J. Eble, G. Sauter, J. Epstein, I. Sesterhenn, and . Editor, World Health Organization classification of tumours. Pathology and genetics of tumours of the urinary system and male genital organs, pp.12-1723, 2004.

J. Gnarra, K. Tory, and Y. Weng, Mutations of the VHL tumour suppressor gene in renal carcinoma, Nature Genetics, vol.88, issue.1, pp.85-90, 1994.
DOI : 10.1038/ng0594-85

W. Kim and W. Kaelin, Gene Mutation in Human Cancer, Journal of Clinical Oncology, vol.22, issue.24, pp.4991-5004, 2004.
DOI : 10.1200/JCO.2004.05.061

N. Barnabas, M. Amin, K. Pindolia, R. Nanavati, M. Amin et al., Mutations in the von Hippel-Lindau (VHL) gene refine differential diagnostic criteria in renal cell carcinoma, Journal of Surgical Oncology, vol.91, issue.1, pp.52-60, 2002.
DOI : 10.1002/jso.10086

W. Kaelin and . Jr, The von Hippel-Lindau Tumor Suppressor Protein and Clear Cell Renal Carcinoma, Clinical Cancer Research, vol.13, issue.2, pp.680-684, 2007.
DOI : 10.1158/1078-0432.CCR-06-1865

M. Oken, R. Creech, and D. Tormey, Toxicity and response criteria of the Eastern Cooperative Oncology Group, AMERICAN JOURNAL OF CLINICAL ONCOLOGY, vol.5, issue.6, pp.649-55, 1982.
DOI : 10.1097/00000421-198212000-00014

S. Edge and C. Compton, The American Joint Committee on Cancer: the 7th Edition of the AJCC Cancer Staging Manual and the Future of TNM, Annals of Surgical Oncology, vol.17, issue.6, pp.1471-1475, 2010.
DOI : 10.1245/s10434-010-0985-4

J. Patard, P. Fergelot, and P. Karakiewicz, Low CAIX expression and absence of VHL gene mutation are associated with tumor aggressiveness and poor survival of clear cell renal cell carcinoma, International Journal of Cancer, vol.98, issue.2, pp.395-400, 2008.
DOI : 10.1002/ijc.23496

URL : https://hal.archives-ouvertes.fr/hal-00278587

N. Rioux-leclercq, P. Fergelot, and S. Zerrouki, Plasma level and tissue expression of vascular endothelial growth factor in renal cell carcinoma: a prospective study of 50 cases, Human Pathology, vol.38, issue.10
DOI : 10.1016/j.humpath.2007.02.014

J. Patard, N. Rioux-leclercq, and D. Masson, Absence of VHL gene alteration and high VEGF expression are associated with tumour aggressiveness and poor survival of renal-cell carcinoma, British Journal of Cancer, vol.94, issue.8, pp.1417-1441, 2009.
DOI : 10.1186/1471-2407-5-57

URL : https://hal.archives-ouvertes.fr/inserm-00418630

J. Jeuken, S. Cornelissen, and M. Vriezen, MS-MLPA: an attractive alternative laboratory assay for robust, reliable, and semiquantitative detection of MGMT promoter hypermethylation in gliomas, Laboratory Investigation, vol.349, issue.10, pp.1055-6543, 2007.
DOI : 10.1038/labinvest.3700664

P. Schraml, K. Struckmann, and F. Hatz, VHL mutations and their correlation with tumour cell proliferation, microvessel density, and patient prognosis in clear cell renal cell carcinoma, The Journal of Pathology, vol.87, issue.2, pp.186-93, 2002.
DOI : 10.1002/path.1034

. Hippel-lindau, VHL) gene alterations and relationship with clinical variables in sporadic renal cancer, Cancer Res, vol.66, issue.4, pp.2000-2011, 2006.

K. Kondo, M. Yao, and M. Yoshida, Comprehensive mutational analysis of theVHL gene in sporadic renal cell carcinoma: Relationship to clinicopathological parameters, Genes, Chromosomes and Cancer, vol.8, issue.1, pp.58-68, 2002.
DOI : 10.1002/gcc.10054

J. Herman, F. Latif, and Y. Weng, Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma., Proceedings of the National Academy of Sciences, vol.91, issue.21, pp.9700-9704, 1994.
DOI : 10.1073/pnas.91.21.9700

C. Kwak, Y. Park, and C. Jeong, Sarcomatoid differentiation as a prognostic factor for immunotherapy in metastatic renal cell carcinoma Expression of hypoxia inducible factor-1? and 2? in conventional renal cell carcinoma with or without sarcomatoid differentiation, J Surg Oncol. Urol Oncol, vol.9529, issue.46, pp.317-23731, 2007.

O. Nakano, M. Sato, and Y. Naito, Proliferative activity of intratumoral CD8(+) Tlymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity, Cancer Res, vol.61, issue.13, pp.5132-5138, 2001.

W. Webster, C. Lohse, and R. Thompson, Mononuclear cell infiltration in clearcell renal cell carcinoma independently predicts patient survival Behavior of immune players in the tumor microenvironment, Cancer. Curr Opin Oncol, vol.10721, issue.221, pp.46-5353, 2006.

J. Brahmer, S. Tykodi, and L. Chow, Safety and Activity of Anti???PD-L1 Antibody in Patients with Advanced Cancer, New England Journal of Medicine, vol.366, issue.26, pp.2455-65, 2012.
DOI : 10.1056/NEJMoa1200694

S. Topalian, F. Hodi, and J. Brahmer, Safety, Activity, and Immune Correlates of Anti???PD-1 Antibody in Cancer, New England Journal of Medicine, vol.366, issue.26, pp.2443-54, 2012.
DOI : 10.1056/NEJMoa1200690

M. Kang, K. Kim, and J. Bae, Tumor-infiltrating PD1-Positive Lymphocytes and

J. Edeline, C. Vigneau, J. Patard, N. Rioux-leclercq, L. Fournier et al., Signalling pathways in renal-cell carcinoma: from the molecular biology to the future therapy] New insights into the management of renal cell cancer, Bull Cancer. Oncology, vol.9784, issue.271, pp.5-1522, 2010.

D. Huang, Y. Ding, and M. Zhou, Interleukin-8 Mediates Resistance to Antiangiogenic Agent Sunitinib in Renal Cell Carcinoma, Cancer Research, vol.70, issue.3, pp.1063-71, 2010.
DOI : 10.1158/0008-5472.CAN-09-3965

C. Swanton, J. Larkin, and M. Gerlinger, Predictive biomarker discovery through the parallel integration of clinical trial and functional genomics datasets, Genome Medicine, vol.2, issue.8, p.53, 2010.
DOI : 10.1186/gm174

B. Rini and M. Atkins, Resistance to targeted therapy in renal-cell carcinoma, The Lancet Oncology, vol.10, issue.10, pp.992-1000, 2009.
DOI : 10.1016/S1470-2045(09)70240-2