P. Harnett, F. Kirsten, and M. Tattersall, Drug resistance in clinical practice: patterns of treatment failure in advanced breast and ovarian cancer., Journal of Clinical Oncology, vol.4, issue.6, pp.952-959, 1986.
DOI : 10.1200/JCO.1986.4.6.952

E. Sato, S. Olson, and J. Ahn, Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer, Proceedings of the National Academy of Sciences, vol.102, issue.51, pp.18538-18581, 2005.
DOI : 10.1073/pnas.0509182102

L. Zhang, J. Conejo-garcia, and D. Katsaros, Intratumoral T Cells, Recurrence, and Survival in Epithelial Ovarian Cancer, New England Journal of Medicine, vol.348, issue.3, pp.203-216, 2003.
DOI : 10.1056/NEJMoa020177

V. Barnaba, M. Paroli, and S. Piconese, The Ambiguity in Immunology, Frontiers in Immunology, vol.3, p.18, 2012.
DOI : 10.3389/fimmu.2012.00018

T. Curiel, S. Wei, and H. Dong, Blockade of B7-H1 improves myeloid dendritic cell???mediated antitumor immunity, Nature Medicine, vol.9, issue.5, pp.562-569, 2003.
DOI : 10.1038/nm863

J. Hamanishi, M. Mandai, and M. Iwasaki, Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer, Proceedings of the National Academy of Sciences, vol.104, issue.9, pp.3360-3365, 2007.
DOI : 10.1073/pnas.0611533104

A. Mantovani, P. Allavena, and A. Sica, Cancer-related inflammation, Nature, vol.342, issue.7203, pp.436-480, 2008.
DOI : 10.1038/nature07205

U. Scarlett, M. Rutkowski, and A. Rauwerdink, Ovarian cancer progression is controlled by phenotypic changes in dendritic cells, The Journal of Experimental Medicine, vol.63, issue.3, pp.495-506, 2012.
DOI : 10.1056/NEJMoa020177

W. Zou, Regulatory T cells, tumour immunity and immunotherapy, Nature Reviews Immunology, vol.306, issue.4, pp.295-307, 2006.
DOI : 10.1038/nri1806

R. Schreiber, L. Old, and M. Smyth, Cancer Immunoediting: Integrating Immunity's Roles in Cancer Suppression and Promotion, Science, vol.331, issue.6024, pp.1565-70, 2011.
DOI : 10.1126/science.1203486

V. Dutoit, C. Herold-mende, and N. Hilf, Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy, Brain, vol.135, issue.4, pp.1042-54, 2012.
DOI : 10.1093/brain/aws042

T. Walther and M. Mann, Mass spectrometry???based proteomics in cell biology, The Journal of Cell Biology, vol.6, issue.Suppl 2, pp.491-500, 2010.
DOI : 10.1126/science.1179689

G. Kroemer, L. Galluzzi, and O. Kepp, Immunogenic Cell Death in Cancer Therapy, Annual Review of Immunology, vol.31, issue.1, pp.51-72, 2013.
DOI : 10.1146/annurev-immunol-032712-100008

R. Zappasodi, S. Pupa, and G. Ghedini, Improved Clinical Outcome in Indolent B-Cell Lymphoma Patients Vaccinated with Autologous Tumor Cells Experiencing Immunogenic Death, Cancer Research, vol.70, issue.22, pp.9062-72, 2010.
DOI : 10.1158/0008-5472.CAN-10-1825

Y. Ma, O. Kepp, and F. Ghiringhelli, Chemotherapy and radiotherapy: Cryptic anticancer vaccines, Seminars in Immunology, vol.22, issue.3, pp.113-137, 2010.
DOI : 10.1016/j.smim.2010.03.001

L. Zitvogel, L. Apetoh, and F. Ghiringhelli, Immunological aspects of cancer chemotherapy, Nature Reviews Immunology, vol.313, issue.1, pp.59-73, 2008.
DOI : 10.1038/nri2216

P. Rawson, C. Molette, and M. Videtta, Cross-presentation of caspase-cleaved apoptotic self antigens in HIV infection, Nature Medicine, vol.162, issue.12, pp.1431-1440, 2007.
DOI : 10.1002/eji.200324539

T. Boon and P. Van-der-bruggen, Human tumor antigens recognized by T lymphocytes, Journal of Experimental Medicine, vol.183, issue.3, pp.725-734, 1996.
DOI : 10.1084/jem.183.3.725

V. Barnaba, A. Franco, and A. Alberti, Selective killing of hepatitis B envelope antigen-specific B cells by class I-restricted, exogenous antigen-specific T lymphocytes, Nature, vol.345, issue.6272, pp.258-60, 1990.
DOI : 10.1038/345258a0

D. Accapezzato, V. Francavilla, and M. Paroli, Hepatic expansion of a virus-specific regulatory CD8+ T cell population in chronic hepatitis C virus infection, Journal of Clinical Investigation, vol.113, issue.7, pp.963-72, 2004.
DOI : 10.1172/JCI200420515

E. Lanitis, D. Dangaj, and I. Hagemann, Primary Human Ovarian Epithelial Cancer Cells Broadly Express HER2 at Immunologically-Detectable Levels, PLoS ONE, vol.173, issue.11, p.49829, 2012.
DOI : 10.1371/journal.pone.0049829.s002

URL : http://doi.org/10.1371/journal.pone.0049829

M. Albert, Opinion: Death-defying immunity: do apoptotic cells influence antigen processing and presentation?, Nature Reviews Immunology, vol.104, issue.3, pp.223-254, 2004.
DOI : 10.1038/nri11308

URL : https://hal.archives-ouvertes.fr/pasteur-01402360

O. Joffre, E. Segura, and A. Savina, Cross-presentation by dendritic cells, Nature Reviews Immunology, vol.195, issue.8, pp.557-69, 2012.
DOI : 10.1038/nri3254

D. Accapezzato, V. Visco, and V. Francavilla, T cell responses against soluble antigens in vivo, The Journal of Experimental Medicine, vol.68, issue.6, pp.817-845, 2005.
DOI : 10.1083/jcb.137.1.93

Y. Ma, S. Adjemian, and S. Mattarollo, Anticancer Chemotherapy-Induced Intratumoral Recruitment and Differentiation of Antigen-Presenting Cells, Immunity, vol.38, issue.4, pp.729-770, 2013.
DOI : 10.1016/j.immuni.2013.03.003

C. Hinrichs, A. Kaiser, and C. Paulos, Type 17 CD8+ T cells display enhanced antitumor immunity, Blood, vol.114, issue.3, pp.596-605, 2009.
DOI : 10.1182/blood-2009-02-203935

I. Kryczek, S. Wei, and W. Szeliga, Endogenous IL-17 contributes to reduced tumor growth and metastasis, Blood, vol.114, issue.2, pp.357-366, 2009.
DOI : 10.1182/blood-2008-09-177360

Y. Ma, L. Aymeric, and C. Locher, Contribution of IL-17???producing ???? T cells to the efficacy of anticancer chemotherapy, The Journal of Experimental Medicine, vol.178, issue.3, pp.491-503, 2011.
DOI : 10.1016/j.cell.2010.02.015

URL : https://hal.archives-ouvertes.fr/pasteur-00576679

N. Martin-orozco, P. Muranski, and Y. Chung, T Helper 17 Cells Promote Cytotoxic T Cell Activation in Tumor Immunity, Immunity, vol.31, issue.5, pp.787-98, 2009.
DOI : 10.1016/j.immuni.2009.09.014

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

J. Hataye, J. Moon, and A. Khoruts, Naive and Memory CD4+ T Cell Survival Controlled by Clonal Abundance, Science, vol.312, issue.5770, pp.114-130, 2006.
DOI : 10.1126/science.1124228

A. Kosmrlj, E. Read, and Y. Qi, Effects of thymic selection of the T-cell repertoire on HLA class???I-associated control of HIV infection, Nature, vol.171, issue.7296, pp.350-354, 2010.
DOI : 10.1046/j.1365-2540.2001.00901.x

A. Marzo, K. Klonowski, L. Bon, and A. , Initial T cell frequency dictates memory CD8+ T cell lineage commitment, Nature Immunology, vol.166, issue.8, pp.793-802, 2005.
DOI : 10.1126/science.279.5354.1223

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2849311

M. Pittet, D. Valmori, and P. Dunbar, T Cells in a Large Proportion of Human Histocompatibility Leukocyte Antigen (Hla)-A2 Individuals, The Journal of Experimental Medicine, vol.59, issue.5, pp.705-720, 1999.
DOI : 10.1097/00002371-199601000-00009

C. Chang, Y. Hsu, and C. Wu, Dose-Dense Chemotherapy Improves Mechanisms of Antitumor Immune Response, Cancer Research, vol.73, issue.1, pp.119-146, 2013.
DOI : 10.1158/0008-5472.CAN-12-2225

W. Lesterhuis, C. Punt, and S. Hato, Platinum-based drugs disrupt STAT6-mediated suppression of immune responses against cancer in humans and mice, Journal of Clinical Investigation, vol.121, issue.8, pp.3100-3108, 2011.
DOI : 10.1172/JCI43656DS1

P. Savage, K. Vosseller, and C. Kang, Recognition of a Ubiquitous Self Antigen by Prostate Cancer-Infiltrating CD8+ T Lymphocytes, Science, vol.319, issue.5860, pp.215-235, 2008.
DOI : 10.1126/science.1148886

A. Schietinger, M. Philip, and R. Liu, T cells during the effector phase, The Journal of Experimental Medicine, vol.21, issue.11, pp.2469-77, 2010.
DOI : 10.1158/0008-5472.CAN-07-5324

J. Walch, Q. Zeng, and Q. Li, Cognate antigen directs CD8+ T cell migration to vascularized transplants, Journal of Clinical Investigation, vol.123, issue.6, pp.2663-71, 2013.
DOI : 10.1172/JCI66722DS1

P. Robbins, Y. Lu, and M. El-gamil, Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells, Nature Medicine, vol.360, issue.6, pp.747-52, 2013.
DOI : 10.1038/nm.3161

D. Ciocca and S. Calderwood, Heat shock proteins in cancer: diagnostic, prognostic, predictive, and treatment implications, Cell Stress & Chaperones, vol.109, issue.2, pp.86-103, 2005.
DOI : 10.1379/CSC-99r.1

K. Nieman, H. Kenny, and C. Penicka, Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth, Nature Medicine, vol.239, issue.11, pp.1498-503, 2011.
DOI : 10.1007/s12014-010-9055-y

C. Balch, D. Matei, and T. Huang, Role of epigenomics in ovarian and endometrial cancers, Epigenomics, vol.2, issue.3, pp.419-466, 2010.
DOI : 10.2217/epi.10.19

L. Seeber and P. Van-diest, Epigenetics in Ovarian Cancer, Methods Mol Biol, vol.863, pp.253-69, 2012.
DOI : 10.1007/978-1-61779-612-8_15

A. Dimberg, S. Rylova, and L. Dieterich, ??B-crystallin promotes tumor angiogenesis by increasing vascular survival during tube morphogenesis, Blood, vol.111, issue.4, pp.2015-2038, 2008.
DOI : 10.1182/blood-2007-04-087841

S. Vemulapalli, A. Mita, and Y. Alvarado, The emerging role of mammalian target of rapamycin inhibitors in the treatment of sarcomas, Targeted Oncology, vol.66, issue.3, pp.29-39, 2011.
DOI : 10.1007/s11523-011-0179-4