J. L. Orgaz and V. Sanz-moreno, Emerging molecular targets in melanoma invasion and metastasis, Pigment Cell Melanoma Res, vol.26, pp.39-57, 2013.

H. A. Tawbi and S. C. Buch, Chemotherapy resistance abrogation in metastatic melanoma, Clinical advances in hematology & oncology : H&O 2010, vol.8, pp.259-266

Y. Cheli, S. Giuliano, T. Botton, S. Rocchi, V. Hofman 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.
URL : https://hal.archives-ouvertes.fr/inserm-02530763

S. Pinner, P. Jordan, K. Sharrock, L. Bazley, L. Collinson et al., Intravital imaging reveals transient changes in pigment production and Brn2 expression during metastatic melanoma dissemination, Cancer Res, vol.69, pp.7969-7977, 2009.

A. Roesch, M. Fukunaga-kalabis, E. C. Schmidt, S. E. Zabierowski, P. A. Brafford et al., A temporarily distinct subpopulation of slowcycling melanoma cells is required for continuous tumor growth, Cell, vol.141, pp.583-594, 2010.

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.

Y. Cheli, S. Giuliano, N. Fenouille, A. M. Hofman, V. Hofman et al., Hypoxia and MITF control metastatic behaviour in mouse and human melanoma cells, Oncogene, vol.31, pp.2461-2470, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-02530643

K. Harbst, J. Staaf, M. Lauss, A. Karlsson, A. Masback et al., Molecular profiling reveals low-and high-grade forms of primary melanoma, Clin Cancer Res, vol.18, pp.4026-4036, 2012.

K. S. Hoek, O. M. Eichhoff, N. C. Schlegel, U. Dobbeling, N. Kobert et al., In vivo switching of human melanoma cells between proliferative and invasive states, Cancer Res, vol.68, pp.650-656, 2008.

E. Steingrimsson, N. G. Copeland, and N. A. Jenkins, Melanocytes and the microphthalmia transcription factor network, Annu Rev Genet, vol.38, pp.365-411, 2004.

S. Giuliano, Y. Cheli, M. Ohanna, C. Bonet, L. Beuret et al., Microphthalmia-associated transcription factor controls the DNA damage response and a lineage-specific senescence program in melanomas, Cancer Res, vol.70, pp.3813-3822, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-02530789

T. Strub, S. Giuliano, T. Ye, C. Bonet, C. Keime et al., Essential role of microphthalmia transcription factor for DNA replication, mitosis and genomic stability in melanoma, Oncogene, vol.30, pp.2319-2332, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02530755

N. M. Mhaidat, X. D. Zhang, J. Allen, K. A. Avery-kiejda, R. J. Scott et al., Temozolomide induces senescence but not apoptosis in human melanoma cells, Br J Cancer, vol.97, pp.1225-1233, 2007.

J. P. Coppe, P. Y. Desprez, A. Krtolica, and J. Campisi, The senescence-associated secretory phenotype: the dark side of tumor suppression, Annual review of pathology, vol.5, pp.99-118, 2010.

T. Kuilman and D. S. Peeper, Senescence-messaging secretome: SMS-ing cellular stress, Nat Rev Cancer, vol.9, pp.81-94, 2009.

M. Ohanna, S. Giuliano, C. Bonet, V. Imbert, V. Hofman et al., Senescent cells develop a PARP-1 and nuclear factor-{kappa}B-associated secretome, Genes Dev www.impactjournals.com/oncotarget, vol.25, pp.1245-1261, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02530715

T. Kuilman, C. Michaloglou, L. C. Vredeveld, S. Douma, R. Van-doorn et al., Oncogene-induced senescence relayed by an interleukindependent inflammatory network, Cell, vol.133, pp.1019-1031, 2008.

J. C. Acosta, A. O'loghlen, A. Banito, M. V. Guijarro, A. Augert et al., Chemokine signaling via the CXCR2 receptor reinforces senescence, vol.133, pp.1006-1018, 2008.

C. Canino, F. Mori, A. Cambria, A. Diamantini, S. Germoni et al., SASP mediates chemoresistance and tumorinitiating-activity of mesothelioma cells, vol.31, pp.3148-3163, 2012.

A. B. Lyons, J. Hasbold, and P. D. Hodgkin, Flow cytometric analysis of cell division history using dilution of carboxyfluorescein diacetate succinimidyl ester, a stably integrated fluorescent probe, Methods in cell biology, vol.63, pp.375-398, 2001.

K. S. Hoek and C. R. Goding, Cancer stem cells versus phenotypeswitching in melanoma, Pigment Cell Melanoma Res, vol.23, pp.746-759, 2010.

R. Santini, M. C. Vinci, S. Pandolfi, J. Y. Penachioni, V. Montagnani et al., Hedgehog-GLI signaling drives self-renewal and tumorigenicity of human melanomainitiating cells, Stem Cells, vol.30, pp.1808-1818, 2012.

D. Iliopoulos, H. A. Hirsch, G. Wang, and K. Struhl, Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion, Proc Natl Acad Sci U S A, vol.108, pp.1397-1402, 2011.

K. Kushiro, R. A. Chu, A. Verma, and N. P. Nunez, Adipocytes Promote B16BL6 Melanoma Cell Invasion and the Epithelial-to-Mesenchymal Transition, Cancer microenvironment : official journal of the International Cancer Microenvironment Society, vol.5, pp.73-82, 2012.

V. Von-felbert, F. Cordoba, J. Weissenberger, C. Vallan, M. Kato et al., Interleukin-6 gene ablation in a transgenic mouse model of malignant skin melanoma, Am J Pathol, vol.166, pp.831-841, 2005.

D. A. Frank, STAT3 as a central mediator of neoplastic cellular transformation, Cancer letters, vol.251, pp.199-210, 2007.

Z. S. Wu, X. W. Cheng, X. N. Wang, and N. J. Song, Prognostic significance of phosphorylated signal transducer and activator of transcription 3 and suppressor of cytokine signaling 3 expression in human cutaneous melanoma, Melanoma Res, vol.21, pp.483-490, 2011.

H. Yu, D. Pardoll, and R. Jove, STATs in cancer inflammation and immunity: a leading role for STAT3, vol.9, pp.798-809, 2009.

T. K. Lee, A. Castilho, V. C. Cheung, K. H. Tang, S. Ma et al., CD24(+) liver tumor-initiating cells drive self-renewal and tumor initiation through STAT3-mediated NANOG regulation, Cell stem cell, vol.9, pp.50-63, 2011.

L. Lin, A. Liu, Z. Peng, H. J. Lin, P. K. Li et al., STAT3 is necessary for proliferation and survival in colon cancerinitiating cells, Cancer Res, vol.71, pp.7226-7237, 2011.

C. Villalva, S. Martin-lanneree, U. Cortes, F. Dkhissi, M. Wager et al., Karayan-Tapon L: STAT3 is essential for the maintenance of neurosphere-initiating tumor cells in patients with glioblastomas: a potential for targeted therapy?, Int J Cancer, vol.128, pp.826-838, 2011.

Y. Hasegawa, N. Takahashi, A. R. Forrest, J. W. Shin, Y. Kinoshita et al., CC chemokine ligand 2 and leukemia inhibitory factor cooperatively promote pluripotency in mouse induced pluripotent cells, Stem Cells, vol.29, pp.1196-1205, 2011.

D. S. Widmer, P. F. Cheng, O. M. Eichhoff, B. C. Belloni, M. C. Zipser et al., Systematic classification of melanoma cells by phenotype-specific gene expression mapping, Pigment Cell Melanoma Res, vol.25, pp.343-353, 2012.

C. Bertolotto, F. Lesueur, S. Giuliano, T. Strub, M. De-lichy et al., A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma, vol.480, pp.94-98, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01791271

Y. Luo, K. Dallaglio, Y. Chen, W. A. Robinson, S. E. Robinson et al., ALDH1A isozymes are markers of human melanoma stem cells and potential therapeutic targets, Stem Cells, vol.30, pp.2100-2113, 2012.

L. Prasmickaite, B. O. Engesaeter, N. Skrbo, T. Hellenes, K. A. Oliver et al., Aldehyde dehydrogenase (ALDH) activity does not select for cells with enhanced aggressive properties in malignant www.impactjournals.com/oncotarget melanoma, PloS one, vol.5, p.10731, 2010.

F. Brellier, K. Hostettler, H. R. Hotz, C. Ozcakir, S. A. Cologlu et al., Chiquet-Ehrismann R: Tenascin-C triggers fibrin accumulation by downregulation of tissue plasminogen activator, FEBS Lett, vol.585, pp.913-920, 2011.

M. Fukunaga-kalabis, G. Martinez, T. K. Nguyen, D. Kim, A. Santiago-walker et al., Tenascin-C promotes melanoma progression by maintaining the ABCB5-positive side population, Oncogene, vol.29, pp.6115-6124, 2010.

R. M. Linger, A. K. Keating, H. S. Earp, and D. K. Graham, TAM receptor tyrosine kinases: biologic functions, signaling, and potential therapeutic targeting in human cancer, Advances in cancer research, vol.100, pp.35-83, 2008.

L. A. Byers, J. Wang, M. B. Nilsson, J. Fujimoto, P. Saintigny et al., Proteomic profiling identifies dysregulated pathways in small cell lung cancer and novel therapeutic targets including PARP1, Cancer discovery, vol.2, pp.798-811, 2012.

M. Sensi, M. Catani, G. Castellano, G. Nicolini, F. Alciato et al., Human cutaneous melanomas lacking MITF and melanocyte differentiation antigens express a functional Axl receptor kinase, J Invest Dermatol, vol.131, pp.2448-2457, 2011.

Y. Tang, Y. Luo, Z. Jiang, Y. Ma, C. J. Lin et al., Jak/Stat3 signaling promotes somatic cell reprogramming by epigenetic regulation, Stem Cells, vol.30, pp.2645-2656, 2012.

L. Larribere, C. Hilmi, M. Khaled, C. Gaggioli, K. Bille et al., The cleavage of microphthalmia-associated transcription factor, MITF, by caspases plays an essential role in melanocyte and melanoma cell apoptosis, Genes Dev, vol.19, pp.1980-1985, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-02531076

C. Hilmi, L. Larribere, S. Giuliano, K. Bille, J. P. Ortonne et al., IGF1 promotes resistance to apoptosis in melanoma cells through an increased expression of BCL2, BCL-X(L), and survivin, J Invest Dermatol, vol.128, pp.1499-1505, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-02530835