J. Ferlay, D. M. Parkin, and E. Steliarova-foucher, Estimates of cancer incidence and mortality in Europe in 2008, Eur J Cancer. mars, vol.46, issue.4, pp.765-81, 2010.

, Le cancer du sein -Les cancers les plus fréquents

D. Sur,

A. Prat, C. Fan, A. Fernández, K. A. Hoadley, R. Martinello et al., Response and survival of breast cancer intrinsic subtypes following multi-agent neoadjuvant chemotherapy, BMC Med. 18 déc, vol.13, issue.1, p.303, 2015.

X. Dai, T. Li, Z. Bai, Y. Yang, X. Liu et al., Breast cancer intrinsic subtype classification, clinical use and future trends, Am J Cancer Res, vol.5, issue.10, pp.2929-2972, 2015.

M. Espié, Prise en charge du cancer du sein, J Radiol Diagn Interv. juill, vol.95, issue.7-8, pp.740-744, 2014.

M. Filipits, T. O. Nielsen, M. Rudas, R. Greil, H. Stöger et al., The PAM50 Riskof-Recurrence Score Predicts Risk for Late Distant Recurrence after Endocrine Therapy in Postmenopausal Women with Endocrine-Responsive Early Breast Cancer, Clin Cancer Res. 1 mars, vol.20, issue.5, pp.1298-305, 2014.

B. Wallden, J. Storhoff, T. Nielsen, N. Dowidar, C. Schaper et al., Development and verification of the PAM50-based Prosigna breast cancer gene signature assay, BMC Med Genomics. déc, vol.8, issue.1, p.54, 2015.

, Poursuivre la recherche clinique pour positionner utilement les signatures génomiques dans la prise en charge des cancers du sein

H. Autorité-de-santé, Disponible sur

P. F. Peddi, Hormone receptor positive breast cancer: state of the art, Curr Opin Obstet Gynecol. déc, vol.1, 2017.

M. Pathak, S. N. Dwivedi, S. Deo, B. Thakur, V. Sreenivas et al., Effectiveness of Added Targeted Therapies to Neoadjuvant Chemotherapy for Breast Cancer: A Systematic Review and Meta-analysis, Clin Breast Cancer. juin, p.1526820919303234, 2019.

, Comparisons between different polychemotherapy regimens for early breast cancer: meta-analyses of longterm outcome among 100 000 women in 123 randomised trials, The Lancet. févr, vol.379, issue.9814, pp.432-476, 2012.

M. C. Strach, T. Prasanna, Y. M. Kirova, S. Alran, S. O'toole et al., Optimise not compromise: The importance of a multidisciplinary breast cancer patient pathway in the era of oncoplastic and reconstructive surgery, Crit Rev Oncol Hematol. févr, vol.134, pp.10-21, 2019.

, Haute Autorité de santé définissant les actes et prestations pour l'ALD n° 30 « Tumeur maligne

S. R. Lakhani, Pathology of Ovarian Cancers in BRCA1 and BRCA2 Carriers, Clin Cancer Res. 1 avr, vol.10, issue.7, pp.2473-81, 2004.

J. M. Helder-woolderink, E. A. Blok, H. Vasen, H. Hollema, M. J. Mourits et al., Ovarian cancer in Lynch syndrome; a systematic review, Eur J Cancer. mars, vol.55, pp.65-73, 2016.

A. Gadducci, V. Guarneri, F. A. Peccatori, G. Ronzino, G. Scandurra et al., Current strategies for the targeted treatment of high-grade serous epithelial ovarian cancer and relevance of BRCA mutational status, J Ovarian Res. déc, vol.12, issue.1, p.9, 2019.

C. Aghajanian, S. V. Blank, B. A. Goff, P. L. Judson, M. G. Teneriello et al., OCEANS: A Randomized, Double-Blind, Placebo-Controlled Phase III Trial of Chemotherapy With or Without Bevacizumab in Patients With Platinum-Sensitive Recurrent Epithelial Ovarian, Primary Peritoneal, or Fallopian Tube Cancer, J Clin Oncol. 10 juin, vol.30, issue.17, pp.2039-2084, 2012.

C. Aghajanian, B. Goff, L. R. Nycum, Y. V. Wang, A. Husain et al., Final overall survival and safety analysis of OCEANS, a phase 3 trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent ovarian cancer, Gynecol Oncol, vol.139, issue.1, pp.10-16, 2015.

J. Ledermann, P. Harter, C. Gourley, M. Friedlander, I. Vergote et al., Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial, Lancet Oncol. juill, vol.15, issue.8, pp.852-61, 2014.

J. A. Ledermann, P. Harter, C. Gourley, M. Friedlander, I. Vergote et al., Overall survival in patients with platinum-sensitive recurrent serous ovarian cancer receiving olaparib maintenance monotherapy: an updated analysis from a randomised, placebocontrolled, double-blind, phase 2 trial, Lancet Oncol, vol.17, issue.11, pp.1579-89, 2016.

S. M. Weissman, S. M. Weiss, and A. C. Newlin, Genetic Testing by Cancer Site: Ovary, Cancer J, vol.18, issue.4, pp.320-327, 2012.

Y. Miki, J. Swensen, D. Shattuck-eidens, P. Futreal, K. Harshman et al., A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1, Science, vol.266, issue.5182, pp.66-71, 1994.

R. Wooster, G. Bignell, J. Lancaster, S. Swift, S. Seal et al., Identification of the breast cancer susceptibility gene BRCA2, Nature. déc, vol.378, issue.6559, pp.789-92, 1995.

, Oncogénétique en 2016 -Consultations, laboratoires et suivi -Ref : ADONCOG17 [Internet

A. G. Knudson, Mutation and Cancer: Statistical Study of Retinoblastoma, Proc Natl Acad Sci. 1 avr, vol.68, issue.4, pp.820-823, 1971.

F. Bunz, Principles of Cancer Genetics, Bunz F, éditeur, pp.75-134, 2016.

J. Hall, M. Lee, B. Newman, J. Morrow, L. Anderson et al., Linkage of earlyonset familial breast cancer to chromosome 17q21, Science. 21 déc, vol.250, issue.4988, pp.1684-1693, 1990.

R. Wooster, S. Neuhausen, J. Mangion, Y. Quirk, D. Ford et al., Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science. 30 sept, vol.265, pp.2088-90, 1994.

E. R. Gold and J. Carbone, Myriad Genetics: In the eye of the policy storm, Genet Med Off J Am Coll Med Genet. avr, vol.12, issue.4, pp.39-70, 2010.

, Association for Molecular Pathology et al. against US Patent and Trademark Office, Document, vol.255

C. Cropper, A. Woodson, B. Arun, C. Barcenas, J. Litton et al., Evaluating the NCCN Clinical Criteria for Recommending BRCA1 and BRCA2 Genetic Testing in Patients With Breast Cancer, J Natl Compr Canc Netw. juin, vol.15, issue.6, pp.797-803, 2017.

N. Petrucelli, M. B. Daly, and G. L. Feldman, Hereditary breast and ovarian cancer due to mutations in BRCA1 and BRCA2, Genet Med. mai, vol.12, issue.5, pp.245-59, 2010.

K. B. Kuchenbaecker, J. L. Hopper, D. R. Barnes, K. Phillips, T. M. Mooij et al., Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers, JAMA. 20 juin, vol.317, issue.23, p.2402, 2017.

E. Castro and R. Eeles, The role of BRCA1 and BRCA2 in prostate cancer, Asian J Androl. mai, vol.14, issue.3, pp.409-423, 2012.

J. Iqbal, A. Ragone, J. Lubinski, H. T. Lynch, P. Moller et al., The incidence of pancreatic cancer in BRCA1 and BRCA2 mutation carriers, Br J Cancer, vol.107, 2005.

A. L. Lucas, L. E. Frado, C. Hwang, S. Kumar, L. G. Khanna et al., BRCA1 and BRCA2 germline mutations are frequently demonstrated in both high-risk pancreatic cancer screening and pancreatic cancer cohorts: BRCA1/2 Germline Mutations in Pancreatic Cancer, Cancer. 1 juill, vol.120, issue.13, pp.1960-1967, 2014.

P. Keefe, S. Bokhari, and . Fanconi-syndrome, Treasure Island (FL): StatPearls Publishing, StatPearls [Internet, 2019.

B. Xia, Q. Sheng, K. Nakanishi, A. Ohashi, J. Wu et al., Control of BRCA2 Cellular and Clinical Functions by a Nuclear Partner, PALB2. Mol Cell. juin, vol.22, issue.6, pp.719-748, 2006.

A. C. Antoniou, S. Casadei, T. Heikkinen, D. Barrowdale, K. Pylkäs et al., Breast-Cancer Risk in Families with Mutations in PALB2, N Engl J Med. 7 août, vol.371, issue.6, pp.497-506, 2014.

E. R. Thompson, S. M. Rowley, N. Li, S. Mcinerny, L. Devereux et al., Panel Testing for Familial Breast Cancer: Calibrating the Tension Between Research and Clinical Care, J Clin Oncol. mai, vol.34, issue.13, pp.1455-1464, 2016.

L. Castéra, V. Harter, E. Muller, S. Krieger, N. Goardon et al., Landscape of pathogenic variations in a panel of 34 genes and cancer risk estimation from 5131 HBOC families, Genet Med Off J Am Coll Med Genet, vol.20, issue.12, pp.1677-86, 2018.

M. C. Southey, D. E. Goldgar, R. Winqvist, K. Pylkäs, F. Couch et al., CHEK2 and ATM rare variants and cancer risk: data from COGS, J Med Genet. déc, vol.53, issue.12, pp.800-811, 2016.

C. Loveday, C. Turnbull, E. Ruark, R. Xicola, E. Ramsay et al., Germline RAD51C mutations confer susceptibility to ovarian cancer, Nat Genet. 26 avr, vol.44, p.475, 2012.

C. Loveday, C. Turnbull, E. Ramsay, D. Hughes, E. Ruark et al., Germline mutations in RAD51D confer susceptibility to ovarian cancer, Nat Genet. 7 août, vol.43, p.879, 2011.

N. Li, S. Mcinerny, M. Zethoven, D. Cheasley, B. Lim et al., Combined Tumor Sequencing and Case-Control Analyses of RAD51C in Breast Cancer, JNCI J Natl Cancer Inst. 5 avr, p.45, 2019.

A. Kourtidis, R. Lu, L. J. Pence, and P. Z. Anastasiadis, A central role for cadherin signaling in cancer, Exp Cell Res. sept, vol.358, issue.1, pp.78-85, 2017.

S. Hansford, P. Kaurah, H. Li-chang, M. Woo, J. Senz et al., Hereditary Diffuse Gastric Cancer Syndrome: CDH1 Mutations and Beyond, JAMA Oncol. 1 avr, vol.1, issue.1, p.23, 2015.

R. Pilarski, R. Burt, W. Kohlman, L. Pho, K. M. Shannon et al., Cowden syndrome and the PTEN hamartoma tumor syndrome: systematic review and revised diagnostic criteria, J Natl Cancer Inst, vol.105, issue.21, pp.1607-1623, 2013.

D. L. Riegert-johnson, F. C. Gleeson, M. Roberts, K. Tholen, L. Youngborg et al., Cancer and Lhermitte-Duclos disease are common in Cowden syndrome patients, Hered Cancer Clin Pract. 17 juin, vol.8, issue.1, p.6, 2010.

V. Bubien, F. Bonnet, V. Brouste, S. Hoppe, E. Barouk-simonet et al., High cumulative risks of cancer in patients with PTEN hamartoma tumour syndrome, J Med Genet. avr, vol.50, issue.4, pp.255-63, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02168104

V. Bonadona, Cancer Risks Associated With Germline Mutations in MLH1, MSH2, and MSH6 Genes in Lynch Syndrome, JAMA. 8 juin, vol.305, issue.22, p.2304, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00780536

G. Bougeard, M. Renaux-petel, J. Flaman, C. Charbonnier, P. Fermey et al.,

, Revisiting Li-Fraumeni Syndrome From TP53 Mutation Carriers, J Clin Oncol. 20 juill, vol.33, issue.21, pp.2345-52, 2015.

E. Kasper, E. Angot, E. Colasse, L. Nicol, J. Sabourin et al., Contribution of genotoxic anticancer treatments to the development of multiple primary tumours in the context of germline TP53 mutations, Eur J Cancer. sept, vol.101, pp.254-62, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02375757

K. C. De-andrade, L. Mirabello, D. R. Stewart, E. Karlins, R. Koster et al., Higherthan-expected population prevalence of potentially pathogenic germline TP53 variants in individuals unselected for cancer history, Hum Mutat. déc, vol.38, issue.12, pp.1723-1753, 2017.

J. Moretta, P. Berthet, V. Bonadona, O. Caron, O. Cohen-haguenauer et al., Recommandations françaises du Groupe Génétique et Cancer pour l'analyse en panel de gènes dans les prédispositions héréditaires au cancer du sein ou de l'ovaire, Bull Cancer, vol.105, issue.10, pp.907-924, 2018.

W. Liu, D. I. Smith, K. J. Rechtzigel, S. N. Thibodeau, and C. D. James, Denaturing high performance liquid chromatography (DHPLC) used in the detection of germline and somatic mutations, Nucleic Acids Res. 1 mars, vol.26, issue.6, pp.1396-400, 1998.

A. Riahi, M. Kharrat, I. Lariani, and H. Chaabouni-bouhamed, High-resolution melting (HRM) assay for the detection of recurrent BRCA1/BRCA2 germline mutations in Tunisian breast/ovarian cancer families, Fam Cancer. déc, vol.13, issue.4, pp.603-612, 2014.

V. Caux-moncoutier, L. Castéra, C. Tirapo, D. Michaux, M. Rémon et al.,

, EMMA, a cost-and time-effective diagnostic method for simultaneous detection of point mutations and large-scale genomic rearrangements: application to BRCA1 and BRCA2 in 1,525 patients, Hum Mutat. mars, vol.32, issue.3, pp.325-359, 2011.

B. Langmead, C. Trapnell, M. Pop, and S. L. Salzberg, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome, Genome Biol, vol.10, issue.3, p.25, 2009.

M. Gerlinger, A. J. Rowan, S. Horswell, J. Larkin, D. Endesfelder et al., Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing, N Engl J Med. 8 mars, vol.366, issue.10, pp.883-92, 2012.

E. Muller, N. Goardon, B. Brault, A. Rousselin, G. Paimparay et al., OutLyzer: software for extracting low-allele-frequency tumor mutations from sequencing background noise in clinical practice, Oncotarget, vol.7, issue.48, 2016.

K. Wang, M. Li, and H. Hakonarson, ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data, Nucleic Acids Res. 1 sept, vol.38, issue.16, pp.164-164, 2010.

J. T. Den-dunnen, R. Dalgleish, D. R. Maglott, R. K. Hart, M. S. Greenblatt et al., HGVS Recommendations for the Description of Sequence Variants, Update. Hum Mutat. juin, vol.37, issue.6, pp.564-573, 2016.

S. Richards, N. Aziz, S. Bale, D. Bick, S. Das et al., Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology, Genet Med Off J Am Coll Med Genet. mai, vol.17, issue.5, pp.405-429, 2015.

S. E. Plon, D. M. Eccles, D. Easton, W. D. Foulkes, M. Genuardi et al., Sequence variant classification and reporting: recommendations for improving the interpretation of cancer susceptibility genetic test results, Hum Mutat, vol.29, issue.11, pp.1282-91, 2008.

S. T. Sherry, M. H. Ward, M. Kholodov, J. Baker, L. Phan et al., dbSNP: the NCBI database of genetic variation, Nucleic Acids Res. 1 janv, vol.29, issue.1, pp.308-319, 2001.

C. Béroud, G. Collod-béroud, C. Boileau, T. Soussi, and J. C. Umd, Universal mutation database): a generic software to build and analyze locus-specific databases, Hum Mutat, vol.15, issue.1, pp.86-94, 2000.

C. Béroud, S. I. Letovsky, C. D. Braastad, S. M. Caputo, O. Beaudoux et al., BRCA Share: A Collection of Clinical BRCA Gene Variants: HUMAN MUTATION, Hum Mutat. déc, vol.37, issue.12, pp.1318-1346, 2016.

M. S. Cline, R. G. Liao, M. T. Parsons, P. B. Alquaddoomi, F. Antoniou et al., BRCA Exchange as a global resource for variants in BRCA1 and BRCA2. Eng C, éditeur, PLOS Genet. 26 déc, vol.14, issue.12, p.1007752, 2018.

I. A. Adzhubei, S. Schmidt, L. Peshkin, V. E. Ramensky, A. Gerasimova et al., A method and server for predicting damaging missense mutations, Nat Methods. avr, vol.7, issue.4, pp.248-257, 2010.

F. Desmet, D. Hamroun, M. Lalande, G. Collod-béroud, M. Claustres et al., Human Splicing Finder: an online bioinformatics tool to predict splicing signals, Nucleic Acids Res. mai, vol.37, issue.9, pp.67-67, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00396239

G. Yeo and C. B. Burge, Maximum Entropy Modeling of Short Sequence Motifs with Applications to RNA Splicing Signals, J Comput Biol. mars, vol.11, issue.2-3, pp.377-94, 2004.

R. Leman, P. Gaildrat, G. L. Gac, C. Ka, Y. Fichou et al., Novel diagnostic tool for prediction of variant spliceogenicity derived from a set of 395 combined in silico/in vitro studies: an international collaborative effort, Nucleic Acids Res. 6 sept, vol.46, issue.15, pp.7913-7936, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01910334

C. Gilissen, J. Y. Hehir-kwa, D. T. Thung, M. Van-de-vorst, B. Van-bon et al., Genome sequencing identifies major causes of severe intellectual disability, Nature. juill, vol.511, issue.7509, pp.344-351, 2014.

P. J. Ainsworth, P. K. Chakraborty, and R. Weksberg, Example of somatic mosaicism in a series of de novo neurofibromatosis type 1 cases due to a maternally derived deletion, Hum Mutat, vol.9, issue.5, pp.452-459, 1997.

D. Evans, A. J. Wallace, C. L. Wu, L. Trueman, R. T. Ramsden et al., Somatic Mosaicism: A Common Cause of Classic Disease in Tumor-Prone Syndromes? Lessons from Type 2 Neurofibromatosis, Am J Hum Genet. sept, vol.63, issue.3, pp.727-763, 1998.

H. Youssoufian and R. E. Pyeritz, Mechanisms and consequences of somatic mosaicism in humans, Nat Rev Genet, vol.3, issue.10, pp.748-58, 2002.

J. G. Hall, Review and hypotheses: somatic mosaicism: observations related to clinical genetics, Am J Hum Genet, vol.43, issue.4, pp.355-63, 1988.

H. J. Parkhurst, . Cleveland, and . Society, Arch Dermatol, vol.24, issue.4, p.712, 1931.

M. T. García-romero, P. Parkin, and I. Lara-corrales, Mosaic Neurofibromatosis Type 1: A Systematic Review, Pediatr Dermatol. janv, vol.33, issue.1, pp.9-17, 2016.

D. G. Evans, M. P. Adam, H. H. Ardinger, R. A. Pagon, S. E. Wallace et al., Neurofibromatosis, vol.2

(. Seattle and . Wa, , 1993.

D. G. Evans, C. L. Hartley, P. T. Smith, A. T. King, N. L. Bowers et al., Incidence of mosaicism in 1055 de novo NF2 cases: much higher than previous estimates with high utility of next-generation sequencing, Genet Med, 2019.

S. Amitrano, A. Marozza, S. Somma, V. Imperatore, T. Hadjistilianou et al., Next generation sequencing in sporadic retinoblastoma patients reveals somatic mosaicism, Eur J Hum Genet, vol.23, issue.11, pp.1523-1553, 2015.

L. Golmard, C. Delnatte, A. Laugé, V. Moncoutier, C. Lefol et al., Breast and ovarian cancer predisposition due to de novo BRCA1 and BRCA2 mutations, Oncogene. mars, vol.35, issue.10, pp.1324-1331, 2016.

J. A. Veltman and H. G. Brunner, De novo mutations in human genetic disease, Nat Rev Genet. août, vol.13, issue.8, pp.565-75, 2012.

R. Moslehi, R. Singh, L. Lessner, and J. M. Friedman, Impact of BRCA mutations on female fertility and offspring sex ratio, Am J Hum Biol, 2009.

F. Kwiatkowski, M. Arbre, Y. Bidet, C. Laquet, N. Uhrhammer et al., BRCA Mutations Increase Fertility in Families at Hereditary Breast/Ovarian Cancer Risk. Fei P, éditeur, PLOS ONE. 5 juin, vol.10, issue.6, p.127363, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01647108

E. Friedman, N. Efrat, L. Soussan-gutman, A. Dvir, Y. Kaplan et al., Low-level constitutional mosaicism of a de novoBRCA1 gene mutation, Br J Cancer. févr, vol.112, issue.4, pp.765-773, 2015.

I. Delon, A. Taylor, A. Molenda, J. Drummond, K. Oakhill et al., A germline mosaic BRCA1 exon deletion in a woman with bilateral basal-like breast cancer: Letter to the Editor, Clin Genet. sept, vol.84, issue.3, pp.297-306, 2013.

J. Mouchawar, C. Korch, T. Byers, T. M. Pitts, E. Li et al., Population-Based Estimate of the Contribution of TP53 Mutations to Subgroups of Early-Onset Breast Cancer: Australian Breast Cancer Family Study: Table 1, Cancer Res. 15 juin, vol.70, issue.12, pp.4795-800, 2010.

D. Evans, A. Moran, R. Hartley, J. Dawson, B. Bulman et al., Long-term outcomes of breast cancer in women aged 30 years or younger, based on family history, pathology and BRCA1/BRCA2/TP53 status, Br J Cancer. mars, vol.102, issue.7, pp.1091-1099, 2010.

K. D. Gonzalez, C. H. Buzin, K. A. Noltner, D. Gu, W. Li et al., High frequency of de novo mutations in Li-Fraumeni syndrome, J Med Genet, vol.46, issue.10, pp.689-93, 2009.

M. Renaux-petel, F. Charbonnier, J. Théry, P. Fermey, G. Lienard et al., Contribution of de novo and mosaic TP53 mutations to Li-Fraumeni syndrome, J Med Genet, pp.2017-104976, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02355680

K. Prochazkova, K. Pavlikova, M. Minarik, D. Sumerauer, R. Kodet et al., Somatic TP53 mutation mosaicism in a patient with Li-Fraumeni syndrome, Am J Med Genet A. févr, vol.149, issue.2, pp.206-217, 2009.

S. Behjati, M. Maschietto, R. D. Williams, L. Side, M. Hubank et al., A Pathogenic Mosaic TP53 Mutation in Two Germ Layers Detected by Next Generation Sequencing. Mazoyer S, éditeur, PLoS ONE. 8 mai, vol.9, issue.5, p.96531, 2014.

J. Mester and C. Eng, Estimate of de novo mutation frequency in probands with PTEN hamartoma tumor syndrome, Genet Med. sept, vol.14, issue.9, pp.819-841, 2012.

J. Mester and C. Eng, When Overgrowth Bumps Into Cancer: The PTEN-Opathies: AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS), Am J Med Genet C Semin Med Genet. mai, vol.163, issue.2, pp.114-135, 2013.

C. C. Pritchard, C. Smith, T. Marushchak, K. Koehler, H. Holmes et al., A mosaic PTEN mutation causing Cowden syndrome identified by deep sequencing, Genet Med. déc, vol.15, issue.12, pp.1004-1011, 2013.

E. E. Salo-mullen, J. Shia, I. Brownell, P. Allen, M. Girotra et al., Mosaic partial deletion of the PTEN gene in a patient with Cowden syndrome, Fam Cancer. sept, vol.13, issue.3, pp.459-67, 2014.

A. Gammon, K. Jasperson, R. Pilarski, T. Prior, and S. Kuwada, PTEN mosaicism with features of Cowden syndrome: PTEN mosaicism with features of CS, Clin Genet. déc, vol.84, issue.6, pp.593-598, 2013.

F. Caux, H. Plauchu, F. Chibon, L. Faivre, O. Fain et al., Segmental overgrowth, lipomatosis, arteriovenous malformation and epidermal nevus (SOLAMEN) syndrome is related to mosaic PTEN nullizygosity, Eur J Hum Genet. juill, vol.15, issue.7, pp.767-73, 2007.

N. Nathan, K. M. Keppler-noreuil, L. G. Biesecker, J. Moss, and T. N. Darling, Mosaic Disorders of the PI3K/PTEN/AKT/TSC/mTORC1 Signaling Pathway, Dermatol Clin. janv, vol.35, issue.1, pp.51-60, 2017.

A. K. Win, M. A. Jenkins, D. D. Buchanan, M. Clendenning, J. P. Young et al., Determining the frequency of de novo germline mutations in DNA mismatch repair genes, J Med Genet. 1 août, vol.48, issue.8, pp.530-534, 2011.

I. Sourrouille, F. Coulet, J. H. Lefevre, C. Colas, M. Eyries et al., Somatic mosaicism and double somatic hits can lead to MSI colorectal tumors, Fam Cancer. mars, vol.12, issue.1, pp.27-33, 2013.

F. K. Ababneh, A. Al-swaid, A. Elhag, T. Youssef, and S. Alsaif, Blepharo-cheilo-dontic (BCD) syndrome: Expanding the phenotype, case report and review of literature, Am J Med Genet A. juin, vol.164, issue.6, pp.1525-1534, 2014.

A. Kievit, F. Tessadori, H. Douben, I. Jordens, M. Maurice et al., Variants in members of the cadherin-catenin complex, CDH1 and CTNND1, cause blepharocheilodontic syndrome, Eur J Hum Genet. févr, vol.26, issue.2, pp.210-219, 2018.

K. Krempely and R. Karam, A novel de novo CDH1 germline variant aids in the classification of carboxy-terminal E-cadherin alterations predicted to escape nonsensemediated mRNA decay. Cold Spring Harb Mol Case Stud, vol.4, 2018.

S. Sugimoto, H. Yamada, M. Takahashi, Y. Morohoshi, N. Yamaguchi et al., Early-onset diffuse gastric cancer associated with a de novo large genomic deletion of CDH1 gene. Gastric Cancer Off J Int Gastric Cancer Assoc Jpn Gastric Cancer Assoc, vol.17, pp.745-754, 2014.

M. A. Shah, E. Salo-mullen, Z. Stadler, J. M. Ruggeri, M. Mirander et al., De novo CDH1 mutation in a family presenting with early-onset diffuse gastric cancer, Clin Genet. sept, vol.82, issue.3, pp.283-290, 2012.

A. Zehir, R. Benayed, R. H. Shah, A. Syed, S. Middha et al., Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients, Nat Med. juin, vol.23, issue.6, pp.703-716, 2017.

S. D. Merajver, T. M. Pham, R. F. Caduff, M. Chen, E. L. Poy et al., Somatic mutations in the BRCA1 gene in sporadic ovarian tumours, Nat Genet. avr, vol.9, issue.4, pp.439-482, 1995.

K. A. Foster, P. Harrington, J. Kerr, P. Russell, R. A. Dicioccio et al., Somatic and germline mutations of the BRCA2 gene in sporadic ovarian cancer, Cancer Res. 15 août, vol.56, issue.16, pp.3622-3627, 1996.

A. Berchuck, K. A. Heron, M. E. Carney, J. M. Lancaster, E. G. Fraser et al., Frequency of germline and somatic BRCA1 mutations in ovarian cancer, Clin Cancer Res Off J Am Assoc Cancer Res. oct, vol.4, issue.10, pp.2433-2440, 1998.

B. Hennessy, K. M. Timms, M. S. Carey, A. Gutin, L. A. Meyer et al., Somatic Mutations in BRCA1 and BRCA2 Could Expand the Number of Patients That Benefit From Poly (ADP Ribose) Polymerase Inhibitors in Ovarian Cancer, J Clin Oncol. août, vol.28, issue.22, pp.3570-3576, 2010.

B. A. Dougherty, Z. Lai, D. R. Hodgson, M. Orr, M. Hawryluk et al., Biological and clinical evidence for somatic mutations in BRCA1 and BRCA2 as predictive markers for olaparib response in high-grade serous ovarian cancers in the maintenance setting, Oncotarget, vol.8, issue.27, 2017.

, The Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours, Nature, vol.490, issue.7418, pp.61-70, 2012.

P. Sharma, J. R. Klemp, B. F. Kimler, J. D. Mahnken, L. J. Geier et al., Germline BRCA mutation evaluation in a prospective triple-negative breast cancer registry: implications for hereditary breast and/or ovarian cancer syndrome testing, Breast Cancer Res Treat. juin, vol.145, issue.3, pp.707-721, 2014.

C. Kandoth, M. D. Mclellan, F. Vandin, K. Ye, B. Niu et al., Mutational landscape and significance across 12 major cancer types, Nature, vol.502, issue.7471, pp.333-342, 2013.

M. Mansha, A. Gill, and P. C. Thomson, Potential risk factors of ovarian cancer and analysis of CA125, a biomarker used for its monitoring and diagnosis, Mol Biol Rep. juin, vol.46, issue.3, pp.3325-3357, 2019.

K. A. Sikaris, CA125-A Test with a Change of Heart. Heart Lung Circ, vol.20, pp.634-674, 2011.

B. Vogelstein, N. Papadopoulos, V. E. Velculescu, S. Zhou, L. A. Diaz et al., Cancer Genome Landscapes. Science. 29 mars, vol.339, issue.6127, pp.1546-58, 2013.

T. R. Ashworth, 1869) A Case of Cancer in Which Cells Similar to Those in the Tumours Were Seen in the Blood after Death, The Medical Journal of Australia, vol.14, pp.146-147

E. Racila, D. Euhus, A. J. Weiss, C. Rao, J. Mcconnell et al., Detection and characterization of carcinoma cells in the blood, Proc Natl Acad Sci. 14 avr, vol.95, issue.8, pp.4589-94, 1998.

M. Stroun, J. Lyautey, C. Lederrey, A. Olson-sand, and P. Anker, About the possible origin and mechanism of circulating DNA apoptosis and active DNA release, Clin Chim Acta Int J Clin Chem, vol.313, issue.1-2, pp.139-181, 2001.

S. Jahr, H. Hentze, S. Englisch, D. Hardt, F. O. Fackelmayer et al., DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells, Cancer Res. 15 févr, vol.61, issue.4, pp.1659-65, 2001.

S. A. Leon, B. Shapiro, D. M. Sklaroff, M. J. Yaros, and D. , Free DNA in the Serum of Cancer Patients and the Effect of Therapy, 1977.

A. B. , Liquid biopsy identifies residual breast cancer

D. Sur,

Y. Tong, S. Shen, H. Jiang, and Z. Chen, Application of Digital PCR in Detecting Human Diseases Associated Gene Mutation, Cell Physiol Biochem, vol.43, issue.4, pp.1718-1748, 2017.

S. Dawson, D. Tsui, M. Murtaza, H. Biggs, O. M. Rueda et al., Analysis of Circulating Tumor DNA to Monitor Metastatic Breast Cancer, N Engl J Med. 28 mars, vol.368, issue.13, pp.1199-209, 2013.

I. B. Hench, J. Hench, and M. Tolnay, Liquid Biopsy in Clinical Management of Breast, Lung, and Colorectal Cancer, Front Med, vol.5, p.9, 2018.

C. Bettegowda, M. Sausen, R. J. Leary, I. Kinde, Y. Wang et al., Detection of Circulating Tumor DNA in Early-and Late-Stage Human Malignancies, Sci Transl Med. 19 févr, vol.6, issue.224, pp.224-248, 2014.

X. Zhang, S. Ju, X. Wang, and H. Cong, Advances in liquid biopsy using circulating tumor cells and circulating cell-free tumor DNA for detection and monitoring of breast cancer, Clin Exp Med. 1 août, vol.19, issue.3, pp.271-280, 2019.

F. Rothé, M. J. Silva, D. Venet, C. Campbell, I. Bradburry et al., Circulating Tumor DNA in HER2-Amplified Breast Cancer: A Translational Research Substudy of the NeoALTTO Phase III Trial, Clin Cancer Res. 15 juin, vol.25, issue.12, pp.3581-3589, 2019.

G. Buono, L. Gerratana, M. Bulfoni, N. Provinciali, D. Basile et al., Circulating tumor DNA analysis in breast cancer: Is it ready for prime-time?, Cancer Treat Rev. févr, vol.73, pp.73-83, 2019.

M. Ratajska, M. Koczkowska, M. ?uk, A. Gorczy?ski, A. Ku?niacka et al., Detection of BRCA1/2 mutations in circulating tumor DNA from patients with ovarian cancer, Oncotarget [Internet, vol.8, issue.60, 2017.

D. P. Steensma, R. Bejar, S. Jaiswal, R. C. Lindsley, M. A. Sekeres et al., Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes, Blood. 2 juill, vol.126, issue.1, pp.9-16, 2015.

C. J. Gibson and D. P. Steensma, New Insights from Studies of Clonal Hematopoiesis, Clin Cancer Res. 1 oct, vol.24, pp.4633-4675, 2018.

S. Jaiswal, P. Fontanillas, J. Flannick, A. Manning, P. V. Grauman et al., Age-Related Clonal Hematopoiesis Associated with Adverse Outcomes, N Engl J Med. 25 déc, vol.371, issue.26, pp.2488-98, 2014.

G. Genovese, A. K. Kähler, R. E. Handsaker, J. Lindberg, S. A. Rose et al., Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence, N Engl J Med. 25 déc, vol.371, issue.26, pp.2477-87, 2014.

M. Xie, C. Lu, J. Wang, M. D. Mclellan, K. J. Johnson et al., Age-related mutations associated with clonal hematopoietic expansion and malignancies, Nat Med. déc, vol.20, issue.12, pp.1472-1480, 2014.

D. W. Craig, J. V. Pearson, S. Szelinger, A. Sekar, M. Redman et al., Identification of genetic variants using bar-coded multiplexed sequencing, Nat Methods. oct, vol.5, issue.10, pp.887-93, 2008.

H. Li and R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinformatics. 15 juill, vol.25, issue.14, pp.1754-60, 2009.

A. Mckenna, M. Hanna, E. Banks, A. Sivachenko, K. Cibulskis et al., The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data, Genome Res. 1 sept, vol.20, issue.9, pp.1297-303, 2010.

E. Mathe, M. Olivier, S. Kato, C. Ishioka, P. Hainaut et al., Computational approaches for predicting the biological effect of p53 missense mutations: a comparison of three sequence analysis based methods, Nucleic Acids Res, vol.34, issue.5, pp.1317-1342, 2006.

S. V. Tavtigian, Comprehensive statistical study of 452 BRCA1 missense substitutions with classification of eight recurrent substitutions as neutral, J Med Genet. 9 sept, vol.43, issue.4, pp.295-305, 2005.

B. Ewing and P. Green, Base-Calling of Automated Sequencer Traces Using Phred. II. Error Probabilities, Genome Res. 1 mars, vol.8, issue.3, pp.186-94, 1998.

B. Ewing, L. Hillier, M. C. Wendl, and P. Green, Base-Calling of Automated Sequencer Traces Using Phred. I. Accuracy Assessment, Genome Res. 1 mars, vol.8, issue.3, pp.175-85, 1998.

P. Rentzsch, D. Witten, G. M. Cooper, J. Shendure, and M. Kircher, CADD: predicting the deleteriousness of variants throughout the human genome, Nucleic Acids Res. 8 janv, vol.47, issue.D1, pp.886-94, 2019.

L. Raphaël, +. Krieger, +. P. Gaildrat, L. Gac, and G. , SILICO/IN VITRO DE 395 VARIANTS Claude Houdayer 5+# des groupes UGG + et ENIGMA # Introduction, 2017.

Q. Li and W. K. Intervar, Clinical Interpretation of Genetic Variants by the 2015 ACMG-AMP Guidelines, Am J Hum Genet. févr, vol.100, issue.2, pp.267-80, 2017.

L. Bouaoun, D. Sonkin, M. Ardin, M. Hollstein, G. Byrnes et al., TP53 Variations in Human Cancers: New Lessons from the IARC TP53 Database and Genomics Data: Human Mutation, Hum Mutat. sept, vol.37, issue.9, pp.865-76, 2016.

C. C. Coombs, N. K. Gillis, X. Tan, J. S. Berg, M. Ball et al., Identification of Clonal Hematopoiesis Mutations in Solid Tumor Patients Undergoing Unpaired Next-Generation Sequencing Assays, Clin Cancer Res, vol.24, issue.23, pp.5918-5942, 2018.

Y. Hu, B. C. Ulrich, J. Supplee, Y. Kuang, P. H. Lizotte et al., False-Positive Plasma Genotyping Due to Clonal Hematopoiesis, Clin Cancer Res. 15 sept, vol.24, issue.18, pp.4437-4480, 2018.

F. J. Couch, H. Shimelis, C. Hu, S. N. Hart, E. C. Polley et al., Associations Between Cancer Predisposition Testing Panel Genes and Breast Cancer, JAMA Oncol. 1 sept, vol.3, issue.9, p.1190, 2017.

K. Packwood, G. Martland, M. Sommerlad, E. Shaw, K. Moutasim et al., Breast cancer in patients with germline TP53 pathogenic variants have typical tumour characteristics: the Cohort study of TP53 carrier early onset breast cancer (COPE study), J Pathol Clin Res. 1 juill, vol.5, issue.3, pp.189-98, 2019.

H. Schwarzenbach, D. Hoon, and K. Pantel, Cell-free nucleic acids as biomarkers in cancer patients, Nat Rev Cancer. juin, vol.11, issue.6, pp.426-463, 2011.

T. Smith, A. Heger, and I. Sudbery, UMI-tools: modeling sequencing errors in Unique Molecular Identifiers to improve quantification accuracy, Genome Res. mars, vol.27, issue.3, pp.491-500, 2017.

D. Castillo, J. Herzog, S. Sand, T. O'connor, C. Clark et al., Well-groomed participants: eyebrow plucks as surrogates for biomarker samples and a viable source of constitutional DNA, Annual Meeting of the American Society of Human Genetics, pp.18-22, 2016.

J. N. Weitzel, E. C. Chao, B. Nehoray, L. R. Van-tongeren, H. Laduca et al., Somatic TP53 variants frequently confound germ-line testing results, Genet Med. août, vol.20, issue.8, pp.809-825, 2018.

C. C. Coombs, A. Zehir, S. M. Devlin, A. Kishtagari, A. Syed et al., Therapy-Related Clonal Hematopoiesis in Patients with Non-hematologic Cancers Is Common and Associated with Adverse Clinical Outcomes, Cell Stem Cell. sept, vol.21, issue.3, pp.374-382, 2017.

K. Takahashi, F. Wang, H. Kantarjian, D. Doss, K. Khanna et al., Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: a case-control study, Lancet Oncol. janv, vol.18, issue.1, pp.100-111, 2017.

N. K. Gillis, M. Ball, Q. Zhang, Z. Ma, Y. Zhao et al., Clonal haemopoiesis and therapy-related myeloid malignancies in elderly patients: a proof-of-concept, casecontrol study, Lancet Oncol. janv, vol.18, issue.1, pp.112-133, 2017.

E. M. Swisher, M. I. Harrell, B. M. Norquist, T. Walsh, M. Brady et al., Somatic Mosaic Mutations in PPM1D and TP53 in the Blood of Women With Ovarian Carcinoma, JAMA Oncol. 1 mars, vol.2, issue.3, p.370, 2016.

J. I. Hsu, T. Dayaram, A. Tovy, D. Braekeleer, E. Jeong et al., PPM1D Mutations Drive Clonal Hematopoiesis in Response to Cytotoxic Chemotherapy, Cell Stem Cell. nov, vol.23, issue.5, pp.700-713, 2018.

T. N. Wong, G. Ramsingh, A. L. Young, C. A. Miller, W. Touma et al., Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia, Nature. févr, vol.518, issue.7540, pp.552-557, 2015.

E. Ruark, K. Snape, P. Humburg, C. Loveday, I. Bajrami et al., Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer, Nature. 16 déc, vol.493, issue.7432, pp.406-416, 2012.

S. Abelson, G. Collord, S. Ng, O. Weissbrod, M. Cohen et al., Prediction of acute myeloid leukaemia risk in healthy individuals, Nature. juill, vol.559, issue.7714, pp.400-404, 2018.

P. Desai, N. Mencia-trinchant, O. Savenkov, M. S. Simon, G. Cheang et al., Somatic mutations precede acute myeloid leukemia years before diagnosis, Nat Med. 1 juill, vol.24, issue.7, pp.1015-1038, 2018.

P. Desai and G. J. Roboz, Clonal Hematopoiesis and therapy related MDS/AML, Best Pract Res Clin Haematol. mars, vol.32, issue.1, pp.13-23, 2019.