S. Kelderman, T. Schumacher, and J. Haanen, Acquired and intrinsic resistance in cancer immunotherapy, Molecular Oncology, vol.348, issue.6, pp.1132-1141, 2014.
DOI : 10.1016/j.molonc.2014.07.011

C. Ng and B. Bonavida, A new challenge for successful immunotherapy by tumors that are resistant to apoptosis: Two complementary signals to overcome cross-resistance
DOI : 10.1016/S0065-230X(02)85005-9

T. Wang, M. Xiao, Y. Ge, C. Krepler, E. Belser et al., BRAF Inhibition Stimulates Melanoma-Associated Macrophages to Drive Tumor Growth, Clinical Cancer Research, vol.21, issue.7, pp.1652-64, 2015.
DOI : 10.1158/1078-0432.CCR-14-1554

D. Giorgi, V. Grazzini, M. Gandini, S. Benemei, S. Lotti et al., Treatment With ??-Blockers and Reduced Disease Progression in Patients With Thick Melanoma, Archives of Internal Medicine, vol.171, issue.8, pp.779-81, 2011.
DOI : 10.1001/archinternmed.2011.131

S. Lemeshow, H. Sørensen, G. Phillips, E. Yang, S. Antonsen et al., ??-Blockers and Survival among Danish Patients with Malignant Melanoma: A Population-Based Cohort Study, Cancer Epidemiology Biomarkers & Prevention, vol.20, issue.10, pp.2273-2282, 2011.
DOI : 10.1158/1055-9965.EPI-11-0249

D. Giorgi, V. Gandini, S. Grazzini, M. Benemei, S. Marchionni et al., Effect of ??-Blockers and Other Antihypertensive Drugs On the Risk of Melanoma Recurrence and Death, Mayo Clinic Proceedings, vol.88, issue.11, pp.1196-203, 2013.
DOI : 10.1016/j.mayocp.2013.09.001

C. Mccourt, H. Coleman, L. Murray, M. Cantwell, O. Dolan et al., Beta-blocker usage after malignant melanoma diagnosis and survival: a population-based nested case-control study, British Journal of Dermatology, vol.39, issue.4, pp.930-938, 2014.
DOI : 10.1111/bjd.12894

S. Cole and A. Sood, Molecular Pathways: Beta-Adrenergic Signaling in Cancer, Clinical Cancer Research, vol.18, issue.5, pp.1201-1207, 2012.
DOI : 10.1158/1078-0432.CCR-11-0641

M. Sanzo, R. Colucci, M. Arunachalam, S. Berti, and S. Moretti, Stress as a Possible Mechanism in Melanoma Progression, Dermatology Research and Practice, vol.158, issue.4, 2010.
DOI : 10.1016/j.bbi.2008.10.005

S. Moretti, D. Massi, V. Farini, G. Baroni, M. Parri et al., ??-adrenoceptors are upregulated in human melanoma and their activation releases pro-tumorigenic cytokines and metalloproteases in melanoma cell lines, Laboratory Investigation, vol.15, issue.3, pp.279-90, 2013.
DOI : 10.1016/j.psyneuen.2005.05.013

A. Ragan, A. Lesniak, M. Bochynska-czyz, A. Kosson, H. Szymanska et al., Chronic mild stress facilitates melanoma tumor growth in mouse lines selected for high and low stress-induced analgesia, Stress, vol.16, issue.5, pp.571-80, 2013.
DOI : 10.1007/s002130050456

M. Kobayashi, M. Jeschke, A. Asai, M. Kogiso, S. Yoshida et al., Propranolol as a modulator of M2b monocytes in severely burned patients, Journal of Leukocyte Biology, vol.89, issue.5, pp.797-803, 2011.
DOI : 10.1189/jlb.1010553

Y. Tsuda, M. Kobayashi, D. Herndon, and F. Suzuki, Impairment of the host's antibacterial resistance by norepinephrine activated neutrophils, Burns, vol.34, issue.4, pp.460-466, 2008.
DOI : 10.1016/j.burns.2007.07.004

A. Scanzano and M. Cosentino, Adrenergic regulation of innate immunity: a review, Frontiers in Pharmacology, vol.289, issue.164, p.171, 2015.
DOI : 10.1016/S0304-3940(00)01254-4

T. Takamoto, Y. Hori, Y. Koga, H. Toshima, A. Hara et al., Norepinephrine Inhibits Human Natural Killer Cell Activity in Vitro, International Journal of Neuroscience, vol.57, issue.1-2, pp.127-158, 1991.
DOI : 10.3109/00207459108987189

D. Vredevoe, D. Moser, X. Gan, and B. Bonavida, Natural Killer Cell Anergy to Cytokine Stimulants in a Subgroup of Patients with Heart Failure: Relationship to Norepinephrine, Neuroimmunomodulation, vol.2, issue.1, pp.16-24, 1995.
DOI : 10.1159/000096830

I. Elenkov and G. Chrousos, Stress Hormones, Th1/Th2 patterns, Pro/Anti-inflammatory Cytokines and Susceptibility to Disease, Trends in Endocrinology & Metabolism, vol.10, issue.9, pp.359-68, 1999.
DOI : 10.1016/S1043-2760(99)00188-5

M. Kato, M. Takahashi, A. Akhand, W. Liu, Y. Dai et al., Transgenic mouse model for skin malignant melanoma, Oncogene, vol.17, issue.14, pp.1885-1893, 1998.
DOI : 10.1038/sj.onc.1202077

J. Eyles, A. Puaux, X. Wang, B. Toh, C. Prakash et al., Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma, Journal of Clinical Investigation, vol.120, issue.6, pp.2030-2039, 2010.
DOI : 10.1172/JCI42002DS1

R. Lengagne, S. Graff-dubois, M. Garcette, L. Renia, M. Kato et al., Distinct Role for CD8 T Cells toward Cutaneous Tumors and Visceral Metastases, The Journal of Immunology, vol.180, issue.1, pp.130-137, 2008.
DOI : 10.4049/jimmunol.180.1.130

A. Pommier, A. Audemard, A. Durand, R. Lengagne, A. Delpoux et al., Inflammatory monocytes are potent antitumor effectors controlled by regulatory CD4+ T cells, Proceedings of the National Academy of Sciences, vol.110, issue.32, pp.13085-90, 2013.
DOI : 10.1073/pnas.1300314110

R. Lengagne, L. Gal, F. Garcette, M. Fiette, L. et al., Spontaneous Vitiligo in an Animal Model for Human Melanoma: Role of Tumor-specific CD8+ T Cells, Cancer Research, vol.64, issue.4, pp.1496-501, 2004.
DOI : 10.1158/0008-5472.CAN-03-2828

R. Lengagne, A. Pommier, J. Caron, L. Douguet, M. Garcette et al., T Cells Contribute to Tumor Progression by Favoring Pro-Tumoral Properties of Intra-Tumoral Myeloid Cells in a Mouse Model for Spontaneous Melanoma, PLoS ONE, vol.3, issue.5, p.20235, 2011.
DOI : 10.1371/journal.pone.0020235.t001

B. Toh, X. Wang, J. Keeble, W. Sim, K. Khoo et al., Mesenchymal Transition and Dissemination of Cancer Cells Is Driven by Myeloid-Derived Suppressor Cells Infiltrating the Primary Tumor, PLoS Biology, vol.11, issue.9, p.1001162, 2011.
DOI : 10.1371/journal.pbio.1001162.s012

K. Guo, Q. Ma, L. Wang, H. Hu, J. Li et al., Norepinephrine-induced invasion by pancreatic cancer cells is inhibited by propranolol, Oncol Rep, vol.22, pp.825-855, 2009.

X. Liao, C. X. Zhao, W. Zhang, D. Bi, T. Wang et al., The ?-adrenoceptor antagonist, propranolol, induces human gastric cancer cell apoptosis and cell cycle arrest via inhibiting nuclear factor ?B signaling, Oncol Rep, vol.24, pp.1669-76, 2010.

D. Powe, M. Voss, K. Zänker, H. Habashy, A. Green et al., Beta-Blocker Drug Therapy Reduces Secondary Cancer Formation in Breast Cancer and Improves Cancer Specific Survival, Oncotarget, vol.1, issue.7, pp.628-666, 2010.
DOI : 10.18632/oncotarget.197

T. Barron, R. Connolly, L. Sharp, K. Bennett, and K. Visvanathan, Beta Blockers and Breast Cancer Mortality: A Population- Based Study, Journal of Clinical Oncology, vol.29, issue.19, pp.2635-2679, 2011.
DOI : 10.1200/JCO.2010.33.5422

P. Chang, W. Huang, C. Lin, T. Huang, Y. Wu et al., Propranolol Reduces Cancer Risk, Medicine, vol.94, issue.27, p.1097, 2015.
DOI : 10.1097/MD.0000000000001097

URL : http://doi.org/10.1097/md.0000000000001097

W. Yuan, J. Z. Wei, J. Liu, Z. Xue, L. Wang et al., Propranolol given orally for proliferating infantile haemangiomas: analysis of efficacy and serological changes in vascular endothelial growth factor and endothelial nitric oxide synthase in 35 patients, British Journal of Oral and Maxillofacial Surgery, vol.51, issue.7, pp.656-61, 2013.
DOI : 10.1016/j.bjoms.2012.12.003

D. Gabrilovich, S. Ostrand-rosenberg, and V. Bronte, Coordinated regulation of myeloid cells by tumours, Nature Reviews Immunology, vol.205, issue.4, pp.253-68, 2012.
DOI : 10.1038/nri3175

S. Ostrand-rosenberg, Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity, Cancer Immunology, Immunotherapy, vol.108, issue.10, pp.1593-600, 2010.
DOI : 10.1007/s00262-010-0855-8

V. Umansky, A. Sevko, C. Gebhardt, and J. Utikal, Myeloidderived suppressor cells in malignant melanoma, JDDG J Dtsch Dermatol Ges, vol.12, pp.1021-1028, 2014.

M. Slattery and C. Dong, Neutrophils influence melanoma adhesion and migration under flow conditions, International Journal of Cancer, vol.168, issue.5, pp.713-735, 2003.
DOI : 10.1002/ijc.11297

C. Dong, M. Slattery, S. Liang, and H. Peng, Melanoma Cell Extravasation under Flow Conditions Is Modulated by Leukocytes and Endogenously Produced Interleukin 8, Mol Cell Biomech MCB, vol.2, pp.145-59, 2005.

M. Hoskins and C. Dong, Kinetics Analysis of Binding between Melanoma Cells and Neutrophils, Mol Cell Biomech MCB, vol.3, pp.79-87, 2006.

S. Tazzyman, C. Lewis, and C. Murdoch, Neutrophils: key mediators of tumour angiogenesis, International Journal of Experimental Pathology, vol.107, issue.Suppl. 16, pp.222-253, 2009.
DOI : 10.1111/j.1365-2613.2009.00641.x

P. Ferrucci, S. Gandini, A. Battaglia, S. Alfieri, D. Giacomo et al., Baseline neutrophil-to-lymphocyte ratio is associated with outcome of ipilimumab-treated metastatic melanoma patients, British Journal of Cancer, vol.102, issue.12, pp.1904-1914, 2015.
DOI : 10.1093/annonc/mdt161

C. Koh, N. Bhoo-pathy, K. Ng, R. Jabir, G. Tan et al., Utility of pre-treatment neutrophil???lymphocyte ratio and platelet???lymphocyte ratio as prognostic factors in breast cancer, British Journal of Cancer, vol.7, issue.1, pp.150-158, 2015.
DOI : 10.1002/jso.20329

S. Walsh, E. Cook, F. Goulder, T. Justin, and N. Keeling, Neutrophil-lymphocyte ratio as a prognostic factor in colorectal cancer, Journal of Surgical Oncology, vol.23, issue.3, pp.181-185, 2005.
DOI : 10.1002/jso.20329

S. Park, J. Kang, K. Jeong, J. Lee, J. Han et al., Norepinephrine induces VEGF expression and angiogenesis by a hypoxia-inducible factor-1?? protein-dependent mechanism, International Journal of Cancer, vol.99, issue.10, pp.2306-2322, 2011.
DOI : 10.1002/ijc.25589

K. Verhoeckx, R. Doornbos, R. Witkamp, J. Van-der-greef, and R. Rodenburg, Beta-adrenergic receptor agonists induce the release of granulocyte chemotactic protein-2, oncostatin M, and vascular endothelial growth factor from macrophages, International Immunopharmacology, vol.6, issue.1, pp.1-7, 2006.
DOI : 10.1016/j.intimp.2005.05.013

P. Filipazzi, V. Huber, and L. Rivoltini, Phenotype, function and clinical implications of myeloid-derived suppressor cells in cancer patients, Cancer Immunology, Immunotherapy, vol.13, issue.16, pp.255-63, 2011.
DOI : 10.1007/s00262-011-1161-9

I. Van-houdt, B. Sluijter, L. Moesbergen, W. Vos, T. De-gruijl et al., Favorable outcome in clinically stage II melanoma patients is associated with the presence of activated tumor infiltrating T-lymphocytes and preserved MHC class I antigen expression, International Journal of Cancer, vol.1, issue.3, pp.609-624, 2008.
DOI : 10.1002/ijc.23543

E. Tjin, G. Krebbers, K. Meijlink, W. Kasteele, . Van-de et al., Immune-Escape Markers in Relation to Clinical Outcome of Advanced Melanoma Patients Following Immunotherapy, Cancer Immunology Research, vol.2, issue.6, pp.538-584, 2014.
DOI : 10.1158/2326-6066.CIR-13-0097

J. Weber, G. Gibney, R. Kudchadkar, Y. B. Cheng, P. Martinez et al., 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

K. Palucka and J. Banchereau, Dendritic-Cell-Based Therapeutic Cancer Vaccines, Immunity, vol.39, issue.1, pp.38-48, 2013.
DOI : 10.1016/j.immuni.2013.07.004

K. Palucka and J. Banchereau, Cancer immunotherapy via dendritic cells, Nature Reviews Cancer, vol.29, issue.4, pp.265-77, 2012.
DOI : 10.1038/nrc3258

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3433802

A. Drutman, S. Jinwala, F. Bahjat, K. Bhardwaj, and N. , CD8+ T Cell Priming by Dendritic Cell Vaccines Requires Antigen Transfer to Endogenous Antigen Presenting Cells, PLOS ONE, vol.5, p.11144, 2010.

Y. Ma, G. Shurin, Z. Peiyuan, and M. Shurin, Dendritic Cells in the Cancer Microenvironment, Journal of Cancer, vol.4, issue.1, pp.36-44, 2012.
DOI : 10.7150/jca.5046

A. Pinzon-charry, T. Maxwell, and J. López, Dendritic cell dysfunction in cancer: A mechanism for immunosuppression, Immunology and Cell Biology, vol.41, issue.5, pp.451-61, 2005.
DOI : 10.1073/pnas.192251199

J. Hervé, L. Dubreil, V. Tardif, M. Terme, S. Pogu et al., ??2-Adrenoreceptor Agonist Inhibits Antigen Cross-Presentation by Dendritic Cells, The Journal of Immunology, vol.190, issue.7, pp.3163-71, 2013.
DOI : 10.4049/jimmunol.1201391

M. Takenaka, L. Araujo, J. Maricato, V. Nascimento, M. Guereschi et al., -Adrenergic Receptor???Mediated Inhibition of NF-??B and AP-1 in Dendritic Cells, The Journal of Immunology, vol.196, issue.2, pp.637-681, 2016.
DOI : 10.4049/jimmunol.1501206

URL : https://hal.archives-ouvertes.fr/hal-01429768

A. Ladányi, J. Kiss, A. Mohos, B. Somlai, G. Liszkay et al., Prognostic impact of B-cell density in cutaneous melanoma, Cancer Immunology, Immunotherapy, vol.4, issue.12, pp.1729-1767, 2011.
DOI : 10.1007/s00262-011-1071-x

K. Garg, M. Maurer, J. Griss, M. Brüggen, I. Wolf et al., Tumor-associated B cells in cutaneous primary melanoma and improved clinical outcome, Human Pathology, vol.54, pp.157-64, 2016.
DOI : 10.1016/j.humpath.2016.03.022