, BP, vol.10448

. Anatomopathology-department, J. Centre, and . Perrin, Comprehensive Cancer Center, vol.58

, Inserm U, vol.1107

D. and C. , , p.63003

C. , I. , and I. Curie, Bat, vol.110

D. Therapeutics and . Sa,-4-rue-pierre-fontaine, 91058 Evry Cedex Disclosure of potential conflict of Interest: M. Dutreix and J-S Sun are cofounders of DNA therapeutics

J. Ferlay, I. Soerjomataram, and R. Dikshit, Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012, Int J Cancer, 2014.

J. A. Lo and D. E. Fisher, The melanoma revolution: from UV carcinogenesis to a new era in therapeutics, Science, vol.346, issue.6212, pp.945-949, 2014.

E. Revskaya, A. M. Jongco, and R. S. Sellers, Radioimmunotherapy of experimental human metastatic melanoma with melanin-binding antibodies and in combination with dacarbazine, Clin Cancer Res, vol.15, issue.7, pp.2373-2379, 2009.

J. Chezal, J. Papon, and P. Labarre, Evaluation of radiolabeled (hetero)aromatic analogues of N-(2-diethylaminoethyl)-4-iodobenzamide for imaging and targeted radionuclide therapy of melanoma, /jm701424g. 5. Bonnet-Duquennoy M, vol.51, pp.708-716, 2008.

M. Bonnet, F. Mishellany, and J. Papon, Anti-melanoma efficacy of internal radionuclide therapy in relation to melanin target distribution, Pigment Cell Melanoma Res, vol.23, issue.5, pp.1-11, 2010.

J. L. Joyal, J. A. Barrett, and J. C. Marquis, Preclinical evaluation of an 131I-labeled benzamide for targeted radiotherapy of metastatic melanoma, Cancer Res, vol.70, issue.10, pp.4045-4053, 2010.

F. Degoul, M. Borel, and N. Jacquemot, In vivo efficacy of melanoma internal radionuclide therapy with a 131I-labelled melanin-targeting heteroarylcarboxamide molecule, Int J Cancer, vol.133, issue.5, pp.1042-1053, 2013.
URL : https://hal.archives-ouvertes.fr/in2p3-00926259

W. Mier, C. Kratochwil, and J. C. Hassel, Radiopharmaceutical therapy of patients with metastasized melanoma with the melanin-binding benzamide 131I-BA52, J Nucl Med, vol.55, issue.1, pp.9-14, 2014.

F. Cachin, E. Miot-noirault, and B. Gillet, )I-BZA2 as a melanin-targeted radiotracer for the identification of melanoma metastases: results and perspectives of a multicenter phase III clinical trial, J Nucl Med, vol.55, issue.123, pp.15-22, 2014.

C. Viallard, Y. Perrot, and Z. Boudhraa, [ 123 I]ICF01012 melanoma imaging and [ 131 I]ICF01012 dosimetry allow adapted internal targeted radiotherapy in preclinical melanoma models, Eur J Dermatol EJD, vol.25, issue.1, pp.29-35, 2015.

M. K. Khan, N. Khan, A. Almasan, and R. Macklis, Future of radiation therapy for malignant melanoma in an era of newer, more effective biological agents, OncoTargets Ther, vol.4, pp.137-148, 2011.

E. Dadachova, R. A. Bryan, and R. C. Howell, The radioprotective properties of fungal melanin are a function of its chemical composition, stable radical presence and spatial arrangement, Pigment Cell Melanoma Res, vol.21, issue.2, pp.192-199, 2008.

A. Munshi, J. F. Kurland, and T. Nishikawa, Histone deacetylase inhibitors radiosensitize human melanoma cells by suppressing DNA repair activity, Clin Cancer Res, vol.11, issue.13, pp.4912-4922, 2005.

R. L. Warters, P. J. Adamson, C. D. Pond, and S. A. Leachman, Melanoma cells express elevated levels of phosphorylated histone H2AX foci, J Invest Dermatol, vol.124, issue.4, pp.807-817, 2005.

W. K. Kaufmann, K. R. Nevis, and P. Qu, Defective cell cycle checkpoint functions in melanoma are associated with altered patterns of gene expression, J Invest Dermatol, vol.128, issue.1, pp.175-187, 2008.

S. A. Amundson, K. T. Do, and L. C. Vinikoor, Integrating global gene expression and radiation survival parameters across the 60 cell lines of the National Cancer Institute Anticancer Drug Screen, Cancer Res, vol.68, issue.2, pp.415-424, 2008.

R. Plummer, P. Lorigan, and N. Steven, A phase II study of the potent PARP inhibitor, Rucaparib (PF-01367338, AG014699), with temozolomide in patients with metastatic melanoma demonstrating evidence of chemopotentiation, Cancer Chemother Pharmacol, vol.71, issue.5, pp.1191-1199, 2013.

A. Croset, F. P. Cordelières, and N. Berthault, Hyperactivation of DNA-PK by double-strand break mimicking molecules disorganizes DNA damage response, Nucleic Acids Res, vol.41, issue.15, pp.7344-7355, 2013.

, , p.6298

J. Biau, F. Devun, and W. Jdey, A preclinical study combining the DNA repair inhibitor dbait with radiotherapy for the treatment of melanoma, Neoplasia, vol.16, issue.10, pp.835-844, 2014.

N. Coquery, N. Pannetier, and R. Farion, Distribution and radiosensitizing effect of cholesterol-coupled Dbait molecule in rat model of glioblastoma, PloS One, vol.7, issue.7, p.40567, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00874418

M. Quanz, N. Berthault, and C. Roulin, Small-molecule drugs mimicking DNA damage: a new strategy for sensitizing tumors to radiotherapy, Clin Cancer Res Off J Am Assoc Cancer Res, vol.15, issue.4, pp.1308-1316, 2009.

F. Devun, G. Bousquet, and J. Biau, Preclinical study of the DNA repair inhibitor Dbait in combination with chemotherapy in colorectal cancer, J Gastroenterol, vol.47, issue.3, pp.266-275, 2012.

A. Schlegel, C. Buhler, and F. Devun, Pharmacokinetics and Toxicity in Rats and Monkeys of coDbait: A Therapeutic Double-stranded DNA Oligonucleotide Conjugated to Cholesterol, Mol Ther Nucleic Acids, vol.1, p.33, 2012.

L. Chen, S. Zheng, and Z. Sun, Cryptotanshinone has diverse effects on cell cycle events in melanoma cell lines with different metastatic capacity, Cancer Chemother Pharmacol, vol.68, issue.1, pp.17-27, 2011.

S. Bamford, E. Dawson, and S. Forbes, The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website, Br J Cancer, vol.91, issue.2, pp.355-358, 2004.

S. Forbes, J. Clements, and E. Dawson, Br J Cancer, vol.94, issue.2, pp.318-322, 2005.

B. Omolo, C. Carson, and H. Chu, A prognostic signature of G(2) checkpoint function in melanoma cell lines, Cell Cycle, vol.12, issue.7, pp.1071-1082, 2013.

F. Zölzer, T. Mußfeldt, and C. Streffer, Differential S-phase progression after irradiation of p53 functional versus non-functional tumour cells, Radiol Oncol, vol.48, issue.4, pp.354-360, 2014.

V. G. Gorgoulis, L. Vassiliou, and P. Karakaidos, Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions, Nature, vol.434, issue.7035, pp.907-913, 2005.

N. Berthault, B. Maury, and C. Agrario, Comparison of distribution and activity of nanoparticles with short interfering DNA (Dbait) in various living systems, Cancer Gene Ther, vol.18, issue.10, pp.695-706, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00683650

T. Kim, R. N. Amaria, C. Spencer, A. Reuben, Z. A. Cooper et al., Combining targeted therapy and immune checkpoint inhibitors in the treatment of metastatic melanoma, Cancer Biol Med, vol.11, issue.4, pp.237-246, 2014.

C. Letourneau, B. Dreno, and Y. Kirova, First-in-human phase I study of the DNA repair inhibitor DT01 in combination with radiotherapy in patients with in transit melanoma, Abstract number : 2555), 2015.

N. R. Abbasi, H. M. Shaw, D. S. Rigel, R. J. Friedman, W. H. Mccarthy et al., Early diagnosis of cutaneous melanoma: revisiting the ABCD criteria, JAMA, vol.292, pp.2771-2776, 2004.

V. Agrawal, S. Maharjan, K. Kim, N. Kim, J. Son et al., Direct endothelial junction restoration results in significant tumor vascular normalization and metastasis inhibition in mice, Oncotarget, vol.5, pp.2761-2777, 2014.

R. Akbani, K. C. Akdemir, B. A. Aksoy, M. Albert, A. Ally et al., Genomic Classification of Cutaneous Melanoma, vol.161, pp.1681-1696, 2015.

J. M. Albert, C. Cao, K. W. Kim, C. D. Willey, L. Geng et al., Inhibition of poly(ADP-ribose) polymerase enhances cell death and improves tumor growth delay in irradiated lung cancer models, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.13, pp.3033-3042, 2007.

E. Aleem and R. J. Arceci, Targeting cell cycle regulators in hematologic malignancies, Front. Cell Dev. Biol, vol.3, 2015.

B. J. Allen, A. A. Singla, S. M. Rizvi, P. Graham, F. Bruchertseifer et al., Analysis of patient survival in a Phase I trial of systemic targeted ?-therapy for metastatic melanoma, Immunotherapy, vol.3, pp.1041-1050, 2011.

J. Amé, C. Spenlehauer, and G. De-murcia, The PARP superfamily, BioEssays News Rev. Mol. Cell. Dev. Biol, vol.26, pp.882-893, 2004.

S. A. Amundson, K. T. Do, L. C. Vinikoor, R. A. Lee, C. A. Koch-paiz et al., Integrating global gene expression and radiation survival parameters across the 60 cell lines of the National Cancer Institute Anticancer Drug Screen, Cancer Res, vol.68, pp.415-424, 2008.

H. Ando, Y. Niki, M. Yoshida, M. Ito, K. Akiyama et al., Involvement of pigment globules containing multiple melanosomes in the transfer of melanosomes from melanocytes to keratinocytes, Cell. Logist, vol.1, pp.12-20, 2011.

H. Ando, Y. Niki, M. Ito, K. Akiyama, M. S. Matsui et al., Melanosomes are transferred from melanocytes to keratinocytes through the processes of packaging, release, uptake, and dispersion, J. Invest. Dermatol, vol.132, pp.1222-1229, 2012.

A. Apel, H. Zentgraf, M. W. Büchler, and I. Herr, Autophagy-A double-edged sword in oncology, Int. J. Cancer, vol.125, pp.991-995, 2009.

C. Arienti, A. Tesei, S. Carloni, P. Ulivi, A. Romeo et al., SLUG silencing increases radiosensitivity of melanoma cells in vitro, Cell. Oncol. Dordr, vol.36, pp.131-139, 2013.

P. A. Ascierto, F. M. Marincola, and M. B. Atkins, What's new in melanoma?, Combination! J. Transl. Med, vol.13, p.213, 2015.

J. M. Atkinson, K. B. Rank, Y. Zeng, A. Capen, V. Yadav et al., Activating the Wnt/?-Catenin Pathway for the Treatment of Melanoma-Application of LY2090314, a Novel Selective Inhibitor of Glycogen Synthase Kinase-3, PLoS ONE, vol.10, 2015.

J. W. Baish, T. Stylianopoulos, R. M. Lanning, W. S. Kamoun, D. Fukumura et al., Scaling rules for diffusive drug delivery in tumor and normal tissues, Proc. Natl. Acad. Sci, vol.108, pp.1799-1803, 2011.

M. Balu, K. M. Kelly, C. B. Zachary, R. M. Harris, T. B. Krasieva et al., Distinguishing between Benign and Malignant Melanocytic Nevi by In Vivo Multiphoton Microscopy, Cancer Res, vol.74, pp.2688-2697, 2014.

B. Bandarchi, L. Ma, R. Navab, A. Seth, R. et al., From melanocyte to metastatic malignant melanoma, Dermatol. Res. Pract, 2010.

C. Bertolotto, Melanoma: from melanocyte to genetic alterations and clinical options, p.635203, 2013.

J. Biau, F. Devun, W. Jdey, E. Kotula, M. Quanz et al., A preclinical study combining the DNA repair inhibitor dbait with radiotherapy for the treatment of melanoma, Neoplasia, vol.16, pp.835-844, 2014.

T. L. Biechele, N. D. Camp, D. M. Fass, R. M. Kulikauskas, N. C. Robin et al., Chemical-genetic screen identifies riluzole as an enhancer of Wnt/?-catenin signaling in melanoma, Chem. Biol, vol.17, pp.1177-1182, 2010.

K. Bixel and J. L. Hays, Olaparib in the management of ovarian cancer, Pharmacogenomics Pers. Med, vol.8, pp.127-135, 2015.

J. S. Blackburn, C. H. Rhodes, C. I. Coon, and C. E. Brinckerhoff, RNA interference inhibition of matrix metalloproteinase-1 prevents melanoma metastasis by reducing tumor collagenase activity and angiogenesis, Cancer Res, vol.67, pp.10849-10858, 2007.

A. Boire, L. Covic, A. Agarwal, S. Jacques, S. Sherifi et al., PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells, Cell, vol.120, pp.303-313, 2005.

W. E. Bolch, K. F. Eckerman, G. Sgouros, T. , and S. R. , MIRD pamphlet No. 21: a generalized schema for radiopharmaceutical dosimetry-standardization of nomenclature, J. Nucl. Med. Off. Publ. Soc. Nucl. Med, vol.50, pp.477-484, 2009.

M. Bonnet, F. Mishellany, J. Papon, A. Cayre, F. Penault-llorca et al., Anti-melanoma efficacy of internal radionuclide therapy in relation to melanin target distribution, Pigment Cell Melanoma Res, vol.23, pp.1-11, 2010.

M. Bonnet-duquennoy, J. Papon, F. Mishellany, P. Labarre, J. Guerquin-kern et al., Targeted radionuclide therapy of melanoma: anti-tumoural efficacy studies of a new 131I labelled potential agent, Int J Cancer, vol.125, pp.708-716, 2009.

V. Boudousq, S. Ricaud, V. Garambois, C. Bascoul-mollevi, S. Boutaleb et al., Brief intraperitoneal radioimmunotherapy of small peritoneal carcinomatosis using high activities of noninternalizing 125I-labeled monoclonal antibodies, J. Nucl. Med. Off. Publ. Soc. Nucl. Med, vol.51, pp.1748-1755, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00531603

V. Boudousq, L. Bobyk, M. Busson, V. Garambois, M. Jarlier et al., Comparison between internalizing anti-HER2 mAbs and non-internalizing anti-CEA mAbs in alpha-radioimmunotherapy of small volume peritoneal carcinomatosis using 212Pb, PloS One, vol.8, p.69613, 2013.

M. M. Bradford, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding, Anal. Biochem, vol.72, pp.248-254, 1976.

K. A. Bridges, H. Hirai, C. A. Buser, C. Brooks, H. Liu et al., MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.17, pp.5638-5648, 2011.

A. A. Bro?yna, W. Jó?wicki, J. A. Carlson, and A. T. Slominski, Melanogenesis affects overall and disease-free survival in patients with stage III and IV melanoma, Hum. Pathol, vol.44, pp.2071-2074, 2013.

F. Cachin, J. Chezal, E. Miot-noirault, N. Moins, P. Auzeloux et al., Nouveaux traceurs TEMP : exemple des traceurs des protéoglycanes et de la mélanine, Médecine Nucl, vol.33, pp.161-167, 2009.

F. Cachin, E. Miot-noirault, B. Gillet, V. Isnardi, B. Labeille et al., )I-BZA2 as a melanin-targeted radiotracer for the identification of melanoma metastases: results and perspectives of a multicenter phase III clinical trial, J Nucl Med, vol.55, issue.123, pp.15-22, 2014.

V. Calugaru, N. Magné, J. Hérault, S. Bonvalot, C. Le-tourneau et al., , 2015.

, Bull. Cancer (Paris), vol.102, pp.83-91

W. J. Cannan and D. S. Pederson, Mechanisms and Consequences of Double-strand DNA Break Formation in Chromatin, J. Cell. Physiol, 2015.

Z. Cao, B. Shang, G. Zhang, L. Miele, F. H. Sarkar et al., Tumor cellmediated neovascularization and lymphangiogenesis contrive tumor progression and cancer metastasis, Biochim. Biophys. Acta BBA-Rev. Cancer, vol.1836, pp.273-286, 2013.

J. Caramel, E. Papadogeorgakis, L. Hill, G. J. Browne, G. Richard et al., A switch in the expression of embryonic EMTinducers drives the development of malignant melanoma, Cancer Cell, vol.24, pp.466-480, 2013.

P. Carmeliet and R. K. Jain, Molecular mechanisms and clinical applications of angiogenesis, Nature, vol.473, pp.298-307, 2011.

P. Carmeliet and R. K. Jain, Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases, Nat. Rev. Drug Discov, vol.10, pp.417-427, 2011.

C. Chang, C. Chang, C. Shen, C. Chen, R. Liu et al., Synthesis and evaluation of 123 / 131 I-Iochlonicotinamide as a novel SPECT probe for malignant melanoma, Bioorg. Med. Chem, vol.23, pp.2261-2269, 2015.

Y. Cheli, S. Giuliano, S. Guiliano, T. Botton, S. Rocchi et al., Mitf is the key molecular switch between mouse or human melanoma initiating cells and their differentiated progeny, Oncogene, vol.30, pp.2307-2318, 2011.

Y. Cheli, S. Giuliano, N. Fenouille, M. Allegra, V. Hofman et al., Hypoxia and MITF control metastatic behaviour in mouse and human melanoma cells, Oncogene, vol.31, pp.2461-2470, 2012.

Y. Cheli, V. F. Bonnazi, A. Jacquel, M. Allegra, G. M. De-donatis et al., CD271 is an imperfect marker for melanoma initiating cells, Oncotarget, vol.5, pp.5272-5283, 2014.

Y. Cheng, G. Zhang, L. , and G. , Targeting MAPK pathway in melanoma therapy, Cancer Metastasis Rev, vol.32, pp.567-584, 2013.

J. Chezal, J. Papon, P. Labarre, C. Lartigue, M. Galmier et al., Evaluation of radiolabeled (hetero)aromatic analogues of N-(2-diethylaminoethyl)-4-iodobenzamide for imaging and targeted radionuclide therapy of melanoma, J. Med. Chem, vol.51, pp.3133-3144, 2008.

A. J. Chien, E. C. Moore, A. S. Lonsdorf, R. M. Kulikauskas, B. G. Rothberg et al., Activated Wnt/beta-catenin signaling in melanoma is associated with decreased proliferation in patient tumors and a murine melanoma model, Proc. Natl. Acad. Sci. U. S. A, vol.106, pp.1193-1198, 2009.

G. Christofori, New signals from the invasive front, Nature, vol.441, pp.444-450, 2006.

M. Cichorek, M. Wachulska, A. Stasiewicz, and A. Tymi?ska, Skin melanocytes: biology and development, Post?py Dermatol. Alergol, vol.30, pp.30-41, 2013.

G. Civenni, A. Walter, N. Kobert, D. Mihic-probst, M. Zipser et al., Human CD271-positive melanoma stem cells associated with metastasis establish tumor heterogeneity and long-term growth, Cancer Res, vol.71, pp.3098-3109, 2011.

M. F. Clarke, J. E. Dick, P. B. Dirks, C. J. Eaves, C. H. Jamieson et al., Cancer Stem Cells-Perspectives on Current Status and Future Directions: AACR Workshop on Cancer Stem Cells, Cancer Res, vol.66, pp.9339-9344, 2006.

H. Clevers, N. , and R. , Wnt/?-Catenin Signaling and Disease, Cell, vol.149, pp.1192-1205, 2012.

S. Colak and J. P. Medema, Cancer stem cells-important players in tumor therapy resistance, FEBS J, vol.281, pp.4779-4791, 2014.

J. Coppé, P. Desprez, A. Krtolica, C. , and J. , The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression, Annu. Rev. Pathol, vol.5, pp.99-118, 2010.

N. Coquery, N. Pannetier, R. Farion, A. Herbette, L. Azurmendi et al., Distribution and radiosensitizing effect of cholesterolcoupled Dbait molecule in rat model of glioblastoma, PloS One, vol.7, p.40567, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00874418

A. Croset, F. P. Cordelières, N. Berthault, C. Buhler, J. Sun et al., Inhibition of DNA damage repair by artificial activation of PARP with siDNA, Nucleic Acids Res, vol.41, pp.7344-7355, 2013.

B. D. Curti and W. J. Urba, Integrating New Therapies in the Treatment of Advanced Melanoma-Springer, Curr. Treat. Options Oncol, vol.13, pp.327-339, 2012.

E. Dadachova, J. D. Nosanchuk, L. Shi, A. D. Schweitzer, A. Frenkel et al., Dead cells in melanoma tumors provide abundant antigen for targeted delivery of ionizing radiation by a mAb to melanin, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.14865-14870, 2004.

C. Dahl, G. , and P. , The genome and epigenome of malignant melanoma, APMIS Acta Pathol. Microbiol. Immunol. Scand, vol.115, pp.1161-1176, 2007.

F. Dantzer, G. Noel, and V. Schreiber, , 2011.

, Bull. Cancer (Paris), vol.98, pp.277-290

H. Davies, G. R. Bignell, C. Cox, P. Stephens, S. Edkins et al., Mutations of the BRAF gene in human cancer, Nature, vol.417, pp.949-954, 2002.

A. J. Davis, C. , and D. J. , DNA double strand break repair via non-homologous end-joining, Transl. Cancer Res, vol.2, pp.130-143, 2013.

F. Degoul, M. Borel, N. Jacquemot, S. Besse, Y. Communal et al., In vivo efficacy of melanoma internal radionuclide therapy with a 131I-labelled melanin-targeting heteroarylcarboxamide molecule, Int J Cancer, vol.133, pp.1042-1053, 2013.
URL : https://hal.archives-ouvertes.fr/in2p3-00926259

C. Delevoye, F. Giordano, G. Van-niel, R. , and G. , , 2011.

, Médecine Sci. MS, vol.27, pp.153-162

F. Devun, G. Bousquet, J. Biau, A. Herbette, C. Roulin et al., Preclinical study of the DNA repair inhibitor Dbait in combination with chemotherapy in colorectal cancer, J. Gastroenterol, vol.47, pp.266-275, 2012.

H. Dodson, S. P. Wheatley, and C. G. Morrison, Involvement of Centrosome Amplification in Radiation-Induced Mitotic Catastrophe, Cell Cycle, vol.6, pp.364-370, 2007.

L. Duc, G. Miladi, I. Alric, C. Mowat, P. Bräuer-krisch et al., Toward an image-guided microbeam radiation therapy using gadolinium-based nanoparticles, ACS Nano, vol.5, pp.9566-9574, 2011.

E. Dupin and L. Sommer, Neural crest progenitors and stem cells: From early development to adulthood, Dev. Biol, vol.366, pp.83-95, 2012.

S. Elmore, Apoptosis: A Review of Programmed Cell Death, Toxicol. Pathol, vol.35, pp.495-516, 2007.

D. Eriksson and T. Stigbrand, Radiation-induced cell death mechanisms, Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med, vol.31, pp.363-372, 2010.

P. Ernfors, Cellular origin and developmental mechanisms during the formation of skin melanocytes, Exp. Cell Res, vol.316, pp.1397-1407, 2010.

L. A. Fecher, S. D. Cummings, M. J. Keefe, A. , and R. M. , Toward a molecular classification of melanoma, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.25, pp.1606-1620, 2007.

M. Feoktistova and M. Leverkus, Programmed necrosis and necroptosis signalling, FEBS J, vol.282, pp.19-31, 2015.

J. Ferlay, I. Soerjomataram, I. Dikshit, R. Eser, S. Mathers et al., Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012, Int. J. Cancer J. Int. Cancer, 2014.

N. Ferrara, Vascular endothelial growth factor, Arterioscler. Thromb. Vasc. Biol, vol.29, pp.789-791, 2009.

I. J. Fidler, Critical determinants of metastasis, Semin. Cancer Biol, vol.12, pp.89-96, 2002.

D. V. Firsanov, L. V. Solovjeva, and M. P. Svetlova, H2AX phosphorylation at the sites of DNA double-strand breaks in cultivated mammalian cells and tissues, Clin. Epigenetics, vol.2, pp.283-297, 2011.

K. T. Flaherty, I. Puzanov, K. B. Kim, A. Ribas, G. A. Mcarthur et al., Inhibition of mutated, activated BRAF in metastatic melanoma, N. Engl. J. Med, vol.363, pp.809-819, 2010.

S. Forbes, J. Clements, E. Dawson, S. Bamford, T. Webb et al., Br. J. Cancer, vol.94, pp.318-322, 2005.

N. Y. Frank, A. Margaryan, Y. Huang, T. Schatton, A. M. Waaga-gasser et al., ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma, Cancer Res, vol.65, pp.4320-4333, 2005.

R. J. Friedman, D. S. Rigel, and A. W. Kopf, Early detection of malignant melanoma: the role of physician examination and self-examination of the skin, CA. Cancer J. Clin, vol.35, pp.130-151, 1985.

S. J. Gallagher, F. Rambow, M. Kumasaka, D. Champeval, A. Bellacosa et al., Beta-catenin inhibits melanocyte migration but induces melanoma metastasis, Oncogene, vol.32, pp.2230-2238, 2013.

C. Garbe, T. K. Eigentler, U. Keilholz, A. Hauschild, and J. M. Kirkwood, Systematic review of medical treatment in melanoma: current status and future prospects, Oncologist, vol.16, pp.5-24, 2011.

Z. Gatalica, C. Snyder, T. Maney, A. Ghazalpour, D. A. Holterman et al., Programmed Cell Death 1 (PD-1) and Its Ligand (PD-L1) in Common Cancers and Their Correlation with Molecular Cancer Type, Cancer Epidemiol. Biomarkers Prev, vol.23, pp.2965-2970, 2014.

A. Gembarska, F. Luciani, C. Fedele, E. A. Russell, M. Dewaele et al., MDM4 is a key therapeutic target in cutaneous melanoma, Nat. Med, vol.18, pp.1239-1247, 2012.

A. Genescà, M. Martín, L. Latre, D. Soler, J. Pampalona et al., Telomere dysfunction: a new player in radiation sensitivity, BioEssays News Rev. Mol. Cell. Dev. Biol, vol.28, pp.1172-1180, 2006.

I. C. Glitza, M. A. Davies, S. Goel, D. G. Duda, L. Xu et al., Normalization of the vasculature for treatment of cancer and other diseases, Chin. Clin. Oncol, vol.3, pp.1071-1121, 2011.

V. K. Goel, A. J. Lazar, C. L. Warneke, M. S. Redston, and F. G. Haluska, Examination of mutations in BRAF, NRAS, and PTEN in primary cutaneous melanoma, J. Invest. Dermatol, vol.126, pp.154-160, 2006.

A. M. Goldstein, M. Chan, M. Harland, N. K. Hayward, F. Demenais et al., Features associated with germline CDKN2A mutations: a GenoMEL study of melanoma-prone families from three continents, J. Med. Genet, vol.44, pp.99-106, 2007.

V. G. Gorgoulis, L. F. Vassiliou, P. Karakaidos, P. Zacharatos, A. Kotsinas et al., Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions, Nature, vol.434, pp.907-913, 2005.

B. E. Gould-rothberg, M. B. Bracken, and D. L. Rimm, Tissue Biomarkers for Prognosis in Cutaneous Melanoma: A Systematic Review and Meta-analysis, JNCI J. Natl. Cancer Inst, vol.101, pp.452-474, 2009.

V. Gray-schopfer, C. Wellbrock, and R. Marais, Melanoma biology and new targeted therapy, Nature, vol.445, pp.851-857, 2007.

I. Greguric, S. R. Taylor, D. Denoyer, P. Ballantyne, P. Berghofer et al., Discovery of [18F]N-(2-(diethylamino)ethyl)-6fluoronicotinamide: a melanoma positron emission tomography imaging radiotracer with high tumor to body contrast ratio and rapid renal clearance, J. Med. Chem, vol.52, pp.5299-5302, 2009.

N. K. Haass and M. Herlyn, Normal human melanocyte homeostasis as a paradigm for understanding melanoma, J. Investig. Dermatol. Symp. Proc. Soc. Investig. Dermatol. Inc Eur. Soc. Dermatol. Res, vol.10, pp.153-163, 2005.

R. Halaban, W. Zhang, A. Bacchiocchi, E. Cheng, F. Parisi et al., PLX4032, a selective BRAF(V600E) kinase inhibitor, activates the ERK pathway and enhances cell migration and proliferation of BRAF melanoma cells, Pigment Cell Melanoma Res, vol.23, pp.190-200, 2010.

T. J. Harris and U. Tepass, Adherens junctions: from molecules to morphogenesis, Nat. Rev. Mol. Cell Biol, vol.11, pp.502-514, 2010.

O. Hashimoto, M. Shinkawa, T. Torimura, T. Nakamura, K. Selvendiran et al., Cell cycle regulation by the Wee1 inhibitor PD0166285, pyrido [2,3-d] pyimidine, in the B16 mouse melanoma cell line, BMC Cancer, vol.6, p.292, 2006.

C. Hennequin, L. Quero, F. , and V. , DNA repair and tumour radiosensitivity: focus on ATM gene, 2011.

, Bull. Cancer (Paris), vol.98, pp.239-246

K. Hoek, D. L. Rimm, K. R. Williams, H. Zhao, S. Ariyan et al., Expression profiling reveals novel pathways in the transformation of melanocytes to melanomas, Cancer Res, vol.64, pp.5270-5282, 2004.

J. H. Houtgraaf, J. Versmissen, and W. J. Van-der-giessen, A concise review of DNA damage checkpoints and repair in mammalian cells, Cardiovasc. Revascularization Med. Mol. Interv, vol.7, pp.165-172, 2006.

J. J. Hsiao and D. E. Fisher, The roles of microphthalmia-associated transcription factor and pigmentation in melanoma, Arch. Biochem. Biophys, vol.563, pp.28-34, 2014.

D. Hu, J. D. Simon, and T. Sarna, Role of Ocular Melanin in Ophthalmic Physiology and Pathology ?, Photochem. Photobiol, vol.84, pp.639-644, 2008.
DOI : 10.1111/j.1751-1097.2008.00316.x

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1751-1097.2008.00316.x/pdf

F. W. Huang, E. Hodis, M. J. Xu, G. V. Kryukov, L. Chin et al., Highly recurrent TERT promoter mutations in human melanoma, Science, vol.339, pp.957-959, 2013.
DOI : 10.1126/science.1229259

URL : http://europepmc.org/articles/pmc4423787?pdf=render

G. S. Inamdar, S. V. Madhunapantula, and G. P. Robertson, Targeting the MAPK pathway in melanoma: why some approaches succeed and other fail, Biochem. Pharmacol, vol.80, pp.624-637, 2010.
DOI : 10.1016/j.bcp.2010.04.029

URL : http://europepmc.org/articles/pmc2897908?pdf=render

M. C. Isoldi, E. A. Pereira, M. A. Visconti, and A. M. Castrucci, The role of calcium, calciumactivated K+ channels, and tyrosine/kinase in psoralen-evoked responses in human melanoma cells. Braz, J. Med. Biol. Res. Rev. Bras. Pesqui. Médicas E Biológicas Soc. Bras. Biofísica Al, vol.37, pp.559-568, 2004.

S. Ito and K. Wakamatsu, Chemistry of mixed melanogenesis-pivotal roles of dopaquinone, Photochem. Photobiol, vol.84, pp.582-592, 2008.
DOI : 10.1111/j.1751-1097.2007.00238.x

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1751-1097.2007.00238.x/pdf

R. K. Jain, Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy, Science, vol.307, pp.58-62, 2005.

T. Jandl, E. Revskaya, Z. Jiang, M. Harris, O. Dorokhova et al., Melanoma stem cells in experimental melanoma are killed by radioimmunotherapy, Nucl. Med. Biol, vol.40, pp.177-181, 2013.
DOI : 10.1016/j.nucmedbio.2012.10.006

A. R. Jazirehi, P. B. Wenn, D. , and M. , Therapeutic implications of targeting the PI3Kinase/AKT/mTOR signaling module in melanoma therapy, Am. J. Cancer Res, vol.2, pp.178-191, 2012.

P. A. Jeggo and M. Löbrich, Contribution of DNA repair and cell cycle checkpoint arrest to the maintenance of genomic stability, DNA Repair, vol.5, pp.1192-1198, 2006.

L. Jennings, M. , and G. M. , Predicting outcome in melanoma: where are we now?, Br. J. Dermatol, vol.161, pp.496-503, 2009.
DOI : 10.1111/j.1365-2133.2009.09324.x

Y. Jiao, X. Feng, Y. Zhan, R. Wang, S. Zheng et al., Matrix metalloproteinase2 promotes ?v?3 integrin-mediated adhesion and migration of human melanoma cells by cleaving fibronectin, PloS One, vol.7, p.41591, 2012.
DOI : 10.1371/journal.pone.0041591

URL : https://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0041591&type=printable

J. L. Joyal, J. A. Barrett, J. C. Marquis, J. Chen, S. M. Hillier et al., Preclinical evaluation of an 131I-labeled benzamide for targeted radiotherapy of metastatic melanoma, Cancer Res, vol.70, pp.4045-4053, 2010.

A. I. Kassis, Therapeutic radionuclides: biophysical and radiobiologic principles, Semin. Nucl. Med, vol.38, pp.358-366, 2008.
DOI : 10.1053/j.semnuclmed.2008.05.002

URL : http://europepmc.org/articles/pmc2584872?pdf=render

A. I. Kassis, Molecular and cellular radiobiological effects of Auger emitting radionuclides, Radiat. Prot. Dosimetry, vol.143, pp.241-247, 2011.
DOI : 10.1093/rpd/ncq385

URL : http://europepmc.org/articles/pmc3108272?pdf=render

A. I. Kassis and S. J. Adelstein, Radiobiologic principles in radionuclide therapy, J. Nucl. Med. Off. Publ. Soc. Nucl. Med, vol.46, issue.1, pp.4-12, 2005.

M. K. Khan, N. Khan, A. Almasan, and R. Macklis, Future of radiation therapy for malignant melanoma in an era of newer, more effective biological agents, OncoTargets Ther, vol.4, pp.137-148, 2011.

M. Klein, M. Lotem, T. Peretz, S. T. Zwas, S. Mizrachi et al., Safety and efficacy of 188-rhenium-labeled antibody to melanin in patients with metastatic melanoma, J. Skin Cancer, p.828329, 2013.

W. M. Klein, B. P. Wu, S. Zhao, H. Wu, A. J. Klein-szanto et al., Increased expression of stem cell markers in malignant melanoma, Mod. Pathol, vol.20, pp.102-107, 2006.

P. Koefinger, C. Wels, S. Joshi, S. Damm, E. Steinbauer et al., The cadherin switch in melanoma instigated by HGF is mediated through epithelial-mesenchymal transition regulators, Pigment Cell Melanoma Res, vol.24, pp.382-385, 2011.

B. Kreiseder, L. Orel, C. Bujnow, S. Buschek, M. Pflueger et al., ?-Catulin downregulates E-cadherin and promotes melanoma progression and invasion, Int. J. Cancer J. Int. Cancer, vol.132, pp.521-530, 2013.

A. Kunwar, B. Adhikary, S. Jayakumar, A. Barik, S. Chattopadhyay et al., Melanin, a promising radioprotector: mechanisms of actions in a mice model, Toxicol. Appl. Pharmacol, vol.264, pp.202-211, 2012.

S. Kuphal and A. K. Bosserhoff, E-cadherin cell-cell communication in melanogenesis and during development of malignant melanoma, Arch. Biochem. Biophys, vol.524, pp.43-47, 2012.

P. Labarre, J. Papon, M. Moreau, N. Moins, M. Bayle et al., Melanin affinity of N-(2-diethylaminoethyl)-4-iodobenzamide, an effective melanoma imaging agent, Melanoma Res, vol.12, pp.115-121, 2002.

M. G. Lampugnani, Endothelial Cell-to-Cell Junctions: Adhesion and Signaling in Physiology and Pathology. Cold Spring Harb, Perspect. Med, vol.2, p.6528, 2012.

D. Lang, J. B. Mascarenhas, and C. R. Shea, Melanocytes, melanocyte stem cells, and melanoma stem cells, Clin. Dermatol, vol.31, pp.166-178, 2013.

J. Lang, X. Lan, Y. Liu, X. Jin, T. Wu et al., Targeting cancer stem cells with an 131I-labeled anti-AC133 monoclonal antibody in human colorectal cancer xenografts, Nucl. Med. Biol, vol.42, pp.505-512, 2015.

L. Larue, F. De-vuyst, and V. Delmas, Modeling melanoblast development, Cell Mol Life Sci, vol.70, pp.1067-1079, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00682405

K. S. Lau and K. M. Haigis, Non-redundancy within the RAS oncogene family: insights into mutational disparities in cancer, Mol. Cells, vol.28, pp.315-320, 2009.

K. Lauber, A. Ernst, M. Orth, M. Herrmann, and C. Belka, Dying cell clearance and its impact on the outcome of tumor radiotherapy, Front. Oncol, issue.2, p.116, 2012.

V. Leblanc, M. , and P. , Activation et modifications post-traductionnelles de p53 après dommage de l'ADN. Médecine/sciences 18, pp.577-584, 2002.

W. Lederle, B. Hartenstein, A. Meides, H. Kunzelmann, Z. Werb et al., MMP13 as a stromal mediator in controlling persistent angiogenesis in skin carcinoma, Carcinogenesis, vol.31, pp.1175-1184, 2010.

S. P. Lees-miller and K. Meek, Repair of DNA double strand breaks by non-homologous end joining, Biochimie, vol.85, pp.1161-1173, 2003.

S. M. De-leeuw, N. P. Smit, M. Van-veldhoven, E. M. Pennings, S. Pavel et al., Melanin content of cultured human melanocytes and UV-induced cytotoxicity, J. Photochem. Photobiol. B, vol.61, pp.106-113, 2001.

C. Letourneau, B. Dreno, Y. Kirova, J. J. Grob, T. Jouary et al., First-in-human phase I study of the DNA repair inhibitor DT01 in combination with radiotherapy in patients with in transit melanoma, J. Clin. Oncol, 2015.

X. Liu, T. Q. Pham, P. Berghofer, J. Chapman, I. Greguric et al., Synthesis and evaluation of novel radioiodinated nicotinamides for malignant melanoma, Nucl. Med. Biol, vol.35, pp.769-781, 2008.

C. J. Lord and A. Ashworth, The DNA damage response and cancer therapy, Nature, vol.481, pp.287-294, 2012.

H. K. Lorenzo and S. A. Susin, Therapeutic potential of AIF-mediated caspase-independent programmed cell death, Drug Resist. Updat, vol.10, pp.235-255, 2007.

M. Lu, P. Miller, L. , and X. , Restoring the tumour suppressive function of p53 as a parallel strategy in melanoma therapy, FEBS Lett, vol.588, pp.2616-2621, 2014.

F. Lux, A. Mignot, P. Mowat, C. Louis, S. Dufort et al., Ultrasmall rigid particles as multimodal probes for medical applications, Angew. Chem. Int. Ed Engl, vol.50, pp.12299-12303, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00673713

J. Ma, J. Y. Lin, A. Alloo, B. J. Wilson, T. Schatton et al., Isolation of tumorigenic circulating melanoma cells, Biochem. Biophys. Res. Commun, vol.402, pp.711-717, 2010.

A. Mamalis, M. Garcha, J. , and J. , Targeting the PD-1 pathway: a promising future for the treatment of melanoma, Arch. Dermatol. Res, 2014.

R. Massoumi, S. Kuphal, C. Hellerbrand, B. Haas, P. Wild et al., Down-regulation of CYLD expression by Snail promotes tumor progression in malignant melanoma, J. Exp. Med, vol.206, pp.221-232, 2009.

T. E. Matthews, I. R. Piletic, M. A. Selim, M. J. Simpson, W. et al., Pump-probe imaging differentiates melanoma from melanocytic nevi, Sci. Transl. Med, vol.3, pp.71-86, 2011.

G. A. Mcarthur, I. Puzanov, R. Amaravadi, A. Ribas, P. Chapman et al., Marked, homogeneous, and early [18F]fluorodeoxyglucose-positron emission tomography responses to vemurafenib in BRAFmutant advanced melanoma, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.30, pp.1628-1634, 2012.

E. Mclafferty, C. Hendry, A. , and F. , The integumentary system: anatomy, physiology and function of skin, Nurs. Stand. R. Coll. Nurs. G. B, vol.27, pp.35-42, 1987.

A. Mehta and J. E. Haber, Sources of DNA double-strand breaks and models of recombinational DNA repair, Cold Spring Harb. Perspect. Biol, vol.6, 2014.

A. Mesbahi, A review on gold nanoparticles radiosensitization effect in radiation therapy of cancer, Rep. Pract. Oncol. Radiother. J. Gt. Cancer Cent. Pozn. Pol. Soc. Radiat. Oncol, vol.15, pp.176-180, 2010.

Y. Miao, Q. , and T. P. , Peptide-targeted radionuclide therapy for melanoma, Crit. Rev. Oncol. Hematol, vol.67, pp.213-228, 2008.

Y. Miao, D. Whitener, W. Feng, N. K. Owen, J. Chen et al., Evaluation of the human melanoma targeting properties of radiolabeled alpha-melanocyte stimulating hormone peptide analogues, Bioconjug. Chem, vol.14, pp.1177-1184, 2003.

M. R. Middleton, J. J. Grob, N. Aaronson, G. Fierlbeck, W. Tilgen et al., Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.18, pp.158-166, 2000.

W. Mier, C. Kratochwil, J. C. Hassel, F. L. Giesel, B. Beijer et al., Radiopharmaceutical therapy of patients with metastasized melanoma with the melanin-binding benzamide 131I-BA52, J Nucl Med, vol.55, pp.9-14, 2014.

I. Miladi, G. L. Duc, D. Kryza, A. Berniard, P. Mowat et al., Biodistribution of ultra small gadolinium-based nanoparticles as theranostic agent: application to brain tumors, J. Biomater. Appl, vol.28, pp.385-394, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01020860

I. Miladi, M. Aloy, E. Armandy, P. Mowat, D. Kryza et al., Combining ultrasmall gadolinium-based nanoparticles with photon irradiation overcomes radioresistance of head and neck squamous cell carcinoma, Nanomedicine Nanotechnol. Biol. Med, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01053787

D. E. Milenic, E. D. Brady, and M. W. Brechbiel, Antibody-targeted radiation cancer therapy, Nat. Rev. Drug Discov, vol.3, pp.488-499, 2004.

A. J. Miller and M. C. Mihm, N. Engl. J. Med, vol.355, pp.51-65, 2006.

J. Mo, B. Sun, X. Zhao, Q. Gu, X. Dong et al., The in-vitro spheroid culture induces a more highly differentiated but tumorigenic population from melanoma cell lines, Melanoma Res, vol.23, pp.254-263, 2013.

N. Moins, M. D'incan, J. Bonafous, F. Bacin, P. Labarre et al., 123I-N-(2-diethylaminoethyl)-2-iodobenzamide: a potential imaging agent for cutaneous melanoma staging, Eur. J. Nucl. Med. Mol. Imaging, vol.29, pp.1478-1484, 2002.

E. Monzani, F. Facchetti, E. Galmozzi, E. Corsini, A. Benetti et al., Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential, Eur. J. Cancer, vol.43, pp.935-946, 2007.

N. Moro, C. Mauch, and P. Zigrino, Metalloproteinases in melanoma, Eur. J. Cell Biol, vol.93, pp.23-29, 2014.

R. Morrison, S. M. Schleicher, Y. Sun, K. J. Niermann, S. Kim et al., Targeting the Mechanisms of Resistance to Chemotherapy and Radiotherapy with the Cancer Stem Cell Hypothesis, J. Oncol, 2011.

P. Mowat, A. Mignot, W. Rima, F. Lux, O. Tillement et al., In vitro radiosensitizing effects of ultrasmall gadolinium based particles on tumour cells, J. Nanosci. Nanotechnol, vol.11, pp.7833-7839, 2011.

G. F. Murphy, B. J. Wilson, S. D. Girouard, N. Y. Frank, and M. H. Frank, Stem cells and targeted approaches to melanoma cure, Mol. Aspects Med, vol.39, pp.33-49, 2014.

V. Muthusamy, C. Hobbs, C. Nogueira, C. Cordon-cardo, P. H. Mckee et al., Amplification of CDK4 and MDM2 in malignant melanoma, Genes. Chromosomes Cancer, vol.45, pp.447-454, 2006.

S. Nag, J. Qin, K. S. Srivenugopal, M. Wang, and R. Zhang, The MDM2-p53 pathway revisited, J. Biomed. Res, vol.27, pp.254-271, 2013.

T. H. Nasti and L. Timares, MC1R, eumelanin and pheomelanin: their role in determining the susceptibility to skin cancer, Photochem. Photobiol, vol.91, pp.188-200, 2015.

I. Navarro-teulon, C. Lozza, A. Pèlegrin, E. Vivès, and J. Pouget, General overview of radioimmunotherapy of solid tumors, Immunotherapy, vol.5, pp.467-487, 2013.

A. A. Nelson and H. Tsao, Melanoma and genetics, Clin. Dermatol, vol.27, pp.46-52, 2009.

N. Ohtani, K. Yamakoshi, A. Takahashi, and E. Hara, The p16INK4a-RB pathway: molecular link between cellular senescence and tumor suppression, J. Med. Investig. JMI, vol.51, pp.146-153, 2004.

M. Olivier, M. Hollstein, and P. Hainaut, TP53 mutations in human cancers: origins, consequences, and clinical use, Cold Spring Harb. Perspect. Biol, vol.2, 1008.

M. Orth, K. Lauber, M. Niyazi, A. A. Friedl, M. Li et al., Current concepts in clinical radiation oncology, Radiat. Environ. Biophys, vol.53, pp.1-29, 2014.

H. Otto, P. A. Reche, F. Bazan, K. Dittmar, F. Haag et al., In silico characterization of the family of PARP-like poly(ADP-ribosyl)transferases (pARTs), BMC Genomics, vol.6, p.139, 2005.

L. Ouyang, Z. Shi, S. Zhao, F. Wang, T. Zhou et al., Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis, Cell Prolif, vol.45, pp.487-498, 2012.

A. M. Oza, D. Cibula, A. O. Benzaquen, C. Poole, R. H. Mathijssen et al., Olaparib combined with chemotherapy for recurrent platinum-sensitive ovarian cancer: a randomised phase 2 trial, Lancet Oncol, vol.16, pp.87-97, 2015.

A. Page-mccaw, A. J. Ewald, and Z. Werb, Matrix metalloproteinases and the regulation of tissue remodelling, Nat. Rev. Mol. Cell Biol, vol.8, pp.221-233, 2007.

L. Panzella, L. Leone, G. Greco, G. Vitiello, G. D'errico et al., Red human hair pheomelanin is a potent pro-oxidant mediating UV-independent contributory mechanisms of melanomagenesis, Pigment Cell Melanoma Res, vol.27, pp.244-252, 2014.

H. Y. Park, M. Kosmadaki, M. Yaar, and B. A. Gilchrest, Cellular mechanisms regulating human melanogenesis, Cell. Mol. Life Sci, vol.66, pp.1493-1506, 2009.

T. M. Pawlik and K. Keyomarsi, Role of cell cycle in mediating sensitivity to radiotherapy, Int. J. Radiat. Oncol. Biol. Phys, vol.59, pp.928-942, 2004.

Y. Perrot, F. Degoul, P. Auzeloux, M. Bonnet, F. Cachin et al., Internal dosimetry through GATE simulations of preclinical radiotherapy using a melanin-targeting ligand, Phys. Med. Biol, vol.59, pp.2183-2198, 2014.
DOI : 10.1088/0031-9155/59/9/2183

A. Pinc, R. Somasundaram, C. Wagner, M. Hörmann, G. Karanikas et al., Targeting CD20 in Melanoma Patients at High Risk of Disease Recurrence, Mol. Ther, vol.20, pp.1056-1062, 2012.
DOI : 10.1038/mt.2012.27

URL : https://doi.org/10.1038/mt.2012.27

L. Portt, G. Norman, C. Clapp, M. Greenwood, and M. T. Greenwood, Anti-apoptosis and cell survival: A review, Biochim. Biophys. Acta BBA-Mol. Cell Res, vol.1813, pp.238-259, 2011.
DOI : 10.1016/j.bbamcr.2010.10.010

URL : https://doi.org/10.1016/j.bbamcr.2010.10.010

G. Poste, J. Doll, I. R. Hart, and I. J. Fidler, In vitro selection of murine B16 melanoma variants with enhanced tissue-invasive properties, Cancer Res, vol.40, pp.1636-1644, 1980.

M. Potente, H. Gerhardt, C. , and P. , Basic and therapeutic aspects of angiogenesis, Cell, vol.146, pp.873-887, 2011.
DOI : 10.1016/j.cell.2011.08.039

URL : https://doi.org/10.1016/j.cell.2011.08.039

J. Pouget and S. J. Mather, General aspects of the cellular response to low-and high-LET radiation, Eur. J. Nucl. Med, vol.28, pp.541-561, 2014.
DOI : 10.1007/s002590100484

J. Pouget, C. Lozza, E. Deshayes, V. Boudousq, and I. Navarro-teulon, Introduction to radiobiology of targeted radionuclide therapy, Front. Med, vol.2, p.12, 2015.
DOI : 10.3389/fmed.2015.00012

URL : https://www.frontiersin.org/articles/10.3389/fmed.2015.00012/pdf

M. A. Price, L. E. Colvin-wanshura, J. Yang, J. Carlson, B. Xiang et al., CSPG4, a potential therapeutic target, facilitates malignant progression of melanoma, Pigment Cell Melanoma Res, vol.24, pp.1148-1157, 2011.
DOI : 10.1111/j.1755-148x.2011.00929.x

URL : http://europepmc.org/articles/pmc3426219?pdf=render

M. Quanz, N. Berthault, C. Roulin, M. Roy, A. Herbette et al., Small-molecule drugs mimicking DNA damage: a new strategy for sensitizing tumors to radiotherapy, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.15, pp.1308-1316, 2009.
DOI : 10.1158/1078-0432.ccr-08-2108

URL : http://clincancerres.aacrjournals.org/content/clincanres/15/4/1308.full.pdf

M. Quanz, D. Chassoux, N. Berthault, C. Agrario, J. Sun et al., Hyperactivation of DNA-PK by double-strand break mimicking molecules disorganizes DNA damage response, PloS One, vol.4, p.6298, 2009.
DOI : 10.1371/journal.pone.0006298

URL : https://doi.org/10.1371/journal.pone.0006298

E. Quintana, M. Shackleton, M. S. Sabel, D. R. Fullen, T. M. Johnson et al., Efficient tumour formation by single human melanoma cells, Nature, vol.456, pp.593-598, 2008.
DOI : 10.1038/nature07567

URL : http://europepmc.org/articles/pmc2597380?pdf=render

C. Raja, P. Graham, S. M. Abbas-rizvi, E. Song, H. Goldsmith et al., Interim analysis of toxicity and response in phase 1 trial of systemic targeted alpha therapy for metastatic melanoma, Cancer Biol. Ther, vol.6, pp.846-852, 2007.

G. Raposo and M. S. Marks, Melanosomes-dark organelles enlighten endosomal membrane transport, Nat. Rev. Mol. Cell Biol, vol.8, pp.786-797, 2007.
DOI : 10.1038/nrm2258

URL : http://europepmc.org/articles/pmc2786984?pdf=render

G. Rappa, O. Fodstad, and A. Lorico, The Stem Cell-Associated Antigen CD133 (Prominin-1) Is a Molecular Therapeutic Target for Metastatic Melanoma, Stem Cells Dayt. Ohio, vol.26, pp.3008-3017, 2008.
DOI : 10.1634/stemcells.2008-0601

URL : http://onlinelibrary.wiley.com/doi/10.1634/stemcells.2008-0601/pdf

E. Rass, A. Grabarz, P. Bertrand, and B. Lopez, Double strand break repair, one mechanism can hide another: alternative non-homologous end joining, Cancer Radiothérapie J. Société Fr. Radiothérapie Oncol, vol.16, pp.1-10, 2012.

T. Redmer, Y. Welte, D. Behrens, I. Fichtner, D. Przybilla et al., The nerve growth factor receptor CD271 is crucial to maintain tumorigenicity and stem-like properties of melanoma cells, PloS One, vol.9, 2014.

S. M. Rizvi, C. F. Qu, Y. J. Song, C. Raja, A. et al., vivo studies of pharmacokinetics and efficacy of Bismuth-213 labeled antimelanoma monoclonal antibody 9, vol.4, pp.763-768, 2005.
DOI : 10.4161/cbt.4.7.1868

URL : http://www.tandfonline.com/doi/pdf/10.4161/cbt.4.7.1868?needAccess=true

F. Rondepierre, B. Bouchon, J. Papon, M. Bonnet-duquennoy, R. Kintossou et al., Proteomic studies of B16 lines: involvement of annexin A1 in melanoma dissemination, Biochim. Biophys. Acta, vol.1794, pp.61-69, 2009.

I. B. Roninson, E. V. Broude, C. , and B. , If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells, Drug Resist. Updat, vol.4, pp.303-313, 2001.
DOI : 10.1054/drup.2001.0213

W. P. Roos, S. Quiros, A. Krumm, S. Merz, O. J. Switzeny et al., B-Raf inhibitor vemurafenib in combination with temozolomide and fotemustine in the killing response of malignant melanoma cells, Oncotarget, vol.5, pp.12607-12620, 2014.

M. Rooseboom, J. N. Commandeur, and N. P. Vermeulen, Enzyme-catalyzed activation of anticancer prodrugs, Pharmacol. Rev, vol.56, pp.53-102, 2004.

J. Rother, J. , and D. , Molecular markers of tumor progression in melanoma, Curr. Genomics, vol.10, pp.231-239, 2009.

A. Rotte, M. Martinka, L. , and G. , MMP2 expression is a prognostic marker for primary melanoma patients, Cell. Oncol. Dordr, vol.35, pp.207-216, 2012.

E. Sala, L. Mologni, S. Truffa, C. Gaetano, G. E. Bollag et al., BRAF silencing by short hairpin RNA or chemical blockade by PLX4032 leads to different responses in melanoma and thyroid carcinoma cells, Mol. Cancer Res. MCR, vol.6, pp.751-759, 2008.

M. J. Sambade, E. C. Peters, N. E. Thomas, W. K. Kaufmann, R. J. Kimple et al., Melanoma cells show a heterogeneous range of sensitivity to ionizing radiation and are radiosensitized by inhibition of B-RAF with PLX-4032, Radiother Oncol, vol.98, pp.394-399, 2011.

B. K. Samulitis, R. T. Dorr, and H. S. Chow, Interaction of dacarbazine and imexon, in vitro and in vivo, in human A375 melanoma cells, Anticancer Res, vol.31, pp.2781-2785, 2011.

M. Sarna, A. Zadlo, P. Hermanowicz, Z. Madeja, K. Burda et al., Cell elasticity is an important indicator of the metastatic phenotype of melanoma cells, Exp. Dermatol, vol.23, pp.813-818, 2014.

K. Satyamoorthy, N. H. Chehab, M. J. Waterman, M. C. Lien, W. S. El-deiry et al., Aberrant regulation and function of wild-type p53 in radioresistant melanoma cells, Cell Growth Differ. Mol. Biol. J. Am. Assoc. Cancer Res, vol.11, pp.467-474, 2000.

D. Schadendorf, S. M. Algarra, L. Bastholt, G. Cinat, B. Dreno et al., Immunotherapy of distant metastatic disease, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. ESMO, vol.20, pp.41-50, 2009.

T. Schatton, G. F. Murphy, N. Y. Frank, K. Yamaura, A. M. Waaga-gasser et al., Identification of cells initiating human melanomas, Nature, vol.451, pp.345-349, 2008.

A. Schlegel, C. Buhler, F. Devun, C. Agrario, S. Urien et al., Pharmacokinetics and Toxicity in Rats and Monkeys of coDbait: A Therapeutic Doublestranded DNA Oligonucleotide Conjugated to Cholesterol, Mol. Ther. Nucleic Acids, vol.1, p.33, 2012.

P. Schmidt, C. Kopecky, A. Hombach, P. Zigrino, C. Mauch et al., Eradication of melanomas by targeted elimination of a minor subset of tumor cells, Proc. Natl. Acad. Sci, vol.108, pp.2474-2479, 2011.

E. Selinheimo, D. Nieidhin, C. Steffensen, J. Nielsen, A. Lomascolo et al., Comparison of the characteristics of fungal and plant tyrosinases, J. Biotechnol, vol.130, pp.471-480, 2007.

G. L. Semenza, Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics, Oncogene, vol.29, pp.625-634, 2010.

O. Shakhova and L. Sommer, Testing the cancer stem cell hypothesis in melanoma: the clinics will tell, Cancer Lett, vol.338, pp.74-81, 2013.

J. D. Simon, D. Peles, K. Wakamatsu, and S. Ito, Current challenges in understanding melanogenesis: bridging chemistry, biological control, morphology, and function, Pigment Cell Melanoma Res, vol.22, pp.563-579, 2009.

J. M. Stahl, A. Sharma, M. Cheung, M. Zimmerman, J. Q. Cheng et al., Deregulated Akt3 activity promotes development of malignant melanoma, Cancer Res, vol.64, pp.7002-7010, 2004.

L. S. Steelman, W. H. Chappell, S. L. Abrams, R. C. Kempf, J. Long et al., Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapyimplications for cancer and aging, Aging, vol.3, pp.192-222, 2011.

R. A. Sturm, A golden age of human pigmentation genetics, Trends Genet. TIG, vol.22, pp.464-468, 2006.

A. Takaoka, S. Hayakawa, H. Yanai, D. Stoiber, H. Negishi et al., Integration of interferon-?/? signalling to p53 responses in tumour suppression and antiviral defence, Nature, vol.424, pp.516-523, 2003.

A. A. Tarhini and S. S. Agarwala, Cutaneous melanoma: available therapy for metastatic disease, Dermatol. Ther, vol.19, pp.19-25, 2006.
DOI : 10.1111/j.1529-8019.2005.00052.x

A. Testori, P. Rutkowski, J. Marsden, L. Bastholt, V. Chiarion-sileni et al., Surgery and radiotherapy in the treatment of cutaneous melanoma, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. ESMO, vol.20, pp.22-29, 2009.

L. H. Thompson, Recognition, signaling, and repair of DNA double-strand breaks produced by ionizing radiation in mammalian cells: the molecular choreography, Mutat. Res, vol.751, pp.158-246, 2012.

J. F. Thompson, R. A. Scolyer, and R. F. Kefford, Cutaneous melanoma, Lancet, vol.365, pp.687-701, 2005.

D. J. Tobin, Human hair pigmentation-biological aspects, Int. J. Cosmet. Sci, vol.30, pp.233-257, 2008.
DOI : 10.1111/j.1468-2494.2008.00456.x

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1468-2494.2008.00456.x/pdf

H. Toshimitsu, Y. Yoshimoto, C. K. Augustine, J. C. Padussis, J. S. Yoo et al., Inhibition of poly(ADP-ribose) polymerase enhances the effect of chemotherapy in an animal model of regional therapy for the treatment of advanced extremity malignant melanoma, Ann. Surg. Oncol, vol.17, pp.2247-2254, 2010.

H. Tsao, J. M. Olazagasti, K. M. Cordoro, J. D. Brewer, S. C. Taylor et al., Early detection of melanoma: Reviewing the ABCDEs, J. Am. Acad. Dermatol, vol.72, pp.717-723, 2015.
DOI : 10.1016/j.jaad.2015.01.025

P. Vandenabeele, L. Galluzzi, T. Vanden-berghe, and G. Kroemer, Molecular mechanisms of necroptosis: an ordered cellular explosion, Nat. Rev. Mol. Cell Biol, vol.11, pp.700-714, 2010.
DOI : 10.1038/nrm2970

L. T. Vassilev, B. T. Vu, B. Graves, D. Carvajal, F. Podlaski et al., Vivo Activation of the p53 Pathway by Small-Molecule Antagonists of MDM2, vol.303, pp.844-848, 2004.

J. Vera, Y. Raatz, O. Wolkenhauer, T. Kottek, A. Bhattacharya et al., Chk1 and Wee1 control genotoxic-stress induced G2-M arrest in melanoma cells, Cell. Signal, vol.27, pp.951-960, 2015.
DOI : 10.1016/j.cellsig.2015.01.020

K. Vermeulen, D. R. Van-bockstaele, and Z. N. Berneman, The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer, Cell Prolif, vol.36, pp.131-149, 2003.

C. Viallard, Y. Perrot, Z. Boudhraa, E. Jouberton, E. Miot-noirault et al., 123 I]ICF01012 melanoma imaging and [ 131 I]ICF01012 dosimetry allow adapted internal targeted radiotherapy in preclinical melanoma models, Eur. J. Dermatol. EJD, vol.25, pp.29-35, 2015.

J. F. Viallard, F. Lacombe, F. Belloc, J. L. Pellegrin, R. et al., Cancer Radiothérapie J. Société Fr. Radiothérapie Oncol, vol.5, pp.109-129, 2001.

I. F. Videira, S. Moura, D. F. Magina, and S. , Mechanisms regulating melanogenesis, An. Bras. Dermatol, vol.88, pp.76-83, 2013.

D. S. Vinay, E. P. Ryan, G. Pawelec, W. H. Talib, J. Stagg et al., Immune evasion in cancer: Mechanistic basis and therapeutic strategies, Semin. Cancer Biol, 2015.

K. Wakamatsu and S. Ito, Advanced chemical methods in melanin determination, Pigment Cell Res. Spons. Eur. Soc. Pigment Cell Res. Int. Pigment Cell Soc, vol.15, pp.174-183, 2002.

P. T. Wan, M. J. Garnett, S. M. Roe, S. Lee, D. Niculescu-duvaz et al., Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF, Cell, vol.116, pp.855-867, 2004.

A. Wang and X. Qi, Targeting RAS/RAF/MEK/ERK signaling in metastatic melanoma, IUBMB Life, vol.65, pp.748-758, 2013.

C. Wang and S. P. Lees-miller, Detection and repair of ionizing radiation-induced DNA double strand breaks: new developments in nonhomologous end joining, Int. J. Radiat. Oncol. Biol. Phys, vol.86, pp.440-449, 2013.

K. A. Ward, D. Lazovich, and M. K. Hordinsky, Germline melanoma susceptibility and prognostic genes: a review of the literature, J. Am. Acad. Dermatol, vol.67, pp.1055-1067, 2012.

C. Wellbrock, MAPK pathway inhibition in melanoma: resistance three ways, Biochem. Soc. Trans, vol.42, pp.727-732, 2014.

C. Wellbrock, M. Karasarides, and R. Marais, The RAF proteins take centre stage, Nat. Rev. Mol. Cell Biol, vol.5, pp.875-885, 2004.

C. Wels, S. Joshi, P. Koefinger, H. Bergler, and H. Schaider, Transcriptional activation of ZEB1 by Slug leads to cooperative regulation of the epithelial-mesenchymal transition-like phenotype in melanoma, J. Invest. Dermatol, vol.131, pp.1877-1885, 2011.

L. Van-der-weyden, E. E. Patton, G. A. Wood, A. Foote, T. Brenn et al., Cross-species models of human melanoma, J. Pathol, 2015.

A. Yabluchanskiy, Y. Ma, R. P. Iyer, M. E. Hall, L. et al., Matrix Metalloproteinase-9: Many Shades of Function in Cardiovascular Disease, Physiology, vol.28, pp.391-403, 2013.

M. Yancovitz, A. Litterman, J. Yoon, E. Ng, R. L. Shapiro et al., Intra-and Inter-Tumor Heterogeneity of BRAFV600EMutations in Primary and Metastatic Melanoma, PLoS ONE, vol.7, 2012.

T. A. Yap, S. K. Sandhu, C. P. Carden, and J. S. Bono, Poly(ADP-ribose) polymerase (PARP) inhibitors: Exploiting a synthetic lethal strategy in the clinic, CA. Cancer J. Clin, vol.61, pp.31-49, 2011.

B. F. Zamarron, C. , and W. , Dual Roles of Immune Cells and Their Factors in Cancer Development and Progression, Int. J. Biol. Sci, vol.7, pp.651-658, 2011.

P. Zigrino, I. Kuhn, T. Bäuerle, J. Zamek, J. W. Fox et al., Stromal expression of MMP-13 is required for melanoma invasion and metastasis, J. Invest. Dermatol, vol.129, pp.2686-2693, 2009.

L. Zimmer, J. Vaubel, E. Livingstone, and D. Schadendorf, Side effects of systemic oncological therapies in dermatology, J. Dtsch. Dermatol. Ges. J. Ger. Soc. Dermatol. JDDG, vol.10, pp.475-486, 2012.

A. Zygogianni, G. Kyrgias, J. Kouvaris, K. Mystakidou, H. Gogas et al., Melanoma: the radiotherapeutic point of view; review of the current literature, Rev. Recent Clin. Trials, vol.6, pp.127-133, 2011.