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, The Oncologist, vol.16, issue.1, pp.5-24, 2011.

B. Guillot, J. Charles, and D. Cupissol, Recommandations/ Traitement des patients atteints de mélanome stade III inopérable ou de stade IV / Société Française de Dermatologie, 2017.

C. Robert, G. V. Long, B. Brady, C. Dutriaux, M. Maio et al., Nivolumab in previously untreated melanoma without BRAF mutation, N Engl J Med, vol.372, issue.4, pp.320-350, 2015.

J. Schachter, A. Ribas, G. V. Long, A. Arance, J. Grob et al., Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006), Lancet Lond Engl, vol.390, pp.1853-62, 2017.

J. S. Weber, M. Postow, C. D. Lao, and D. Schadendorf, Management of Adverse Events Following Treatment With Anti-Programmed Death-1 Agents. The Oncologist, vol.21, pp.1230-1270, 2016.

E. Saâda-bouzid, C. Defaucheux, A. Karabajakian, V. P. Coloma, V. Servois et al., Hyperprogression during anti-PD-1/PD-L1 therapy in patients with recurrent and/or metastatic head and neck squamous cell carcinoma, Ann Oncol, vol.28, issue.7, pp.1605-1616, 2017.

S. Champiat, L. Dercle, S. Ammari, C. Massard, A. Hollebecque et al., Hyperprogressive Disease Is a New Pattern of Progression in Cancer Patients Treated by Anti-PD-1/PD-L1, Clin Cancer Res Off J Am Assoc Cancer Res, vol.23, issue.8, pp.1920-1928, 201715.

L. Zitvogel, D. Hannani, and F. Martin, Immunothérapie des cancers au troisième millénaire, 2014.

L. Galluzzi, E. Vacchelli, J. Bravo-san-pedro, A. Buqué, L. Senovilla et al., Classification of current anticancer immunotherapies, Oncotarget, vol.5, issue.24, pp.12472-508, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01142463

B. Bonnotte, C. Hoarau, and M. Rosenzwaig, Les cellules dendritiques

F. Lemoine, Y. Lebranchu, and O. Boyer, Immunité adaptative : lymphocytes T régulateurs et notion de tolérance

D. Carvalho, M. Adotevi, O. Frenkel, and V. , Immunité adaptative : activation et polarisation des lymphoctes T

R. D. Schreiber, L. J. Old, and M. J. Smyth, Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion, Science, vol.331, issue.6024, pp.1565-70, 2011.

C. Caux, Immunothérapie : les checkpoints, Rev Mal Respir Actual, vol.8, issue.5, pp.387-98, 2016.

G. P. Dunn, A. T. Bruce, H. Ikeda, L. J. Old, and R. D. Schreiber, Cancer immunoediting: from immunosurveillance to tumor escape, Nat Immunol, vol.3, issue.11, pp.991-999, 2002.

W. Ma, B. M. Gilligan, J. Yuan, and T. Li, Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy, J Hematol OncolJ Hematol Oncol, vol.9, issue.1, p.47, 201627.

M. Nishino, N. H. Ramaiya, H. Hatabu, and F. S. Hodi, Monitoring immune-checkpoint blockade: response evaluation and biomarker development, Nat Rev Clin Oncol, vol.14, issue.11, pp.655-68, 2017.

S. L. Topalian, F. S. Hodi, J. R. Brahmer, S. N. Gettinger, D. C. Smith et al., Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer, N Engl J Med, vol.366, issue.26, pp.2443-54, 2012.

S. P. Patel and R. Kurzrock, PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy, Mol Cancer Ther, vol.14, issue.4, pp.847-56, 2015.

S. L. Topalian, J. M. Taube, R. A. Anders, and D. M. Pardoll, Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy, Nat Rev Cancer, vol.16, issue.5, pp.275-87, 2016.

P. C. Tumeh, C. L. Harview, J. H. Yearley, I. P. Shintaku, E. Taylor et al., PD-1 blockade induces responses by inhibiting adaptive immune resistance, Nature, vol.515, issue.7528, pp.568-71, 2014.

R. S. Herbst, J. Soria, M. Kowanetz, G. D. Fine, O. Hamid et al., Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients, Nature, vol.515, issue.7528, pp.563-570, 2014.

J. Larkin, V. Chiarion-sileni, R. Gonzalez, J. J. Grob, C. L. Cowey et al., Combined Nivolumab and Ipilimumab or Monotherapy in Previously Untreated Melanoma, N Engl J Med, vol.373, issue.1, pp.23-34, 2015.

M. Reck, D. Rodríguez-abreu, A. G. Robinson, R. Hui, T. Cs?szi et al., Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer, N Engl J Med, vol.375, issue.19, pp.1823-1856, 201610.

, Pembrolizumab (KEYTRUDA) checkpoint inhibitor, 2016.

A. I. Daud, K. Loo, M. L. Pauli, R. Sanchez-rodriguez, P. M. Sandoval et al., Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma, J Clin Invest, vol.126, issue.9, pp.3447-52, 2016.

R. E. Vilain, A. M. Menzies, J. S. Wilmott, H. Kakavand, J. Madore et al., Dynamic Changes in PD-L1 Expression and Immune Infiltrates Early During Treatment Predict Response to PD-1 Blockade in Melanoma, Clin Cancer Res Off J Am Assoc Cancer Res, vol.23, issue.17, pp.5024-5057, 2017.

A. Frelau, M. Pracht, L. Sourd, S. Lespagnol, A. Corre et al., Biomarqueurs prédictifs de réponse aux inhibiteurs de points de contrôle immuns, Bull Cancer, 2018.

W. Hugo, J. M. Zaretsky, L. Sun, C. Song, B. H. Moreno et al., Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma, Cell, vol.165, issue.1, pp.35-44, 2016.

D. T. Le, J. N. Uram, H. Wang, B. R. Bartlett, H. Kemberling et al., PD-1 Blockade in Tumors with Mismatch-Repair Deficiency, N Engl J Med, vol.372, issue.26, pp.2509-2529, 2015.

A. C. Huang, M. A. Postow, R. J. Orlowski, R. Mick, B. Bengsch et al., T-cell invigoration to tumour burden ratio associated with anti-PD-1 response, Nature, vol.545, issue.7652, pp.60-65, 2017.

C. Krieg, M. Nowicka, S. Guglietta, S. Schindler, F. J. Hartmann et al., Highdimensional single-cell analysis predicts response to anti-PD-1 immunotherapy, Nat Med, vol.24, issue.2, pp.144-53, 2018.

L. S. Fournier, S. Ammari, R. Thiam, and C. Cuénod, Critères de la réponse tumorale en imagerie : RECIST, mRECIST, Cheson. Datarevues22115706v95i7-8S2211570614002859, 2014.

M. Veldhoen, C. Uyttenhove, J. Van-snick, H. Helmby, A. Westendorf et al., Transforming growth factor-beta "reprograms" the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset, Nat Immunol, vol.9, issue.12, pp.1341-1347, 2008.

V. Dardalhon, A. Awasthi, H. Kwon, G. Galileos, W. Gao et al., IL-4 inhibits TGF-beta-induced Foxp3+ T cells and, together with TGF-beta, generates IL-9+ IL-10+ Foxp3(-) effector T cells, Nat Immunol, vol.9, issue.12, pp.1347-55, 2008.

Y. Lu, S. Hong, H. Li, J. Park, B. Hong et al., Th9 cells promote antitumor immune responses in vivo, J Clin Invest, vol.122, issue.11, pp.4160-71, 2012.

Y. Nonomura, A. Otsuka, C. Nakashima, J. A. Seidel, A. Kitoh et al., Peripheral blood Th9 cells are a possible pharmacodynamic biomarker of nivolumab treatment efficacy in metastatic melanoma patients, Oncoimmunology, vol.5, issue.12, p.1248327, 2016.

J. Saison, J. Demaret, F. Venet, C. Chidiac, C. Malcus et al., CD4+CD25+CD127-assessment as a surrogate phenotype for FOXP3+ regulatory T cells in HIV-1 infected viremic and aviremic subjects, Cytometry B Clin Cytom, vol.84, issue.1, pp.50-54, 2013.

Y. Chen, X. Chen, L. Qin, Y. Yang, and Y. Tang,

. Foxp3,

/. Cd127,

, regulatory T cells in peripheral blood of HIV

X. Bao-yu-fen-zi-mian and Y. Xue, Za Zhi Chin J Cell Mol Immunol, vol.28, issue.11, pp.1188-91, 2012.

W. H. Fridman, F. Pagès, C. Sautès-fridman, and J. Galon, The immune contexture in human tumours: impact on clinical outcome, Nat Rev Cancer, vol.15, issue.4, pp.298-306, 2012.

S. Nair, V. Elkord, and E. , Immune checkpoint inhibitors in cancer therapy: a focus on T-regulatory cells, Immunol Cell Biol, vol.96, issue.1, pp.21-33, 2018.

P. Salama, M. Phillips, F. Grieu, M. Morris, N. Zeps et al., Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer, J Clin Oncol Off J Am Soc Clin Oncol, vol.27, issue.2, pp.186-92, 2009.

B. Chaudhary and E. Elkord, Regulatory T Cells in the Tumor Microenvironment and Cancer Progression: Role and Therapeutic Targeting. Vaccines, vol.4, 2016.

K. A. Ward-hartstonge and R. A. Kemp, Regulatory T-cell heterogeneity and the cancer immune response, Clin Transl Immunol, vol.6, issue.9, p.154, 2017.

Z. Ouyang, H. Wu, L. Li, Y. Luo, X. Li et al., Regulatory T cells in the immunotherapy of melanoma, Tumour Biol J Int Soc Oncodevelopmental Biol Med, vol.37, issue.1, pp.77-85, 2016.

M. Viguier, F. Lemaître, O. Verola, M. Cho, G. Gorochov et al., Foxp3 expressing CD4+CD25(high) regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells, J Immunol Baltim Md, vol.173, issue.2, pp.1444-53, 1950.

L. R. Javia and S. A. Rosenberg, CD4+CD25+ suppressor lymphocytes in the circulation of patients immunized against melanoma antigens, J Immunother Hagerstown Md, vol.26, issue.1, pp.85-93, 1997.

C. Jandus, G. Bioley, D. E. Speiser, and P. Romero, Selective accumulation of differentiated FOXP3(+) CD4 (+) T cells in metastatic tumor lesions from melanoma patients compared to peripheral blood, Cancer Immunol Immunother CII, vol.57, issue.12, pp.1795-805, 2008.

C. Miracco, V. Mourmouras, M. Biagioli, P. Rubegni, S. Mannucci et al., Utility of tumour-infiltrating CD25+FOXP3+ regulatory T cell evaluation in predicting local recurrence in vertical growth phase cutaneous melanoma, Oncol Rep, vol.18, issue.5, pp.1115-1137, 2007.

D. Mougiakakos, C. C. Johansson, E. Trocme, C. All-ericsson, M. A. Economou et al., Intratumoral forkhead box P3-positive regulatory T cells predict poor survival in cyclooxygenase-2-positive uveal melanoma, Cancer, vol.116, issue.9, p.78, 2010.

J. M. Baumgartner, R. Gonzalez, K. D. Lewis, W. A. Robinson, D. A. Richter et al., Increased survival from stage IV melanoma associated with fewer regulatory T Cells, J Surg Res, vol.154, issue.1, pp.13-20, 2009.

L. Halim, M. Romano, R. Mcgregor, I. Correa, P. Pavlidis et al., An Atlas of Human Regulatory T Helper-like Cells Reveals Features of Th2-like Tregs that Support a Tumorigenic Environment, Cell Rep, vol.20, issue.3, pp.757-70, 2017.

A. S. Syed-khaja, S. M. Toor, E. Salhat, H. Faour, I. et al., Preferential accumulation of regulatory T cells with highly immunosuppressive characteristics in breast tumor microenvironment, Oncotarget, vol.16, issue.20, pp.33159-71, 2017.

M. J. Selby, J. J. Engelhardt, M. Quigley, K. A. Henning, T. Chen et al., Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells, Cancer Immunol Res, vol.1, issue.1, pp.32-42, 2013.

C. T. Berger and M. Recher, Immuno-régulation par les lymphocytes T régulateurs, 2015.

T. R. Simpson, F. Li, W. Montalvo-ortiz, M. A. Sepulveda, K. Bergerhoff et al., Fcdependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma, J Exp Med, vol.210, issue.9, pp.1695-710, 2013.

F. Tang, X. Du, M. Liu, P. Zheng, and Y. Liu, Anti-CTLA-4 antibodies in cancer immunotherapy: selective depletion of intratumoral regulatory T cells or checkpoint blockade?, Cell Biosci, vol.8, p.30, 2018.

E. Simeone, G. Gentilcore, D. Giannarelli, A. M. Grimaldi, C. Caracò et al., Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma, Cancer Immunol Immunother CII, vol.63, issue.7, pp.675-83, 2014.

M. A. Curran, W. Montalvo, H. Yagita, and J. P. Allison, PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors, Proc Natl Acad Sci, vol.107, issue.9, pp.4275-80, 2010.

W. Wang, R. Lau, D. Yu, W. Zhu, A. Korman et al., PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells, Int Immunol, vol.21, issue.9, pp.1065-77, 2009.