. Surveillance, E. Epidemiology, and . Program, URL https://seer.cancer.gov/ [accessed on 31, 2017.

P. Austin, C. Cruse, and G. Lyman, Age as a prognostic factor in the malignant melanoma population, Annals of Surgical Oncology, vol.218, issue.6, pp.487-94, 1994.
DOI : 10.1093/ajcp/74.3.247

C. Chao, R. Martin, and M. Ross, Correlation Between Prognostic Factors and Increasing Age in Melanoma, Annals of Surgical Oncology, vol.86, issue.(9 Suppl), pp.259-64, 2004.
DOI : 10.1245/ASO.2004.04.015

P. Chapman, A. Hauschild, and C. Robert, Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation, New England Journal of Medicine, vol.364, issue.26, pp.2507-2523, 2011.
DOI : 10.1056/NEJMoa1103782

G. Long, D. Stroyakovskiy, and H. Gogas, Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial, The Lancet, vol.386, issue.9992, pp.444-51, 2015.
DOI : 10.1016/S0140-6736(15)60898-4

C. Balch, J. Gershenwald, and S. Soong, Final Version of 2009 AJCC Melanoma Staging and Classification, Journal of Clinical Oncology, vol.27, issue.36, pp.6199-206, 2009.
DOI : 10.1200/JCO.2009.23.4799

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2793035/pdf

M. Rousset, K. Titier, and S. Bouchet, An UPLC-MS/MS method for the quantification of BRAF inhibitors (vemurafenib, dabrafenib) and MEK inhibitors (cobimetinib, trametinib, binimetinib) in human plasma. Application to treated melanoma patients, Clinica Chimica Acta, vol.470, pp.8-13, 2017.
DOI : 10.1016/j.cca.2017.04.009

M. Rousset, C. Dutriaux, and P. Bosco-lévy, Trough dabrafenib plasma concentrations can predict occurrence of adverse events requiring dose reduction in metastatic melanoma, Clinica Chimica Acta, vol.472
DOI : 10.1016/j.cca.2017.07.012

J. Larkin, P. Ascierto, and B. Dréno, -Mutated Melanoma, New England Journal of Medicine, vol.371, issue.20, pp.1867-76, 2014.
DOI : 10.1056/NEJMoa1408868

G. Falchook, G. Long, and R. Kurzrock, Dose Selection, Pharmacokinetics, and Pharmacodynamics of BRAF Inhibitor Dabrafenib (GSK2118436), Clinical Cancer Research, vol.20, issue.17, pp.4449-58, 2014.
DOI : 10.1158/1078-0432.CCR-14-0887

URL : http://clincancerres.aacrjournals.org/content/clincanres/20/17/4449.full.pdf

D. Ouellet, E. Gibiansky, and C. Leonowens, Population pharmacokinetics of dabrafenib, a BRAF inhibitor: Effect of dose, time, covariates, and relationship with its metabolites, The Journal of Clinical Pharmacology, vol.12, issue.Suppl. 1, pp.696-706, 2014.
DOI : 10.2174/138920011796504527

D. Ouellet, N. Kassir, and J. Chiu, Population pharmacokinetics and exposure???response of trametinib, a MEK inhibitor, in patients with BRAF V600 mutation-positive melanoma, Cancer Chemotherapy and Pharmacology, vol.45, issue.2
DOI : 10.1016/j.ejca.2008.10.026

G. Mcarthur, P. Chapman, and C. Robert, Safety and efficacy of vemurafenib in BRAFV600E and BRAFV600K mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study, The Lancet Oncology, vol.15, issue.3, pp.323-355, 2014.
DOI : 10.1016/S1470-2045(14)70012-9

P. Ascierto, D. Minor, and A. Ribas, Phase II Trial (BREAK-2) of the BRAF Inhibitor Dabrafenib (GSK2118436) in Patients With Metastatic Melanoma, Journal of Clinical Oncology, vol.31, issue.26, pp.3205-3216, 2013.
DOI : 10.1200/JCO.2013.49.8691

C. Lee, A. Menzies, and L. Haydu, Features and management of pyrexia with combined dabrafenib and trametinib in metastatic melanoma, Melanoma Research, vol.24, issue.5, pp.468-74, 2014.
DOI : 10.1097/CMR.0000000000000110

J. Larkin, D. Vecchio, M. Ascierto, and P. , Vemurafenib in patients with BRAFV600 mutated metastatic melanoma: an open-label, multicentre, safety study, The Lancet Oncology, vol.15, issue.4, pp.436-480, 2014.
DOI : 10.1016/S1470-2045(14)70051-8

C. Robert, B. Karaszewska, and J. Schachter, Improved Overall Survival in Melanoma with Combined Dabrafenib and Trametinib, New England Journal of Medicine, vol.372, issue.1, pp.30-39, 2015.
DOI : 10.1056/NEJMoa1412690

R. Anforth, T. Blumetti, and R. Kefford, Cutaneous manifestations of dabrafenib (GSK2118436): a selective inhibitor of mutant BRAF in patients with metastatic melanoma, British Journal of Dermatology, vol.230, issue.Suppl., pp.1153-60, 2012.
DOI : 10.1006/bbrc.1996.5891

V. Belum, A. Fischer, J. Choi, and M. Lacouture, Dermatological Adverse Events from BRAF Inhibitors: A Growing Problem, Current Oncology Reports, vol.365, issue.15, pp.249-59, 2013.
DOI : 10.1074/jbc.273.34.21730

J. Curry, C. Torres-cabala, and K. Kim, Dermatologic toxicities to targeted cancer therapy: shared clinical and histologic adverse skin reactions, International Journal of Dermatology, vol.163, issue.3, pp.376-84, 2014.
DOI : 10.1111/j.1365-2133.2010.09751.x

S. Heidorn, C. Milagre, and S. Whittaker, Kinase-Dead BRAF and Oncogenic RAS Cooperate to Drive Tumor Progression through CRAF, Cell, vol.140, issue.2, pp.209-230, 2010.
DOI : 10.1016/j.cell.2009.12.040

URL : https://doi.org/10.1016/j.cell.2009.12.040

G. Hatzivassiliou, K. Song, and I. Yen, RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth, Nature, vol.50, issue.7287, pp.431-436, 2010.
DOI : 10.1128/MCB.14.12.8212

C. Hughes, Medication Non-Adherence in the Elderly, Drugs & Aging, vol.15, issue.12, pp.793-811, 2004.
DOI : 10.2165/00002512-200421120-00004

N. Campbell, M. Boustani, and E. Skopelja, Medication Adherence in Older Adults With Cognitive Impairment: A Systematic Evidence-Based Review, The American Journal of Geriatric Pharmacotherapy, vol.10, issue.3, pp.165-77, 2012.
DOI : 10.1016/j.amjopharm.2012.04.004

M. Sharma, K. Loh, and G. Nightingale, Polypharmacy and potentially inappropriate medication use in geriatric oncology, Journal of Geriatric Oncology, vol.7, issue.5, pp.346-53, 2016.
DOI : 10.1016/j.jgo.2016.07.010

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5037024/pdf

J. Greer, N. Amoyal, and L. Nisotel, A Systematic Review of Adherence to Oral Antineoplastic Therapies, The Oncologist, vol.21, issue.3, pp.354-76, 2016.
DOI : 10.1634/theoncologist.2015-0405

P. Kardas, P. Lewek, and M. Matyjaszczyk, Determinants of patient adherence: a review of systematic reviews, Frontiers in Pharmacology, vol.4, p.91, 2013.
DOI : 10.3389/fphar.2013.00091

J. Grob, M. Amonkar, and S. Martin-algarra, Patient perception of the benefit of a BRAF inhibitor in metastatic melanoma: quality-of-life analyses of the BREAK-3 study comparing dabrafenib with dacarbazine, Annals of Oncology, vol.25, issue.7, pp.1428-1464, 2014.
DOI : 10.1093/annonc/mdu154

. Les-cancers-en-france-en, Boulogne-Billancourt: Collec-tion état des lieux et des connaissances, ouvrage collectif éditépar l'INCa, 2013.

F. Binder-foucard, N. Bossard, and P. Delafosse, Cancer incidence and mortality in France over the 1980???2012 period: Solid tumors, Revue d'??pid??miologie et de Sant?? Publique, vol.62, issue.2, pp.95-108, 2014.
DOI : 10.1016/j.respe.2013.11.073

S. Gandini, F. Sera, and M. Cattaruzza, Meta-analysis of risk factors for cutaneous melanoma: II. Sun exposure, European Journal of Cancer, vol.41, issue.1, pp.45-60, 1990.
DOI : 10.1016/j.ejca.2004.10.016

T. Fears, J. Scotto, and M. Schneiderman, MATHEMATICAL MODELS OF AGE AND ULTRAVIOLET EFFECTS ON THE INCIDENCE OF SKIN CANCER AMONG WHITES IN THE UNITED STATES, American Journal of Epidemiology, vol.105, issue.5, pp.420-427, 1977.
DOI : 10.1093/oxfordjournals.aje.a112400

J. Elwood and J. Jopson, Melanoma and sun exposure: An overview of published studies, International Journal of Cancer, vol.52, issue.2, pp.198-203, 1997.
DOI : 10.1002/ijc.2910520128

URL : http://onlinelibrary.wiley.com/doi/10.1002/(SICI)1097-0215(19971009)73:2<198::AID-IJC6>3.0.CO;2-R/pdf

D. Whiteman, W. Pavan, and B. Bastian, The melanomas: a synthesis of epidemiological, clinical, histopathological, genetic, and biological aspects, supporting distinct subtypes, causal pathways, and cells of origin, Pigment Cell & Melanoma Research, vol.65, issue.10, pp.879-97, 2011.
DOI : 10.1086/302494

F. Grange, L. Mortier, and A. Crine, Prevalence of sunbed use, and characteristics and knowledge of sunbed users: results from the French population-based Edifice Melanoma survey, Journal of the European Academy of Dermatology and Venereology, vol.28, issue.Suppl 3, pp.23-30, 2015.
DOI : 10.1177/109019810102800304

M. Greene, The genetics of hereditary melanoma and nevi, Cancer, vol.14, issue.S11, pp.2464-77, 1999.
DOI : 10.1200/JCO.1996.14.5.1730

A. Kamb, D. Shattuck-eidens, and R. Eeles, Analysis of the p16 gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus, Nature Genetics, vol.52, issue.1, pp.23-29, 1994.
DOI : 10.1001/jama.269.5.558

E. Holland, H. Schmid, R. Kefford, and G. Mann, CDKN2A (P16INK4a) andCDK4 mutation analysis in 131 Australian melanoma probands: Effect of family history and multiple primary melanomas, Genes, Chromosomes and Cancer, vol.12, issue.4, pp.339-387, 1999.
DOI : 10.1038/bjc.1996.460

J. Harbour, M. Onken, and E. Roberson, Frequent Mutation of BAP1 in Metastasizing Uveal Melanomas, Science, vol.35, issue.suppl_1, pp.1410-1413, 2010.
DOI : 10.1093/nar/gkl952

T. Wiesner, A. Obenauf, and R. Murali, Germline mutations in BAP1 predispose to melanocytic tumors, Nature Genetics, vol.43, issue.10, pp.1018-1039, 2011.
DOI : 10.1186/gb-2007-8-11-r232

L. Titus-ernstoff, A. Perry, and S. Spencer, Pigmentary characteristics and moles in relation to melanoma risk, International Journal of Cancer, vol.46, issue.1, pp.144-153, 2005.
DOI : 10.1038/bjc.1996.302

URL : http://onlinelibrary.wiley.com/doi/10.1002/ijc.21001/pdf

B. Hazen, A. Bhatia, T. Zaim, and R. Brodell, The clinical diagnosis of early malignant melanoma: expansion of the ABCD criteria to improve diagnostic sensitivity, Dermatol Online J, vol.5, p.3, 1999.

M. Vestergaard, P. Macaskill, P. Holt, and S. Menzies, Dermoscopy compared with naked eye examination for the diagnosis of primary melanoma: a meta-analysis of studies performed in a clinical setting, British Journal of Dermatology, vol.141, pp.669-76, 2008.
DOI : 10.1001/archderm.137.10.1343

S. Rajpara, A. Botello, J. Townend, and A. Ormerod, Systematic review of dermoscopy and digital dermoscopy/ artificial intelligence for the diagnosis of melanoma, British Journal of Dermatology, vol.6, issue.Suppl. 1, pp.591-604, 2009.
DOI : 10.7326/0003-4819-120-8-199404150-00008

G. Salerni, T. Terán, and S. Puig, Meta-analysis of digital dermoscopy follow-up of melanocytic skin lesions: a study on behalf of the International Dermoscopy Society, Journal of the European Academy of Dermatology and Venereology, vol.36, issue.7, pp.805-819, 2013.
DOI : 10.1016/S0190-9622(97)80324-6

V. Mcgovern, M. Mihm, and C. Bailly, The classification of malignant melanoma and its histologic reporting, Cancer, vol.18, issue.6, pp.1446-57, 1973.
DOI : 10.1016/S0031-3025(16)39458-2

D. Ivan and V. Prieto, Use of immunohistochemistry in the diagnosis of melanocytic lesions: applications and pitfalls, Future Oncology, vol.22, issue.7, pp.1163-75, 2010.
DOI : 10.1038/modpathol.2009.116

M. Rastrelli, S. Tropea, C. Rossi, and M. Alaibac, Melanoma: Epidemiology, Risk Factors, Pathogenesis, Diagnosis and Classification, In Vivo, vol.28, pp.1005-1016, 2014.

H. Higgins, K. Lee, A. Galan, and D. Leffell, Melanoma in situ, Journal of the American Academy of Dermatology, vol.73, issue.2, pp.181-90, 2015.
DOI : 10.1016/j.jaad.2015.04.014

J. Goydos and S. Shoen, Acral Lentiginous Melanoma, Cancer Treat Res, vol.167, pp.321-330, 2016.
DOI : 10.1007/978-3-319-22539-5_14

R. Patrick, N. Fenske, and J. Messina, Primary mucosal melanoma, Journal of the American Academy of Dermatology, vol.56, issue.5, pp.828-862, 2007.
DOI : 10.1016/j.jaad.2006.06.017

L. Chen, N. Jaimes, and C. Barker, Desmoplastic melanoma: A review, Journal of the American Academy of Dermatology, vol.68, issue.5, pp.825-858, 2013.
DOI : 10.1016/j.jaad.2012.10.041

C. Balch, J. Gershenwald, and S. Soong, Final Version of 2009 AJCC Melanoma Staging and Classification, Journal of Clinical Oncology, vol.27, issue.36, pp.6199-206, 2009.
DOI : 10.1200/JCO.2009.23.4799

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2793035/pdf

L. Serrone, M. Zeuli, F. Sega, and F. Cognetti, Dacarbazine-based chemotherapy for metastatic melanoma: thirty-year experience overview, J Exp Clin Cancer Res CR, vol.19, pp.21-34, 2000.

M. Avril, S. Aamdal, and J. Grob, Fotemustine Compared With Dacarbazine in Patients With Disseminated Malignant Melanoma: A Phase III Study, Journal of Clinical Oncology, vol.22, issue.6, pp.1118-1143, 2004.
DOI : 10.1200/JCO.2004.04.165

M. Middleton, J. Grob, and N. Aaronson, Randomized Phase III Study of Temozolomide Versus Dacarbazine in the Treatment of Patients With Advanced Metastatic Malignant Melanoma, Journal of Clinical Oncology, vol.18, issue.1, pp.158-66, 2000.
DOI : 10.1200/JCO.2000.18.1.158

P. Dariavach, M. Mattéi, P. Golstein, and M. Lefranc, Human Ig superfamily CTLA-4 gene: chromosomal localization and identity of protein sequence between murine and human CTLA-4 cytoplasmic domains, European Journal of Immunology, vol.13, issue.12, pp.1901-1906, 1988.
DOI : 10.1002/eji.1830181206

N. Karandikar, C. Vanderlugt, and T. Walunas, CTLA-4: a negative regulator of autoimmune disease, Journal of Experimental Medicine, vol.184, issue.2, pp.783-791, 1996.
DOI : 10.1084/jem.184.2.783

G. Phan, J. Yang, and R. Sherry, Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma, Proceedings of the National Academy of Sciences, vol.163, issue.14, pp.8372-8379, 2003.
DOI : 10.1073/pnas.0830997100

URL : http://www.pnas.org/content/100/14/8372.full.pdf

T. Walunas, C. Bakker, and J. Bluestone, CTLA-4 ligation blocks CD28-dependent T cell activation [published erratum appears in J Exp Med 1996 Jul 1;184(1):301], Journal of Experimental Medicine, vol.183, issue.6, pp.2541-50, 1996.
DOI : 10.1084/jem.183.6.2541

URL : http://jem.rupress.org/content/jem/183/6/2541.full.pdf

C. Kyi and M. Postow, Checkpoint blocking antibodies in cancer immunotherapy, FEBS Letters, vol.31, issue.2, pp.368-76, 2014.
DOI : 10.1056/NEJMe1205943

URL : http://onlinelibrary.wiley.com/doi/10.1016/j.febslet.2013.10.015/pdf

D. Mcdermott and M. Atkins, PD-1 as a potential target in cancer therapy, Cancer Medicine, vol.16, issue.Suppl., pp.662-73, 2013.
DOI : 10.1016/j.molmed.2010.02.001

R. Wong, R. Scotland, and R. Lau, Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs, International Immunology, vol.19, issue.10, pp.1223-1257, 2007.
DOI : 10.1093/intimm/dxm091

URL : https://academic.oup.com/intimm/article-pdf/19/10/1223/2230147/dxm091.pdf

C. Blank, I. Brown, and A. Peterson, T Cells, Cancer Research, vol.64, issue.3, pp.1140-1145, 2004.
DOI : 10.1158/0008-5472.CAN-03-3259

URL : https://hal.archives-ouvertes.fr/inserm-01408787

B. Almagro, M. Steyls, and N. Navarro, Occurrence of Subacute Cutaneous Lupus Erythematosus After Treatment With Systemic Fluorouracil, Journal of Clinical Oncology, vol.29, issue.20, pp.613-615, 2011.
DOI : 10.1200/JCO.2011.35.0686

D. Leach, M. Krummel, and J. Allison, Enhancement of Antitumor Immunity by CTLA-4 Blockade, Science, vol.271, issue.5256, pp.1734-1740, 1996.
DOI : 10.1126/science.271.5256.1734

C. Robert, L. Thomas, and I. Bondarenko, Ipilimumab plus Dacarbazine for Previously Untreated Metastatic Melanoma, New England Journal of Medicine, vol.364, issue.26, pp.2517-2543, 2011.
DOI : 10.1056/NEJMoa1104621

P. Prieto, J. Yang, and R. Sherry, CTLA-4 Blockade with Ipilimumab: Long-term Follow-up of 177 Patients with Metastatic Melanoma, Clinical Cancer Research, vol.18, issue.7, pp.2039-2086, 2012.
DOI : 10.1158/1078-0432.CCR-11-1823

F. Hodi, O. Day, S. Mcdermott, and D. , Improved Survival with Ipilimumab in Patients with Metastatic Melanoma, New England Journal of Medicine, vol.363, issue.8, pp.711-734, 2010.
DOI : 10.1056/NEJMoa1003466

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3549297/pdf

C. Robert, A. Ribas, and J. Wolchok, Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial, The Lancet, vol.384, issue.9948, pp.1109-1126, 2014.
DOI : 10.1016/S0140-6736(14)60958-2

A. Ribas, O. Hamid, and A. Daud, Association of Pembrolizumab With Tumor Response and Survival Among Patients With Advanced Melanoma, JAMA, vol.315, issue.15, pp.1600-1609, 2016.
DOI : 10.1001/jama.2016.4059

A. Ribas, I. Puzanov, and R. Dummer, Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial, The Lancet Oncology, vol.16, issue.8, pp.908-926, 2015.
DOI : 10.1016/S1470-2045(15)00083-2

J. Weber, G. Gibney, and R. Kudchadkar, Phase I/II Study of Metastatic Melanoma Patients Treated with Nivolumab Who Had Progressed after Ipilimumab, Cancer Immunology Research, vol.4, issue.4, pp.345-53, 2016.
DOI : 10.1158/2326-6066.CIR-15-0193

J. Weber, D. Angelo, S. Minor, and D. , Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial, The Lancet Oncology, vol.16, issue.4, pp.375-84, 2015.
DOI : 10.1016/S1470-2045(15)70076-8

J. Larkin, C. Lao, and W. Urba, Wild-Type Advanced Melanoma, JAMA Oncology, vol.1, issue.4
DOI : 10.1001/jamaoncol.2015.1184

C. Robert, G. Long, and B. Brady, Mutation, New England Journal of Medicine, vol.372, issue.4, pp.320-350, 2015.
DOI : 10.1056/NEJMoa1412082

URL : https://hal.archives-ouvertes.fr/hal-00872683

E. Buchbinder and A. Desai, CTLA-4 and PD-1 Pathways, American Journal of Clinical Oncology, vol.39, issue.1, pp.98-106, 2016.
DOI : 10.1097/COC.0000000000000239

URL : http://doi.org/10.1097/coc.0000000000000239

Z. Abdel-malek, A. Kadekaro, and V. Swope, Stepping up melanocytes to the challenge of UV exposure, Pigment Cell & Melanoma Research, vol.90, issue.7, pp.171-86, 2010.
DOI : 10.1073/pnas.90.9.4216

T. Hirobe, How are proliferation and differentiation of melanocytes regulated?, Pigment Cell & Melanoma Research, vol.33, issue.Suppl. 8, pp.462-78, 2011.
DOI : 10.1038/ng1087

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1755-148X.2011.00845.x/pdf

R. Sullivan and K. Flaherty, MAP kinase signaling and inhibition in melanoma, Oncogene, vol.18, issue.19, pp.2373-2382, 2013.
DOI : 10.1158/1078-0432.CCR-11-2479

D. Matallanas, M. Birtwistle, and D. Romano, Raf Family Kinases: Old Dogs Have Learned New Tricks, Genes & Cancer, vol.2, issue.3, pp.232-60, 2011.
DOI : 10.1177/1947601911407323

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3128629/pdf

R. Buscà, A. P. Mantoux, and F. , Ras mediates the cAMP-dependent activation of extracellular signal-regulated kinases (ERKs) in melanocytes, The EMBO Journal, vol.19, issue.12, pp.2900-2910, 2000.
DOI : 10.1093/emboj/19.12.2900

H. Davies, G. Bignell, and C. Cox, Mutations of the BRAF gene in human cancer, Nature, vol.7, issue.6892, pp.949-54, 2002.
DOI : 10.1016/S0960-9822(97)70094-0

A. Menzies, L. Haydu, and L. Visintin, Distinguishing Clinicopathologic Features of Patients with V600E and V600K BRAF-Mutant Metastatic Melanoma, Clinical Cancer Research, vol.18, issue.12, pp.3242-3251, 2012.
DOI : 10.1158/1078-0432.CCR-12-0052

J. Solus, S. Kraft, and R. Ras, Ras, Raf, and MAP Kinase in Melanoma, Advances In Anatomic Pathology, vol.20, issue.4, pp.217-243, 2013.
DOI : 10.1097/PAP.0b013e3182976c94

G. Bollag, P. Hirth, and J. Tsai, Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma, Nature, vol.26, issue.7315, pp.596-605, 2010.
DOI : 10.1200/JCO.2008.20.3745

K. Flaherty, I. Puzanov, and K. Kim, Inhibition of Mutated, Activated BRAF in Metastatic Melanoma, New England Journal of Medicine, vol.363, issue.9, pp.809-828, 2010.
DOI : 10.1056/NEJMoa1002011

A. Hauschild, J. Grob, and L. Demidov, Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial, The Lancet, vol.380, issue.9839, pp.358-65, 2012.
DOI : 10.1016/S0140-6736(12)60868-X

P. Chapman, A. Hauschild, and C. Robert, Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation, New England Journal of Medicine, vol.364, issue.26, pp.2507-2523, 2011.
DOI : 10.1056/NEJMoa1103782

G. Mcarthur, P. Chapman, and C. Robert, Safety and efficacy of vemurafenib in BRAFV600E and BRAFV600K mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study, The Lancet Oncology, vol.15, issue.3, pp.323-355, 2014.
DOI : 10.1016/S1470-2045(14)70012-9

A. Hauschild, J. Grob, and L. Demidov, An update on BREAK-3, a phase III, randomized trial: Dabrafenib (DAB) versus dacarbazine (DTIC) in patients with BRAF V600E-positive mutation metastatic melanoma (MM), J Clin Oncol, vol.31, pp.9013-9013, 2013.

J. Grippo, W. Zhang, and D. Heinzmann, A phase I, randomized, open-label study of the multiple-dose pharmacokinetics of vemurafenib in patients with BRAF V600E mutation-positive metastatic melanoma, Cancer Chemotherapy and Pharmacology, vol.26, issue.1, pp.103-114, 2014.
DOI : 10.1016/j.ejps.2005.05.010

J. Sosman, K. Kim, and L. Schuchter, Survival in BRAF V600???Mutant Advanced Melanoma Treated with Vemurafenib, New England Journal of Medicine, vol.366, issue.8, pp.707-721, 2012.
DOI : 10.1056/NEJMoa1112302

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3724515/pdf

W. Zhang, D. Heinzmann, and J. Grippo, Clinical Pharmacokinetics of Vemurafenib, Clinical Pharmacokinetics, vol.11, issue.1, pp.1033-1076, 2017.
DOI : 10.1007/s11523-015-0375-8

G. Falchook, G. Long, and R. Kurzrock, Dose Selection, Pharmacokinetics, and Pharmacodynamics of BRAF Inhibitor Dabrafenib (GSK2118436), Clinical Cancer Research, vol.20, issue.17, pp.4449-58, 2014.
DOI : 10.1158/1078-0432.CCR-14-0887

URL : http://clincancerres.aacrjournals.org/content/clincanres/20/17/4449.full.pdf

S. Goldinger, J. Rinderknecht, and R. Dummer, A single-dose mass balance and metabolite-profiling study of vemurafenib in patients with metastatic melanoma, Pharmacology Research & Perspectives, vol.30, issue.Suppl., p.113, 2015.
DOI : 10.1200/JCO.2011.41.1660

D. Ouellet, N. Kassir, and J. Chiu, Population pharmacokinetics and exposure???response of trametinib, a MEK inhibitor, in patients with BRAF V600 mutation-positive melanoma, Cancer Chemotherapy and Pharmacology, vol.45, issue.2, pp.807-824, 2016.
DOI : 10.1016/j.ejca.2008.10.026

G. Bollag, J. Tsai, and J. Zhang, Vemurafenib: the first drug approved for BRAF-mutant cancer, Nature Reviews Drug Discovery, vol.61, issue.11, pp.873-86, 2012.
DOI : 10.1038/383181a0

D. Bershas, D. Ouellet, and D. Mamaril-fishman, Metabolism and Disposition of Oral Dabrafenib in Cancer Patients: Proposed Participation of Aryl Nitrogen in Carbon-Carbon Bond Cleavage via Decarboxylation following Enzymatic Oxidation, Drug Metabolism and Disposition, vol.41, issue.12, pp.2215-2239, 2013.
DOI : 10.1124/dmd.113.053785

M. Ho, M. Morris, and J. Pirhalla, Trametinib, a first-in-class oral MEK inhibitor mass balance study with limited enrollment of two male subjects with advanced cancers, Xenobiotica, vol.66, issue.4, pp.352-68, 2014.
DOI : 10.1128/AEM.66.6.2336-2342.2000

J. Park, A. Boddy, and X. Liu, Pharmacokinetics of dabrafenib in a patient with metastatic melanoma undergoing haemodialysis, Pigment Cell & Melanoma Research, vol.372, issue.1, pp.68-71, 2017.
DOI : 10.1056/NEJMoa1412690

P. Ascierto, D. Minor, and A. Ribas, Phase II Trial (BREAK-2) of the BRAF Inhibitor Dabrafenib (GSK2118436) in Patients With Metastatic Melanoma, Journal of Clinical Oncology, vol.31, issue.26, pp.3205-3216, 2013.
DOI : 10.1200/JCO.2013.49.8691

R. Anforth, T. Blumetti, and R. Kefford, Cutaneous manifestations of dabrafenib (GSK2118436): a selective inhibitor of mutant BRAF in patients with metastatic melanoma, British Journal of Dermatology, vol.230, issue.Suppl., pp.1153-60, 2012.
DOI : 10.1006/bbrc.1996.5891

J. Curry, C. Torres-cabala, and K. Kim, Dermatologic toxicities to targeted cancer therapy: shared clinical and histologic adverse skin reactions, International Journal of Dermatology, vol.163, issue.3, pp.376-84, 2014.
DOI : 10.1111/j.1365-2133.2010.09751.x

K. Young, A. Minchom, and J. Larkin, mutation, Future Oncology, vol.29, issue.5, pp.499-507, 2012.
DOI : 10.1056/NEJMoa1105358

M. Lacouture, M. Duvic, and A. Hauschild, Analysis of Dermatologic Events in Vemurafenib-Treated Patients With Melanoma, The Oncologist, vol.18, issue.3, pp.314-336, 2013.
DOI : 10.1634/theoncologist.2012-0333

J. Larkin, D. Vecchio, M. Ascierto, and P. , Vemurafenib in patients with BRAFV600 mutated metastatic melanoma: an open-label, multicentre, safety study, The Lancet Oncology, vol.15, issue.4, pp.436-480, 2014.
DOI : 10.1016/S1470-2045(14)70051-8

L. Peuvrel, G. Quéreux, and M. Saint-jean, Profile of vemurafenib-induced severe skin toxicities, Journal of the European Academy of Dermatology and Venereology, vol.115, issue.2, pp.250-257, 2016.
DOI : 10.1002/cncr.24088

M. Wantz, I. Spanoudi-kitrimi, and A. Lasek, N??crolyse ??pidermique toxique au vemurafenib, Annales de Dermatologie et de V??n??r??ologie, vol.141, issue.3, pp.215-223, 2014.
DOI : 10.1016/j.annder.2013.10.054

A. Teixeira, P. Morlière, and J. Ferreira, Interplay Between Membrane Lipid Peroxidation and Photoproduct Formation in the Ultraviolet A-Induced Phototoxicity of Vemurafenib in Skin Keratinocytes, Toxicological Sciences, vol.154, issue.2, pp.289-95, 2016.
DOI : 10.1093/toxsci/kfw159

R. Gabeff, H. Dutartre, and A. Khammari, Phototoxicity of B-RAF inhibitors: Exclusively due to UVA radiation and rapidly regressive, Eur J Dermatol EJD, vol.25, pp.452-458, 2015.

L. Boussemart, E. Routier, and C. Mateus, Prospective study of cutaneous side-effects associated with the BRAF inhibitor vemurafenib: a study of 42 patients, Annals of Oncology, vol.24, issue.6, pp.1691-1698, 2013.
DOI : 10.1093/annonc/mdt015

R. Anforth, C. G. Clements, and A. , Cutaneous adverse events in patients treated with BRAF inhibitor-based therapies for metastatic melanoma for longer than 52??weeks, British Journal of Dermatology, vol.134, issue.15 Suppl., pp.239-282, 2015.
DOI : 10.1038/jid.2013.462

F. Su, A. Viros, and C. Milagre, Mutations in Cutaneous Squamous-Cell Carcinomas in Patients Treated with BRAF Inhibitors, New England Journal of Medicine, vol.366, issue.3, pp.207-222, 2012.
DOI : 10.1056/NEJMoa1105358

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3724537/pdf

P. Oberholzer, D. Kee, and P. Dziunycz, Mutations Are Associated With the Development of Cutaneous Squamous Cell Tumors in Patients Treated With RAF Inhibitors, Journal of Clinical Oncology, vol.30, issue.3, pp.316-337, 2012.
DOI : 10.1200/JCO.2011.36.7680

S. Heidorn, C. Milagre, and S. Whittaker, Kinase-Dead BRAF and Oncogenic RAS Cooperate to Drive Tumor Progression through CRAF, Cell, vol.140, issue.2, pp.209-230, 2010.
DOI : 10.1016/j.cell.2009.12.040

URL : https://doi.org/10.1016/j.cell.2009.12.040

E. Hodis, I. Watson, and G. Kryukov, A Landscape of Driver Mutations in Melanoma, Cell, vol.150, issue.2, pp.251-63, 2012.
DOI : 10.1016/j.cell.2012.06.024

P. Poulikakos, C. Zhang, and G. Bollag, RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF, Nature, vol.6, issue.7287, pp.427-457, 2010.
DOI : 10.1016/S1359-6349(09)72036-1

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3178447/pdf

R. Nazarian, H. Shi, and Q. Wang, Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation, Nature, vol.66, issue.7326, pp.973-980, 2010.
DOI : 10.1158/0008-5472.CAN-05-4227

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3143360/pdf

J. Villanueva, A. Vultur, and J. Lee, Acquired Resistance to BRAF Inhibitors Mediated by a RAF Kinase Switch in Melanoma Can Be Overcome by Cotargeting MEK and IGF-1R/PI3K, Cancer Cell, vol.18, issue.6, pp.683-95, 2010.
DOI : 10.1016/j.ccr.2010.11.023

D. Thakur, M. Salangsang, F. Landman, and A. , Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance, Nature, vol.7, issue.7436, pp.251-256, 2013.
DOI : 10.1097/JTO.0b013e31823c5aee

H. Shi, G. Moriceau, and X. Kong, Melanoma whole-exome sequencing identifies V600E B- RAF amplification-mediated acquired B-RAF inhibitor resistance, Nat Commun, vol.3, p.1727, 2012.
DOI : 10.1038/ncomms1727

URL : http://www.nature.com/articles/ncomms1727.pdf

C. Johannessen, J. Boehm, and S. Kim, COT drives resistance to RAF inhibition through MAP kinase pathway reactivation, Nature, vol.21, issue.7326, pp.968-72, 2010.
DOI : 10.1038/sj.onc.1205550

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3058384/pdf

P. Poulikakos, Y. Persaud, and M. Janakiraman, RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E), Nature, vol.11, issue.7377, pp.387-90, 2011.
DOI : 10.1002/cyto.990110707

J. Choi, S. Landrette, and T. Wang, Identification of PLX4032-resistance mechanisms and implications for novel RAF inhibitors, Pigment Cell & Melanoma Research, vol.487, issue.2, pp.253-62, 2014.
DOI : 10.1038/nature11249

T. Wagenaar, L. Ma, and B. Roscoe, Resistance to vemurafenib resulting from a novel mutation in the BRAFV600E kinase domain, Pigment Cell & Melanoma Research, vol.2, issue.1, pp.124-157, 2014.
DOI : 10.1126/scitranslmed.3000758

J. Charles, C. Martel, and F. De-fraipont, M??canismes de r??sistance aux inhibiteurs de BRAF, Annales de Dermatologie et de V??n??r??ologie, vol.141, issue.11, pp.671-81, 2014.
DOI : 10.1016/j.annder.2014.06.021

I. Sansal and W. Sellers, Tumor Suppressor Pathway, Journal of Clinical Oncology, vol.22, issue.14, pp.2954-63, 2004.
DOI : 10.1200/JCO.2004.02.141

D. Dankort, D. Curley, and R. Cartlidge, BrafV600E cooperates with Pten loss to induce metastatic melanoma, Nature Genetics, vol.17, issue.5, pp.544-52, 2009.
DOI : 10.1038/75973

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2705918/pdf

K. Paraiso, Y. Xiang, and V. Rebecca, PTEN Loss Confers BRAF Inhibitor Resistance to Melanoma Cells through the Suppression of BIM Expression, Cancer Research, vol.71, issue.7, pp.2750-60, 2011.
DOI : 10.1158/0008-5472.CAN-10-2954

T. Wilson, J. Fridlyand, and Y. Yan, Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors, Nature, vol.1, issue.7408, pp.505-514, 2012.
DOI : 10.1158/2159-8290.CD-11-0175

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3724525/pdf

K. Smalley, M. Lioni, and M. Palma, Increased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAF V600E-mutated melanomas, Molecular Cancer Therapeutics, vol.7, issue.9, pp.2876-83, 2008.
DOI : 10.1158/1535-7163.MCT-08-0431

URL : http://mct.aacrjournals.org/content/molcanther/7/9/2876.full.pdf

O. Maertens, B. Johnson, and P. Hollstein, in Melanomagenesis, Cancer Discovery, vol.3, issue.3, pp.338-387, 2013.
DOI : 10.1158/2159-8290.CD-12-0313

J. Larkin, P. Ascierto, and B. Dréno, -Mutated Melanoma, New England Journal of Medicine, vol.371, issue.20, pp.1867-76, 2014.
DOI : 10.1056/NEJMoa1408868

G. Long, D. Stroyakovskiy, and H. Gogas, Combined BRAF and MEK Inhibition versus BRAF Inhibition Alone in Melanoma, New England Journal of Medicine, vol.371, issue.20, pp.1877-88, 2014.
DOI : 10.1056/NEJMoa1406037

G. Long, D. Stroyakovskiy, and H. Gogas, Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial, The Lancet, vol.386, issue.9992, pp.444-51, 2015.
DOI : 10.1016/S0140-6736(15)60898-4

M. Sanlorenzo, A. Choudhry, and I. Vujic, Comparative profile of cutaneous adverse events: BRAF/MEK inhibitor combination therapy versus BRAF monotherapy in melanoma, Journal of the American Academy of Dermatology, vol.71, issue.6, pp.1102-1109, 2014.
DOI : 10.1016/j.jaad.2014.09.002

K. Kim, R. Kefford, and A. Pavlick, -Mutant Cutaneous Melanoma Previously Treated With or Without a BRAF Inhibitor, Journal of Clinical Oncology, vol.31, issue.4, pp.482-491, 2013.
DOI : 10.1200/JCO.2012.43.5966

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4878037/pdf

G. Long, C. Fung, and A. Menzies, Increased MAPK reactivation in early resistance to dabrafenib/trametinib combination therapy of BRAF-mutant metastatic melanoma, Nature Communications, vol.129, p.5694, 2014.
DOI : 10.1038/jid.2009.5

H. Shi, W. Hugo, and X. Kong, Acquired Resistance and Clonal Evolution in Melanoma during BRAF Inhibitor Therapy, Cancer Discovery, vol.4, issue.1, pp.80-93, 2014.
DOI : 10.1158/2159-8290.CD-13-0642

URL : http://cancerdiscovery.aacrjournals.org/content/candisc/4/1/80.full.pdf

R. Carvajal, C. Antonescu, and J. Wolchok, KIT as a Therapeutic Target in Metastatic Melanoma, JAMA, vol.305, issue.22, pp.2327-2361, 2011.
DOI : 10.1001/jama.2011.746

S. Lee, T. Kim, and Y. Kim, Phase II Trial of Nilotinib in Patients With Metastatic Malignant Melanoma Harboring KIT Gene Aberration: A Multicenter Trial of Korean Cancer Study Group (UN10-06), The Oncologist, vol.20, issue.11, pp.1312-1321, 2015.
DOI : 10.1634/theoncologist.2015-0161

J. Guo, L. Si, and Y. Kong, Mutation or Amplification, Journal of Clinical Oncology, vol.29, issue.21, pp.2904-2913, 2011.
DOI : 10.1200/JCO.2010.33.9275

J. Delyon, S. Chevret, and T. Jouary, STAT3 Mediates Nilotinib Response in KIT-Altered Melanoma: A Phase II Multicenter Trial of the French Skin Cancer Network, Journal of Investigative Dermatology, 2017.
DOI : 10.1016/j.jid.2017.07.839

A. Platz, S. Egyhazi, U. Ringborg, and J. Hansson, mutation frequencies in relation to histogenetic subclass and body site, Molecular Oncology, vol.19, issue.4, pp.395-405, 2008.
DOI : 10.1093/emboj/19.20.5429

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5543839/pdf

D. Johnson and I. Puzanov, Treatment of NRAS-Mutant Melanoma, Current Treatment Options in Oncology, vol.70, issue.9, p.15, 2015.
DOI : 10.1158/0008-5472.CAN-10-0118

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4830486/pdf

R. Dummer, D. Schadendorf, and P. Ascierto, Binimetinib versus dacarbazine in patients with advanced NRAS -mutant melanoma (NEMO): a multicentre, open-label, randomised, phase 3 trial, The Lancet Oncology, vol.18, issue.4, pp.435-480, 2017.
DOI : 10.1016/S1470-2045(17)30180-8

L. Balducci and M. Extermann, Management of Cancer in the Older Person: A Practical Approach, The Oncologist, vol.5, issue.3, pp.224-261, 2000.
DOI : 10.1634/theoncologist.5-3-224

P. Soubeyran, C. Bellera, and J. Goyard, Screening for Vulnerability in Older Cancer Patients: The ONCODAGE Prospective Multicenter Cohort Study, PLoS ONE, vol.49, issue.12, 2014.
DOI : 10.1371/journal.pone.0115060.s002

R. Corre, L. Greillier, L. Caër, and H. , Use of a Comprehensive Geriatric Assessment for the Management of Elderly Patients With Advanced Non???Small-Cell Lung Cancer: The Phase III Randomized ESOGIA-GFPC-GECP 08-02 Study, Journal of Clinical Oncology, vol.34, issue.13, pp.1476-83, 2016.
DOI : 10.1200/JCO.2015.63.5839

URL : https://hal.archives-ouvertes.fr/hal-01282337

M. Extermann and A. Hurria, Comprehensive Geriatric Assessment for Older Patients With Cancer, Journal of Clinical Oncology, vol.25, issue.14, pp.1824-1855, 2007.
DOI : 10.1200/JCO.2007.10.6559

A. Iuga and M. Mcguire, Adherence and health care costs, Risk Manag Healthc Policy, vol.7, pp.35-44, 2014.

C. Townsley, G. Pond, and A. Oza, Evaluation of Adverse Events Experienced by Older Patients Participating in Studies of Molecularly Targeted Agents Alone or in Combination, Clinical Cancer Research, vol.12, issue.7, pp.2141-2150, 2006.
DOI : 10.1158/1078-0432.CCR-05-1798

A. Daste, C. Chakiba, and C. Domblides, Targeted therapy and elderly people: A review, European Journal of Cancer, vol.69
DOI : 10.1016/j.ejca.2016.10.005

O. Merimsky, C. Cheng, and J. Au, Efficacy and safety of first-line erlotinib in elderly patients with advanced non-small cell lung cancer, Oncology Reports, vol.28, pp.721-728, 2012.
DOI : 10.3892/or.2012.1824

H. Yoshioka, K. Komuta, and F. Imamura, Efficacy and safety of erlotinib in elderly patients in the phase IV POLARSTAR surveillance study of Japanese patients with non-small-cell lung cancer, Lung Cancer, vol.86, issue.2, pp.201-207, 2014.
DOI : 10.1016/j.lungcan.2014.09.015

K. Yamada, K. Azuma, and M. Takeshita, Phase II Trial of Erlotinib in Elderly Patients with Previously Treated Non-small Cell Lung Cancer: Results of the Lung Oncology Group in Kyushu (LOGiK-0802), Anticancer Res, vol.36, pp.2881-2888, 2016.

K. Tateishi, T. Ichiyama, and K. Hirai, Clinical outcomes in elderly patients administered gefitinib as first-line treatment in epidermal growth factor receptor-mutated non-smallcell lung cancer: retrospective analysis in a Nagano Lung Cancer Research Group study

L. Crinò, F. Cappuzzo, and P. Zatloukal, Gefitinib Versus Vinorelbine in Chemotherapy-Na??ve Elderly Patients With Advanced Non???Small-Cell Lung Cancer (INVITE): A Randomized, Phase II Study, Journal of Clinical Oncology, vol.26, issue.26, pp.4253-60, 2008.
DOI : 10.1200/JCO.2007.15.0672

T. Kuwako, H. Imai, and T. Masuda, First-line gefitinib treatment in elderly patients (aged ???75??years) with non-small cell lung cancer harboring EGFR mutations, Cancer Chemotherapy and Pharmacology, vol.98, issue.Suppl 7, pp.761-770, 2015.
DOI : 10.1111/j.1349-7006.2007.00607.x

T. Losanno and C. Gridelli, Recent advances in targeted advanced lung cancer therapy in the elderly, Expert Review of Anticancer Therapy, vol.36, issue.6, pp.787-97, 2017.
DOI : 10.1007/s10238-017-0460-7

T. Hutson, R. Bukowski, and B. Rini, Efficacy and safety of sunitinib in elderly patients with metastatic renal cell carcinoma, British Journal of Cancer, vol.33, issue.5, pp.1125-1157, 2014.
DOI : 10.1093/annonc/mdq070

A. Brunello, U. Basso, and C. Sacco, Safety and activity of sunitinib in elderly patients (???70 years) with metastatic renal cell carcinoma: a multicenter study, Annals of Oncology, vol.24, issue.2, pp.336-378, 2013.
DOI : 10.1093/annonc/mds431

D. Giorgi, U. Scarpi, E. Sacco, and C. , Standard vs Adapted Sunitinib Regimen in Elderly Patients With Metastatic Renal Cell Cancer: Results From a Large Retrospective Analysis, Clinical Genitourinary Cancer, vol.12, issue.3, pp.182-191, 2014.
DOI : 10.1016/j.clgc.2013.11.005

T. Eisen, S. Oudard, and C. Szczylik, Sorafenib for Older Patients With Renal Cell Carcinoma: Subset Analysis From a Randomized Trial, JNCI Journal of the National Cancer Institute, vol.100, issue.20, pp.1454-63, 2008.
DOI : 10.1093/jnci/djn319

F. Zustovich and G. Novara, Advanced kidney cancer: treating the elderly, Expert Review of Anticancer Therapy, vol.69, issue.15, pp.1389-98, 2013.
DOI : 10.1056/NEJMoa066838

C. Porta, E. Calvo, and M. Climent, Efficacy and Safety of Everolimus in Elderly Patients With Metastatic Renal Cell Carcinoma: An Exploratory Analysis of the Outcomes of Elderly Patients in the RECORD-1 Trial, European Urology, vol.61, issue.4, pp.826-859, 2012.
DOI : 10.1016/j.eururo.2011.12.057

G. Rosti, I. Iacobucci, and S. Bassi, Impact of age on the outcome of patients with chronic myeloid leukemia in late chronic phase: results of a phase II study of the GIMEMA CML Working Party, Haematologica, vol.92, issue.1, pp.101-106, 2007.
DOI : 10.3324/haematol.10239

J. Cortes, M. Talpaz, O. Brien, and S. , Effects of age on prognosis with imatinib mesylate therapy for patients with Philadelphia chromosome-positive chronic myelogenous leukemia, Cancer, vol.11, issue.6, pp.1105-1118, 2003.
DOI : 10.1002/cncr.11629

R. Latagliata, M. Breccia, and I. Carmosino, ???Real-life??? results of front-line treatment with Imatinib in older patients (???65 years) with newly diagnosed chronic myelogenous leukemia, Leukemia Research, vol.34, issue.11, pp.1472-1477, 2010.
DOI : 10.1016/j.leukres.2010.07.001

O. Sheehy, A. Irvine, and R. Cuthbert, Use of imatinib mesylate in elderly patients in Northern Ireland: evidence of comparable haematological and molecular responses to younger patients, Hematology, vol.25, issue.3, pp.133-139, 2008.
DOI : 10.1016/j.blre.2005.01.008

A. Italiano, E. Saada, and A. Cioffi, Treatment of advanced gastrointestinal stromal tumors in patients over 75??years old: clinical and pharmacological implications, Targeted Oncology, vol.63, issue.4, pp.295-300, 2013.
DOI : 10.1093/gerona/63.7.724

M. Kozloff, J. Berlin, and P. Flynn, Clinical Outcomes in Elderly Patients with Metastatic Colorectal Cancer Receiving Bevacizumab and Chemotherapy: Results from the BRiTE Observational Cohort Study, Oncology, vol.78, issue.5-6, pp.329-368, 2010.
DOI : 10.1159/000320222

S. Jang, C. Zheng, and H. Tsai, Cardiovascular toxicity after antiangiogenic therapy in persons older than 65 years with advanced renal cell carcinoma, Cancer, vol.52, issue.1, pp.124-154, 2016.
DOI : 10.1097/MLR.0b013e31825a8c22

URL : http://onlinelibrary.wiley.com/doi/10.1002/cncr.29728/pdf

D. Ciocan, C. Barbe, and F. Aubin, Distinctive Features of Melanoma and Its Management in Elderly Patients, JAMA Dermatology, vol.149, issue.10, pp.1150-1157, 2013.
DOI : 10.1001/jamadermatol.2013.706

F. Grange, C. Barbe, and L. Mas, The role of general practitioners in diagnosis of cutaneous melanoma: a population-based study in France, British Journal of Dermatology, vol.144, issue.Suppl. 1, pp.1351-1360, 2012.
DOI : 10.1001/archderm.144.5.629

URL : https://hal.archives-ouvertes.fr/hal-00727660

S. Weiss, J. Han, and F. Darvishian, Impact of aging on host immune response and survival in melanoma: an analysis of 3 patient cohorts, Journal of Translational Medicine, vol.77, issue.1, pp.10-1186, 2016.
DOI : 10.1002/(SICI)1097-0142(19960401)77:7<1303::AID-CNCR12>3.0.CO;2-5

A. Du-thanh, C. Lesage, and E. Ferreira, Place des traitements innovants du m??lanome m??tastatique chez le sujet ??g??, Annales de Dermatologie et de V??n??r??ologie, vol.142, issue.10, pp.549-56, 2015.
DOI : 10.1016/j.annder.2015.03.022

C. Friedman and J. Wolchok, Checkpoint inhibition and melanoma: Considerations in treating the older adult, Journal of Geriatric Oncology, vol.8, issue.4, pp.237-278, 2017.
DOI : 10.1016/j.jgo.2017.04.003

T. Nishijima, H. Muss, S. Shachar, and S. Moschos, Comparison of efficacy of immune checkpoint inhibitors (ICIs) between younger and older patients: A systematic review and meta-analysis, Cancer Treatment Reviews, vol.45, pp.30-37, 2016.
DOI : 10.1016/j.ctrv.2016.02.006

C. Sileni, V. Pigozzo, J. Ascierto, and P. , Efficacy and safety of ipilimumab in elderly patients with pretreated advanced melanoma treated at Italian centres through the expanded access programme, Journal of Experimental & Clinical Cancer Research, vol.33, issue.1, p.30, 2014.
DOI : 10.1189/jlb.0108023

F. Hodi, J. Chesney, and A. Pavlick, Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial, The Lancet Oncology, vol.17, issue.11, pp.1558-68, 2016.
DOI : 10.1016/S1470-2045(16)30366-7

C. Friedman, T. Horvat, and J. Minehart, Efficacy and safety of checkpoint blockade for treatment of advanced melanoma (mel) in patients (pts) age 80 and older (80+), J Clin Oncol, vol.34, pp.10009-10009, 2016.

P. Austin, C. Cruse, and G. Lyman, Age as a prognostic factor in the malignant melanoma population, Annals of Surgical Oncology, vol.218, issue.6, pp.487-94, 1994.
DOI : 10.1093/ajcp/74.3.247

C. Chao, R. Martin, and M. Ross, Correlation Between Prognostic Factors and Increasing Age in Melanoma, Annals of Surgical Oncology, vol.86, issue.(9 Suppl), pp.259-64, 2004.
DOI : 10.1245/ASO.2004.04.015

M. Rousset, K. Titier, and S. Bouchet, An UPLC-MS/MS method for the quantification of BRAF inhibitors (vemurafenib, dabrafenib) and MEK inhibitors (cobimetinib, trametinib, binimetinib) in human plasma. Application to treated melanoma patients, Clinica Chimica Acta, vol.470, pp.8-13, 2017.
DOI : 10.1016/j.cca.2017.04.009

M. Rousset, C. Dutriaux, and P. Bosco-lévy, Trough dabrafenib plasma concentrations can predict occurrence of adverse events requiring dose reduction in metastatic melanoma, Clinica Chimica Acta, vol.472, pp.26-35, 2017.
DOI : 10.1016/j.cca.2017.07.012

C. Lee, A. Menzies, and L. Haydu, Features and management of pyrexia with combined dabrafenib and trametinib in metastatic melanoma, Melanoma Research, vol.24, issue.5, pp.468-74, 2014.
DOI : 10.1097/CMR.0000000000000110

C. Robert, B. Karaszewska, and J. Schachter, Improved Overall Survival in Melanoma with Combined Dabrafenib and Trametinib, New England Journal of Medicine, vol.372, issue.1, pp.30-39, 2015.
DOI : 10.1056/NEJMoa1412690

V. Belum, A. Fischer, J. Choi, and M. Lacouture, Dermatological Adverse Events from BRAF Inhibitors: A Growing Problem, Current Oncology Reports, vol.365, issue.15, pp.249-59, 2013.
DOI : 10.1074/jbc.273.34.21730

G. Hatzivassiliou, K. Song, and I. Yen, RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth, Nature, vol.50, issue.7287, pp.431-436, 2010.
DOI : 10.1128/MCB.14.12.8212

C. Hughes, Medication Non-Adherence in the Elderly, Drugs & Aging, vol.15, issue.12, pp.793-811, 2004.
DOI : 10.2165/00002512-200421120-00004

N. Campbell, M. Boustani, and E. Skopelja, Medication Adherence in Older Adults With Cognitive Impairment: A Systematic Evidence-Based Review, The American Journal of Geriatric Pharmacotherapy, vol.10, issue.3, pp.165-77, 2012.
DOI : 10.1016/j.amjopharm.2012.04.004

M. Sharma, K. Loh, and G. Nightingale, Polypharmacy and potentially inappropriate medication use in geriatric oncology, Journal of Geriatric Oncology, vol.7, issue.5, pp.346-53, 2016.
DOI : 10.1016/j.jgo.2016.07.010

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5037024/pdf

J. Greer, N. Amoyal, and L. Nisotel, A Systematic Review of Adherence to Oral Antineoplastic Therapies, The Oncologist, vol.21, issue.3, pp.354-76, 2016.
DOI : 10.1634/theoncologist.2015-0405

P. Kardas, P. Lewek, and M. Matyjaszczyk, Determinants of patient adherence: a review of systematic reviews, Frontiers in Pharmacology, vol.4, p.91, 2013.
DOI : 10.3389/fphar.2013.00091

R. Harvey, T. Mckibbin, M. Behera, and S. Ramalingam, Adherence reporting in phase III trials of approved oral anticancer agents: A systematic review, J Clin Oncol, vol.31, pp.20596-20596, 2013.

M. Verbrugghe, V. Duprez, and D. Beeckman, Factors Influencing Adherence in Cancer Patients Taking Oral Tyrosine Kinase Inhibitors, Cancer Nursing, vol.39, issue.2, pp.153-62, 2016.
DOI : 10.1097/NCC.0000000000000250

H. Klepin, A. Geiger, and H. Bandos, Cognitive Factors Associated with Adherence to Oral Antiestrogen Therapy: Results from the Cognition in the Study of Tamoxifen and Raloxifene (Co-STAR) Study, Cancer Prevention Research, vol.7, issue.1, pp.161-169, 2014.
DOI : 10.1158/1940-6207.CAPR-13-0165

A. Mislang, T. Wildes, and R. Kanesvaran, Adherence to oral cancer therapy in older adults: The International Society of Geriatric Oncology (SIOG) taskforce recommendations, Cancer Treatment Reviews, vol.57, pp.58-66, 2017.
DOI : 10.1016/j.ctrv.2017.05.002

R. Haynes, E. Ackloo, and N. Sahota, Interventions for enhancing medication adherence, Cochrane Database Syst Rev, vol.288, issue.3, p.11, 2008.
DOI : 10.1161/01.HYP.4.3.415

D. Ouellet, K. Grossmann, and G. Limentani, Effects of particle size, food, and capsule shell composition on the oral bioavailability of dabrafenib, a BRAF inhibitor, in patients with BRAF mutation-positive tumors, Journal of Pharmaceutical Sciences, vol.102, issue.9, pp.3100-3109, 2013.
DOI : 10.1002/jps.23519

D. Ouellet, E. Gibiansky, and C. Leonowens, Population pharmacokinetics of dabrafenib, a BRAF inhibitor: Effect of dose, time, covariates, and relationship with its metabolites, The Journal of Clinical Pharmacology, vol.12, issue.Suppl. 1, pp.696-706, 2014.
DOI : 10.2174/138920011796504527

A. Mangoni and S. Jackson, Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications, British Journal of Clinical Pharmacology, vol.20, issue.Suppl 8, pp.6-14, 2004.
DOI : 10.1111/j.1365-2125.1985.tb05041.x

URL : http://onlinelibrary.wiley.com/doi/10.1046/j.1365-2125.2003.02007.x/pdf

N. Kramkimel, A. Thomas-schoemann, and L. Sakji, Vemurafenib pharmacokinetics and its correlation with efficacy and safety in outpatients with advanced BRAF-mutated melanoma, Targeted Oncology, vol.25, issue.1, pp.59-69, 2016.
DOI : 10.1093/annonc/mdt587

S. Park, S. Park, and S. Lee, Pretreatment albumin-to-globulin ratio as a predictive marker for tyrosine kinase inhibitor in non-small cell lung cancer, Cancer Biomarkers, vol.27, issue.3, pp.425-458, 2016.
DOI : 10.1200/JCO.2008.19.8440

V. Ziller, I. Kyvernitakis, and D. Knöll, Influence of a patient information program on adherence and persistence with an aromatase inhibitor in breast cancer treatment - the COMPAS study, BMC Cancer, vol.37, issue.4, p.407, 2013.
DOI : 10.3109/02770900009055457

S. Simons, S. Ringsdorf, and M. Braun, Enhancing adherence to capecitabine chemotherapy by means of multidisciplinary pharmaceutical care, Supportive Care in Cancer, vol.8, issue.Suppl 1, pp.1009-1027, 2011.
DOI : 10.1016/j.ejon.2004.06.008

F. Stevenson, K. Cox, N. Britten, and Y. Dundar, A systematic review of the research on communication between patients and health care professionals about medicines: the consequences for concordance, Health Expectations, vol.50, issue.3, pp.235-280, 2004.
DOI : 10.1053/seiz.1998.0232

M. Viswanathan, C. Golin, and C. Jones, Interventions to improve adherence to selfadministered medications for chronic diseases in the United States: a systematic review

P. Creel, Optimizing Patient Adherence to Targeted Therapies in Renal Cell Carcinoma, Clinical Journal of Oncology Nursing, vol.18, issue.6, pp.694-700, 2014.
DOI : 10.1188/14.CJON.694-700

A. Brunot, M. Sadek, A. , L. Roy, and F. , Les infirmi??res cliniciennes am??liorent la gestion des toxicit??s des patients trait??s par soraf??nib pour carcinome h??patocellulaire, Bulletin du Cancer, vol.103, issue.11, pp.941-949, 2016.
DOI : 10.1016/j.bulcan.2016.09.017

J. Alvarez, E. Funck-brentano, and E. Abe, A LC/MS/MS micro-method for human plasma quantification of vemurafenib. Application to treated melanoma patients, Journal of Pharmaceutical and Biomedical Analysis, vol.97, pp.29-32, 2014.
DOI : 10.1016/j.jpba.2014.04.014

N. Lankheet, I. Desar, and S. Mulder, Optimizing the dose in cancer patients treated with imatinib, sunitinib and pazopanib, Br J Clin Pharmacol, 2017.

V. Gotta, N. Widmer, and L. Decosterd, Clinical usefulness of therapeutic concentration monitoring for imatinib dosage individualization: results from a randomized controlled trial, Cancer Chemotherapy and Pharmacology, vol.71, issue.1, pp.1307-1326, 2014.
DOI : 10.1007/s00280-012-1998-4

D. Frederick, A. Piris, and A. Cogdill, BRAF Inhibition Is Associated with Enhanced Melanoma Antigen Expression and a More Favorable Tumor Microenvironment in Patients with Metastatic Melanoma, Clinical Cancer Research, vol.19, issue.5, pp.1225-1256, 2013.
DOI : 10.1158/1078-0432.CCR-12-1630

J. Wilmott, G. Long, and J. Howle, Selective BRAF Inhibitors Induce Marked T-cell Infiltration into Human Metastatic Melanoma, Clinical Cancer Research, vol.18, issue.5, pp.1386-94, 2012.
DOI : 10.1158/1078-0432.CCR-11-2479

URL : http://clincancerres.aacrjournals.org/content/clincanres/18/5/1386.full.pdf

D. Minor, I. Puzanov, and M. Callahan, Severe gastrointestinal toxicity with administration of trametinib in combination with dabrafenib and ipilimumab, Pigment Cell & Melanoma Research, vol.28, issue.5, pp.611-613, 2015.
DOI : 10.1111/pcmr.12383

A. Ribas, M. Butler, and J. Lutzky, Phase I study combining anti-PD-L1 (MEDI4736) with BRAF (dabrafenib) and/or MEK (trametinib) inhibitors in advanced melanoma, J Clin Oncol, vol.33, pp.3003-3003, 2015.

A. Ribas, F. Hodi, and D. Lawrence, Pembrolizumab (pembro) in combination with dabrafenib (D) and trametinib (T) for BRAF-mutant advanced melanoma: Phase 1 KEYNOTE-022 study, J Clin Oncol, vol.34, pp.3014-3014, 2016.

A. Ackerman, O. Klein, and D. Mcdermott, Outcomes of patients with metastatic melanoma treated with immunotherapy prior to or after BRAF inhibitors, Cancer, vol.13, issue.suppl, pp.1695-701, 2014.
DOI : 10.1016/S1470-2045(12)70431-X

P. Ascierto, E. Simeone, and V. Sileni, Sequential Treatment with Ipilimumab and BRAF Inhibitors in Patients With Metastatic Melanoma: Data From the Italian Cohort of the Ipilimumab Expanded Access Program, Cancer Investigation, vol.11, issue.6, pp.144-153, 2014.
DOI : 10.1200/JCO.2009.23.4799