapes of human breast and colorectal cancers.Science, pp.1108-1121, 2007.

L. Kleinsmith, Science Behind the News : Understanding Cancer, Bethesda: National Cancer Institut

J. Ecsedy, Textbook of cancer epidemiology The origin of cancer, 2008.

A. Erson and E. Petty, Molecular and Genetic Events in Neoplastic Transformation, J Natl Cancer Inst, 2009.
DOI : 10.1093/acprof:oso/9780195149616.003.0004

G. Liu and H. Robins, The natural history and biology of cancer UICC Manual of Clinical Oncology, pp.1-18, 2004.

P. Futreal, R. Wooster, and M. Stratton, Somatic Mutations in Human Cancer: Insights from Resequencing the Protein Kinase Gene Family, Cold Spring Harbor Symposia on Quantitative Biology, vol.65, issue.0, pp.43-52, 2005.
DOI : 10.1002/ajmg.10775

C. Greenman, P. Stephens, R. Smith, G. Dalgliesh, C. Hunter et al., Patterns of somatic mutation in human cancer genomes, Nature, vol.62, issue.7132, pp.153-161, 2007.
DOI : 10.1038/nature05610

D. Hanahan and R. Weinberg, The hallmarks of cancer.Cell, pp.57-70, 2000.

K. Kinzler and B. Vogelstein, Gatekeepers and caretakers, Nature, vol.386, issue.6627, pp.761-763, 1997.
DOI : 10.1038/386761a0

H. Chial, Proto-oncogènes to oncogènes to cancer, Nature Education, 2008.

B. Vogelstein and K. Kinzler, Cancer genes and the pathways they control, Nature Medicine, vol.1, issue.8, pp.789-99, 2004.
DOI : 10.1038/sj.onc.1207130

J. Campisi, CANCER: Suppressing Cancer: The Importance of Being Senescent, Science, vol.309, issue.5736, pp.886-893, 2005.
DOI : 10.1126/science.1116801

M. Collado and M. Serrano, Senescence in tumours: evidence from mice and humans, Nature Reviews Cancer, vol.15, issue.1, 2010.
DOI : 10.1038/nrc2772

S. Lowe, E. Cepero, and G. Evan, Intrinsic tumour suppression, Nature, vol.35, issue.7015, pp.307-322, 2004.
DOI : 10.1038/nature03098

URL : http://www.nature.com/nature/journal/v432/n7015/pdf/nature03098.pdf

T. Kuilman, C. Michaloglou, W. Mooi, and D. Peeper, The essence of senescence, Genes & Development, vol.24, issue.22, pp.2463-79, 2010.
DOI : 10.1101/gad.1971610

A. Sarasin, Les gènes Humains de la réparation de l'ADN. Médecine/Sciences, 1994.

A. Knudson, Cancer genetics, Am J Med Genet Jul, vol.22111, issue.1, pp.96-102, 2002.

A. Knudson, Mutation and cancer: statistical study of retinoblastoma. ProcNatlAcadSci U S A, 1971.

D. Comings, A general theory of carcinogenesis.ProcNatlAcadSci U S A, pp.3324-3332, 1973.

S. Friend, R. Bernards, S. Rogelj, R. Weinberg, J. Rapaport et al., A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma, Nature, vol.4, issue.6089, pp.643-649, 1986.
DOI : 10.1038/323643a0

A. Machens, P. Niccoli-sire, J. Hoegel, K. Frank-raue, T. Van-vroonhoven et al., Early Malignant Progression of Hereditary Medullary Thyroid Cancer, New England Journal of Medicine, vol.349, issue.16, pp.1517-1542, 2003.
DOI : 10.1056/NEJMoa012915

I. Schuffenecker, B. Chambe, and G. Lenoir, Analysis of the RET gene and medullary cancer of the thyroid. Contribution to the diagnosis and treatment], Ann Endocrinol, vol.57, issue.1, pp.9-14, 1996.

A. Sidle, C. Palaty, P. Dirks, O. Wiggan, M. Kiess et al., Activity of the Retinoblastoma Family Proteins, pRB, p107, and p130, during Cellular Proliferation and Differentiation, Critical Reviews in Biochemistry and Molecular Biology, vol.15, issue.3, pp.237-71, 1996.
DOI : 10.1001/archpedi.1978.02120260032004

A. Manning and N. Dyson, pRB, a tumor suppressor with a stabilizing presence, Trends in Cell Biology, vol.21, issue.8, 2011.
DOI : 10.1016/j.tcb.2011.05.003

S. Boulton, BRCA1-mediated ubiquitylation. Cell Cycle Georget Tex, pp.1481-1487, 2006.
DOI : 10.4161/cc.5.14.2930

C. Harris, p53: at the crossroads of molecular carcinogenesis and risk assessment, Science, vol.262, issue.5142, 1993.
DOI : 10.1126/science.8266092

D. Malkin, F. Li, L. Strong, J. Fraumeni, C. Nelson et al., Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms, Science, vol.250, issue.4985, pp.1233-1241, 1990.
DOI : 10.1126/science.1978757

D. Haber, A. Buckler, T. Glaser, K. Call, J. Pelletier et al., An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms' tumor. Cell, pp.1257-69, 1990.

A. Menon, J. Gusella, and B. Seizinger, Progress toward the Isolation and Characterization of the Genes Causing Neurofibromatosis, Brain Pathology, vol.46, issue.1, pp.33-40, 1990.
DOI : 10.1016/0092-8674(90)90253-B

W. Bodmer, C. Bailey, J. Bodmer, H. Bussey, A. Ellis et al., Localization of the gene for familial adenomatous polyposis on chromosome 5, Nature, vol.328, issue.6131, pp.614-620, 1987.
DOI : 10.1038/328614a0

J. Groden, A. Thliveris, W. Samowitz, M. Carlson, L. Gelbert et al., Identification and characterization of the familial adenomatous polyposis coli gene, Cell, vol.66, issue.3, pp.589-600, 1991.
DOI : 10.1016/0092-8674(81)90021-0

Y. Nakamura, I. Nishisho, K. Kinzler, B. Vogelstein, Y. Miyoshi et al., Mutations of the APC(Adenomatous Polyposis Coli) Gene in FAP(Familial Polyposis Coli) Patients and in Sporadic Colorectal Tumors., The Tohoku Journal of Experimental Medicine, vol.168, issue.2, pp.285-92, 1991.
DOI : 10.1620/tjem.168.141

S. Narod, D. Parry, J. Parboosingh, G. Lenoir, M. Ruttledge et al., Neurofibromatosis type 2 appears to be a genetically homogeneous disease, Am J Hum Genet, vol.51, issue.3, pp.486-96, 1992.

F. Latif, K. Tory, J. Gnarra, M. Yao, F. Duh et al., Identification of the von Hippel-Lindau disease tumor suppressor gene, Science, vol.260, issue.5112, pp.1317-1337, 1993.
DOI : 10.1126/science.8493574

J. Whaley, J. Naglich, L. Gelbert, Y. Hsia, J. Lamiell et al., Germ-line mutations in the von Hippel-Lindau tumor-suppressor gene are similar to somatic von Hippel-Lindau aberrations in sporadic renal cell carcinoma, Am J Hum Genet, vol.55, issue.6, pp.1092-102, 1994.

L. Mulligan, J. Kwok, C. Healey, M. Elsdon, C. Eng et al., Germ-line mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A, Nature, vol.363, issue.6428, pp.458-60, 1993.
DOI : 10.1038/363458a0

S. Baker, A. Plug, T. Prolla, C. Bronner, A. Harris et al., Involvement of mouse Mlh1 in DNA mismatch repair and meiotic crossing over, Nature Genetics, vol.12, issue.3, pp.336-378, 1996.
DOI : 10.1083/jcb.105.1.93

C. Wyman, D. Ristic, and R. Kanaar, Homologous recombination-mediated double-strand break repair. DNA Repair, pp.8-9827, 2004.
DOI : 10.1016/j.dnarep.2004.03.037

D. Bishop, B. Brca1, and B. , BRCA1, BRCA2, BRCA3??? A myriad of breast cancer genes, European Journal of Cancer, vol.30, issue.12, pp.1738-1747, 1990.
DOI : 10.1016/0959-8049(94)00455-E

N. Nicolaides, N. Papadopoulos, B. Liu, Y. Wei, K. Carter et al., Mutations of two P/WS homologues in hereditary nonpolyposis colon cancer, Nature, vol.371, issue.6492, pp.75-80, 1994.
DOI : 10.1038/371075a0

M. King, The race " to clone BRCA1.Science, pp.1462-1467, 2014.

B. Knoppers and R. Chadwick, le projet du génome humain : aspects éthiques sous un angle international " Recueil international de législation sanitaire, 1995.

H. Lynch and A. De-la-chapelle, Hereditary colorectal cancer, N Engl J Med, vol.348, issue.10, pp.919-951, 2003.

H. Lynch, E. Silva, C. Snyder, and J. Lynch, Hereditary Breast Cancer: Part I. Diagnosing Hereditary Breast Cancer Syndromes, The Breast Journal, vol.93, issue.1, pp.3-13, 2008.
DOI : 10.1111/j.1524-4741.2007.00515.x

M. Daly, J. Axilbund, S. Buys, C. B. Farrell, C. Friedman et al., Genetic/familial high-risk assessment: breast and ovarian, J NatlCompr Cancer Netw JNCCN, vol.8, issue.5, pp.562-94, 2010.
DOI : 10.6004/jnccn.2010.0043

D. Pauw, A. Jolissaint, L. Fréneaux, P. Rouleau, E. Stoppa-lyonnet et al., [Hereditary forms of ovarian cancer]. Bull Cancer (Paris), pp.453-62, 2012.

S. Ramus and S. Gayther, The contribution of BRCA1 and BRCA2 to ovarian cancer.MolOncol, 2009.

M. Dunlop, S. Farrington, I. Nicholl, L. Aaltonen, G. Petersen et al., Population carrier frequency of hMSH2 and hMLH1 mutations, British Journal of Cancer, vol.83, issue.12, pp.1643-1648, 2000.
DOI : 10.1054/bjoc.2000.1520

B. Bonaïti, F. Alarcon, V. Bonadona, S. Pennec, N. Andrieu et al., A new scoring system for the diagnosis of BRCA1/2 associated breast-ovarian cancer predisposition], Bull Cancer, 2011.

G. Petersen, J. Brensinger, K. Johnson, and F. Giardiello, Genetic testing and counseling for hereditary forms of colorectal cancer, Cancer, vol.150, issue.S8, pp.2540-50, 1999.
DOI : 10.1002/(SICI)1097-0142(19991015)86:8+<1720::AID-CNCR11>3.0.CO;2-0

M. Esplen, L. Madlensky, K. Butler, W. Mckinnon, B. Bapat et al., Motivations and psychosocial impact of genetic testing for HNPCC, American Journal of Medical Genetics, vol.8, issue.1, pp.9-15, 2001.
DOI : 10.1002/ajmg.1493

S. Vadaparampil, M. Ropka, and M. Stefanek, Measurement of psychological factors associated with genetic testing for hereditary breast, ovarian and colon cancers, Familial Cancer, vol.44, issue.102, pp.195-206, 2005.
DOI : 10.1007/s10689-004-1446-7

F. Collins, ??? Lots of Mutations, Lots of Dilemmas, New England Journal of Medicine, vol.334, issue.3, pp.186-194, 1996.
DOI : 10.1056/NEJM199601183340311

D. Bonneau, S. Marlin, D. Sanlaville, J. Dupont, H. Sobol et al., Genetic testing in the context of the revision of the French law on bioethics].PatholBiol (Paris), pp.396-401, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00461832

P. Pujol, D. Lyonnet, T. Frebourg, J. Blin, M. Picot et al., Lack of referral for genetic counseling and testing in BRCA1/2 and Lynch syndromes: a nationwide study based on 240,134 consultations and 134,652 genetic tests, Breast Cancer Research and Treatment, vol.75, issue.2193, pp.135-179, 2013.
DOI : 10.1007/s10549-013-2669-9

B. Buecher, Consultations d'oncogénétique. Journées Francophones d'Hépato-gastroentérologie et d'Oncologie Digestive, 2014.

A. Bourrillon and J. Chouraqui, Pédiatrie pour le praticien. Issy-les-Moulineaux, 2008.

E. Desandes, B. Lacour, A. Belot, F. Molinie, P. Delafosse et al., Cancer incidence and survival in adolescents and young adults in France, pp.291-306, 2000.
URL : https://hal.archives-ouvertes.fr/hal-00487242

B. Strahm and D. Malkin, Hereditary cancer predisposition in children: Genetic basis and clinical implications, International Journal of Cancer, vol.285, issue.9, 2006.
DOI : 10.1002/ijc.21962

D. Friedman, N. Kadan-lottick, J. Whitton, A. Mertens, Y. Yasui et al., Increased risk of cancer among siblings of long-term childhood cancer survivors: a report from the childhood cancer survivor study. Cancer EpidemiolBiomarkPrevPubl Am Assoc Cancer Res Cosponsored Am SocPrevOncol, 2005.

A. Meadows, D. Friedman, J. Neglia, A. Mertens, S. Donaldson et al., Second Neoplasms in Survivors of Childhood Cancer: Findings From the Childhood Cancer Survivor Study Cohort, Journal of Clinical Oncology, vol.27, issue.14, pp.2356-62, 2009.
DOI : 10.1200/JCO.2008.21.1920

D. Sommelet, J. Clavel, and B. Lacour, E?pidémiologie des cancers de l'enfant [Internet]. Paris, 2009.

H. Ganjavi and D. Malkin, Genetics of Childhood Cancer, Clinical Orthopaedics and Related Research, vol.401, issue.401, pp.75-87, 2002.
DOI : 10.1097/00003086-200208000-00011

S. Narod, C. Stiller, and G. Lenoir, An estimate of the heritable fraction of childhood cancer, British Journal of Cancer, vol.63, issue.6, pp.993-1002, 1991.
DOI : 10.1038/bjc.1991.216

S. Pakakasama and G. Tomlinson, Genetic predisposition and screening in pediatric cancer. PediatrClin North Am, pp.1393-413, 2002.

S. Knapke, R. Nagarajan, J. Correll, D. Kent, and K. Burns, Hereditary cancer risk assessment in a pediatric oncology follow-up clinic. Pediatr Blood Cancer, pp.85-94, 2012.

S. Plon and K. Nathanson, Inherited Susceptibility for Pediatric Cancer, The Cancer Journal, vol.11, issue.4, 2005.
DOI : 10.1097/00130404-200507000-00002

M. Gauthier-villars and D. Stoppa-lyonnet, Genetic predisposition in children cancers in, Bull Cancer, vol.98, issue.5, pp.459-75, 2011.

C. Abadie, M. Gauthier-villars, N. Sirvent, and I. Coupier, Genetic predisposition to childhood cancer]. Arch PédiatrieOrgane Off Sociéte Fr Pédiatrie, pp.863-75, 2012.

F. Doz, Retinoblatoma: a review] Arch PédiatrieOrgane Off Sociéte Fr Pédiatrie, pp.1329-1366, 2006.

M. Gauthier-villars, G. Schleiermacher, and I. Coupier, Prédisposition génétique aux cancers de l'enfant. Le point en 2003 Médecine thérapeutique/Pédiatrie, 2003.

A. Teplick, M. Kowalski, J. Biegel, and K. Nichols, Educational paper, European Journal of Pediatrics, vol.149, issue.4, pp.285-94, 2011.
DOI : 10.1007/s00431-010-1377-2

D. Héron, Rôle du généticien clinicien en pédiatrie Médecine thérapeutique/Pédiatrie, 2014.

C. Colas, Conseils génétiques en oncologie pédiatrique Médecine thérapeutique/Pédiatrie, 2014.

M. Tischkowitz and E. Rosser, Inherited cancer in children: practical/ethical problems and challenges, European Journal of Cancer, vol.40, issue.16, pp.2459-70, 1990.
DOI : 10.1016/j.ejca.2004.06.005

G. Jeanblanc, M. Michel, and C. Rumeau-pichon, Dépistage du cancer du sein en France : identification des femmes à haut risque et modalités de dépistage, HAS Avril, vol.53, 2011.

F. Li and J. Fraumeni, Soft-tissue sarcomas, breast cancer, and other neoplasms.A familial syndrome? Ann Intern Med, pp.747-52, 1969.

A. Chompret, L. Brugières, M. Ronsin, M. Gardes, F. Dessarps-freichey et al., P53 germline mutations in childhood cancers and cancer risk for carrier individuals, Br J Cancer, vol.82, issue.12, pp.1932-1939, 2000.
URL : https://hal.archives-ouvertes.fr/inserm-00117136

T. Frebourg, A. Abel, C. Bonaiti-pellie, L. Brugières, P. Berthet et al., Li-Fraumeni syndrome: update, new data and guidelines for clinical management], Bull Cancer, 2001.

F. Gauthier-villars, F. Doz, and . Desjardins, Le conseil génétique du rétinoblastome, pp.35-40, 2003.

S. Pinson, A. Créange, S. Barbarot, J. Stalder, Y. Chaix et al., Arch Pédiatrie Organe Off Sociéte Fr Pédiatrie, Neurofibromatosis, vol.19, issue.1, pp.49-60, 2002.

V. Williams, J. Lucas, M. Babcock, D. Gutmann, B. Korf et al., Neurofibromatosis type 1 revisited. Pediatrics, pp.124-157, 2009.
DOI : 10.1542/peds.2007-3204

D. Evans, P. Farndon, L. Burnell, H. Gattamaneni, and J. Birch, The incidence of Gorlin syndrome in 173 consecutive cases of medulloblastoma, British Journal of Cancer, vol.64, issue.5, pp.959-61, 1991.
DOI : 10.1038/bjc.1991.435

S. Amlashi, L. Riffaud, G. Brassier, and X. Morandi, Nevoid basal cell carcinoma syndrome: Relation with desmoplastic medulloblastoma in infancy, Cancer, vol.37, issue.3, pp.618-642, 2003.
DOI : 10.1002/cncr.11537

N. Sévenet, A. Lellouch-tubiana, D. Schofield, K. Hoang-xuan, M. Gessler et al., Spectrum of hSNF5IINI1 Somatic Mutations in Human Cancer and Genotype-Phenotype Correlations, Human Molecular Genetics, vol.8, issue.13, pp.2359-68, 1999.
DOI : 10.1093/hmg/8.13.2359

F. Bourdeaut, D. Lequin, L. Brugières, S. Reynaud, C. Dufour et al., Frequent hSNF5/INI1 Germline Mutations in Patients with Rhabdoid Tumor, Clinical Cancer Research, vol.17, issue.1, pp.31-39, 2011.
DOI : 10.1158/1078-0432.CCR-10-1795

J. Varley, G. Mcgown, M. Thorncroft, L. James, G. Margison et al., Are There Low-Penetrance TP53 Alleles? Evidence from Childhood Adrenocortical Tumors, The American Journal of Human Genetics, vol.65, issue.4, pp.995-1006, 1999.
DOI : 10.1086/302575

J. Wagner, C. Portwine, K. Rabin, J. Leclerc, S. Narod et al., High Frequency of Germline p53 Mutations in Childhood Adrenocortical Cancer, JNCI Journal of the National Cancer Institute, vol.86, issue.22, pp.1707-1717, 1994.
DOI : 10.1093/jnci/86.22.1707

M. Steenman, A. Westerveld, and M. Mannens, Genetics of Beckwith-Wiedemann syndrome-associated tumors: Common genetic pathways, Genes, Chromosomes and Cancer, vol.1, issue.1, pp.1-13, 2000.
DOI : 10.1002/(SICI)1098-2264(200005)28:1<1::AID-GCC1>3.0.CO;2-#

A. Feinberg, Multiple genetic abnormalities of 11p15 in Wilms' tumor. Med PediatrOncol, pp.484-493, 1996.

A. Ward, Beck-Wiedemann syndrome and Wilms' tumour, Molecular Human Reproduction, vol.3, issue.2, pp.157-68, 1997.
DOI : 10.1093/molehr/3.2.157

I. Janoueix-lerosey, D. Lequin, L. Brugières, A. Ribeiro, L. De-pontual et al., Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma, Nature, vol.40, issue.7215, pp.967-70, 2008.
DOI : 10.1038/nature07398

Y. Mossé, M. Laudenslager, L. Longo, K. Cole, A. Wood et al., Identification of ALK as a major familial neuroblastoma predisposition gene Triplication of a 21q22 region contributes to B cell transformation through HMGN1 overexpression and loss of histone H3 Lys27 trimethylation, Nature Nat Genet, vol.45546, issue.72156, pp.618-641, 2008.

K. Shannon, O. Connell, P. Martin, G. Paderanga, D. Olson et al., Loss of the normal NF1 allele from the bone marrow of children with type 1 neurofibromatosis and malignant myeloid disorders Neurofibromatosis type 1-associated tumours: their somatic mutational spectrum and pathogenesis. Hum Genomics Neoplasms associated with germline and somatic NF1 gene mutations. The Oncologist The NF1 gene revisited -from bench to bedside Incidence of childhood cancer in France: National Children Cancer Registries The international classification of childhood cancerCancer genetic predisposition: current events and perspectives, TandonnetArégui J. Leucémie aiguë myéloblastique de l'enfant trisomique 21597?601. 104. Laycock-van Spyk S5873?92. 107. Lacour B173?81. 108. Steliarova-Foucher E1457?67. 109. Kramárová E, Stiller CA, pp.101-117, 1990.

D. Easton and J. Peto, The contribution of inherited predisposition to cancer incidence, Cancer Surv, vol.9, issue.3, pp.395-416, 1990.