J. A. Joyce and D. T. Fearon, T cell exclusion, immune privilege, and the tumor microenvironment, Science, vol.91, issue.9, pp.74-80
DOI : 10.1073/pnas.1211850110

D. He, H. Li, N. Yusuf, C. A. Elmets, J. Li et al., IL-17 Promotes Tumor Development through the Induction of Tumor Promoting Microenvironments at Tumor Sites and Myeloid-Derived Suppressor Cells, The Journal of Immunology, vol.184, issue.5, pp.2281-2288, 2010.
DOI : 10.4049/jimmunol.0902574

URL : http://www.jimmunol.org/content/jimmunol/184/5/2281.full.pdf

L. De-la-cruz-merino, A. Barco-sanchez, and F. Henao-carrasco,

B. Molina, J. Martinez-peinado, A. Lopez, A. Manuel-de-villena, and M. , New insights into the role of the immune microenvironment in breast carcinoma Microenvironmental regulation of tumor progression and metastasis, Clin. Dev. Immunol. Nat. Med. 2013, vol.19, pp.785317-785328, 2013.

J. A. Joyce, The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas, Science, 2016.

R. L. Bowman and J. A. Joyce, Therapeutic targeting of tumor-associated macrophages and microglia in glioblastoma, Immunotherapy, vol.6, issue.6, pp.663-666
DOI : 10.1097/CJI.0b013e3182562d59

G. P. Dunn, L. J. Old, and R. D. Schreiber, The Immunobiology of Cancer Immunosurveillance and Immunoediting, Immunity, vol.21, issue.2, pp.137-148, 2004.
DOI : 10.1016/j.immuni.2004.07.017

G. P. Dunn, A. T. Bruce, H. Ikeda, L. J. Old, and R. D. Schreiber, Cancer immunoediting: from immunosurveillance to tumor escape, Nature Immunology, vol.93, issue.11, pp.991-998, 2002.
DOI : 10.1073/pnas.93.21.11798

A. Mantovani, P. Romero, A. K. Palucka, and F. M. Marincola, Tumour immunity: effector response to tumour and role of the microenvironment, The Lancet, vol.371, issue.9614, pp.771-783, 2008.
DOI : 10.1016/S0140-6736(08)60241-X

Z. Yao, S. L. Painter, W. C. Fanslow, D. Ulrich, B. M. Macduff et al., Human IL-17: A novel cytokine derived from T cells, J. Immunol, vol.155, pp.5483-5486, 1995.

E. Maniati, R. Soper, and T. Hagemann, Up for Mischief? IL-17/Th17 in the tumour microenvironment, Oncogene, vol.178, issue.42, pp.5653-5662, 2010.
DOI : 10.1038/nri2742

S. V. Novitskiy, M. W. Pickup, A. E. Gorska, P. Owens, A. Chytil et al., TGF-? receptor II loss promotes mammary carcinoma progression by Th17 dependent mechanisms. Cancer Discov, pp.430-441, 2011.

N. R. West, S. E. Kost, S. D. Martin, K. Milne, R. J. Deleeuw et al., Tumour-infiltrating FOXP3+ lymphocytes are associated with cytotoxic immune responses and good clinical outcome in oestrogen receptor-negative breast cancer, British Journal of Cancer, vol.5, issue.1, pp.155-162, 2013.
DOI : 10.1371/journal.pone.0014282

X. Su, J. Ye, E. C. Hsueh, Y. Zhang, D. F. Hoft et al., Tumor Microenvironments Direct the Recruitment and Expansion of Human Th17 Cells, The Journal of Immunology, vol.184, issue.3, pp.1630-1641, 2010.
DOI : 10.4049/jimmunol.0902813

M. Miyashita, H. Sasano, K. Tamaki, H. Hirakawa, Y. Takahashi et al.,

A. Suzuki and N. Ohuchi, Prognostic significance of tumor-infiltrating CD8+ and FOXP3+ lymphocytes in residual tumors and alterations in these parameters after neoadjuvant chemotherapy in triple-negative breast cancer: A retrospective multicenter study, Breast Cancer Res, vol.17, pp.124-137, 2015.

C. Gu, L. Wu, and X. Li, IL-17 family: Cytokines, receptors and signaling, Cytokine, vol.64, issue.2, pp.477-485
DOI : 10.1016/j.cyto.2013.07.022

L. A. Tesmer, S. K. Lundy, S. Sarkar, and D. A. Fox, Th17 cells in human disease, Immunological Reviews, vol.29, issue.(Suppl.), pp.87-113, 2008.
DOI : 10.1111/j.1600-065X.2008.00628.x

X. O. Yang, B. P. Pappu, R. Nurieva, A. Akimzhanov, H. S. Kang et al., T Helper 17 Lineage Differentiation Is Programmed by Orphan Nuclear Receptors ROR?? and ROR??, Immunity, vol.28, issue.1, pp.29-39, 2008.
DOI : 10.1016/j.immuni.2007.11.016

L. Zhou, I. I. Ivanov, R. Spolski, R. Min, K. Shenderov et al., IL-6 programs TH-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways, Nature Immunology, vol.178, issue.9, pp.967-974, 2007.
DOI : 10.1128/MCB.24.6.2385-2396.2004

D. J. Cua, J. Sherlock, Y. Chen, C. A. Murphy, B. Joyce et al., Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain, Nature, vol.1, issue.6924, pp.744-748, 2003.
DOI : 10.1111/j.1750-3639.1991.tb00646.x

N. J. Wilson, K. Boniface, J. R. Chan, B. S. Mckenzie, W. M. Blumenschein et al., Development, cytokine profile and function of human interleukin 17???producing helper T cells, Nature Immunology, vol.176, issue.9, pp.950-957, 2007.
DOI : 10.4049/jimmunol.176.2.1098

X. O. Yang, S. H. Chang, H. Park, R. Nurieva, B. Shah et al., Regulation of inflammatory responses by IL-17F, The Journal of Experimental Medicine, vol.158, issue.5, pp.1063-1075, 2008.
DOI : 10.4049/jimmunol.168.12.6294

URL : http://jem.rupress.org/content/jem/205/5/1063.full.pdf

A. J. Van-beelen, Z. Zelinkova, E. W. Taanman-kueter, F. J. Muller, D. W. Hommes et al., Stimulation of the Intracellular Bacterial Sensor NOD2 Programs Dendritic Cells to Promote Interleukin-17 Production in Human Memory T Cells, Immunity, vol.27, issue.4, pp.660-669, 2007.
DOI : 10.1016/j.immuni.2007.08.013

I. Kryczek, S. Wei, W. Gong, X. Shu, W. Szeliga et al., Cutting Edge: IFN-?? Enables APC to Promote Memory Th17 and Abate Th1 Cell Development, The Journal of Immunology, vol.181, issue.9, pp.5842-5846, 2008.
DOI : 10.4049/jimmunol.181.9.5842

URL : http://www.jimmunol.org/content/jimmunol/181/9/5842.full.pdf

N. Martin-orozco, P. Muranski, Y. Chung, X. O. Yang, T. Yamazaki et al., T Helper 17 Cells Promote Cytotoxic T Cell Activation in Tumor Immunity, Immunity, vol.31, issue.5, pp.787-798, 2009.
DOI : 10.1016/j.immuni.2009.09.014

URL : https://doi.org/10.1016/j.immuni.2009.09.014

I. Kryczek, M. Banerjee, P. Cheng, L. Vatan, W. Szeliga et al., Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments, Blood, vol.114, issue.6, pp.1141-1149, 2009.
DOI : 10.1182/blood-2009-03-208249

P. Ghadjar, C. Rubie, D. M. Aebersold, and U. Keilholz, The chemokine CCL20 and its receptor CCR6 in human malignancy with focus on colorectal cancer, International Journal of Cancer, vol.27, issue.4, pp.741-745, 2009.
DOI : 10.1002/ijc.24468

URL : http://onlinelibrary.wiley.com/doi/10.1002/ijc.24468/pdf

P. Muranski, A. Boni, P. A. Antony, L. Cassard, K. R. Irvine et al., Tumor-specific Th17-polarized cells eradicate large established melanoma, Blood, vol.112, issue.2, pp.362-373, 2008.
DOI : 10.1182/blood-2007-11-120998

URL : http://www.bloodjournal.org/content/bloodjournal/112/2/362.full.pdf

I. Kryczek, R. Liu, G. Wang, K. Wu, X. Shu et al., FOXP3 Defines Regulatory T Cells in Human Tumor and Autoimmune Disease, Cancer Research, vol.69, issue.9, pp.3995-4000, 2009.
DOI : 10.1158/0008-5472.CAN-08-3804

URL : http://cancerres.aacrjournals.org/content/canres/69/9/3995.full.pdf

E. Rouvier, M. F. Luciani, M. G. Mattei, F. Denizot, and P. Golstein, CTLA-8, cloned from an activated T cell, bearing AU-rich messenger RNA instability sequences, and homologous to a herpesvirus saimiri gene

, J. Immunol, vol.150, pp.5445-5456, 1993.

J. K. Kolls and A. Linden, Interleukin-17 Family Members and Inflammation, Immunity, vol.21, issue.4, pp.467-476, 2004.
DOI : 10.1016/j.immuni.2004.08.018

T. A. Moseley, D. R. Haudenschild, L. Rose, and A. H. Reddi, Interleukin-17 family and IL-17 receptors, Cytokine & Growth Factor Reviews, vol.14, issue.2, pp.155-174, 2003.
DOI : 10.1016/S1359-6101(03)00002-9

S. Xu and X. Cao, Interleukin-17 and its expanding biological functions, Cellular & Molecular Immunology, vol.162, issue.3, pp.164-174, 2010.
DOI : 10.1016/j.tips.2008.11.004

W. Jin and C. Dong, IL-17 cytokines in immunity and inflammation, Emerging Microbes & Infections, vol.179, issue.9, pp.60-65
DOI : 10.1038/ni.2427

X. Song and Y. Qian, The activation and regulation of IL-17 receptor mediated signaling, Cytokine, vol.62, issue.2, pp.175-182
DOI : 10.1016/j.cyto.2013.03.014

Z. Chen and J. J. Shea, Regulation of IL-17 production in human lymphocytes, Cytokine, vol.41, issue.2, pp.71-78, 2008.
DOI : 10.1016/j.cyto.2007.09.009

B. W. Kirkham, A. Kavanaugh, and K. Reich, Interleukin-17A: a unique pathway in immune-mediated diseases: psoriasis, psoriatic arthritis and rheumatoid arthritis, Immunology, vol.61, issue.Suppl. 10, pp.133-142, 2014.
DOI : 10.1136/gutjnl-2011-301668

D. Toy, D. Kugler, M. Wolfson, T. Vanden-bos, J. Gurgel et al., Cutting Edge: Interleukin 17 Signals through a Heteromeric Receptor Complex, The Journal of Immunology, vol.177, issue.1, pp.36-39, 2006.
DOI : 10.4049/jimmunol.177.1.36

E. A. Rickel, L. A. Siegel, B. R. Yoon, J. B. Rottman, D. G. Kugler et al., Identification of Functional Roles for Both IL-17RB and IL-17RA in Mediating IL-25-Induced Activities, The Journal of Immunology, vol.181, issue.6, pp.4299-4310, 2008.
DOI : 10.4049/jimmunol.181.6.4299

URL : http://www.jimmunol.org/content/jimmunol/181/6/4299.full.pdf

T. Shalom-barak, J. Quach, and M. Lotz, Interleukin-17-induced Gene Expression in Articular Chondrocytes Is Associated with Activation of Mitogen-activated Protein Kinases and NF-??B, Journal of Biological Chemistry, vol.157, issue.42, pp.27467-27473, 1998.
DOI : 10.1038/sj.onc.1201253

URL : http://www.jbc.org/content/273/42/27467.full.pdf

S. H. Chang, H. Park, and C. Dong, Act1 Adaptor Protein Is an Immediate and Essential Signaling Component of Interleukin-17 Receptor, Journal of Biological Chemistry, vol.77, issue.47, pp.35603-35607, 2006.
DOI : 10.4049/jimmunol.176.2.711

URL : http://www.jbc.org/content/281/47/35603.full.pdf

B. Zhang, C. Liu, W. Qian, Y. Han, X. Li et al., Structure of the unique SEFIR domain from human interleukin 17 receptor A reveals a composite ligand-binding site containing a conserved ??-helix for Act1 binding and IL-17 signaling, Acta Crystallographica Section D Biological Crystallography, vol.190, issue.5, pp.1476-1483, 2014.
DOI : 10.4049/jimmunol.1202922

F. Sun, Z. Qu, Y. Xiao, J. Zhou, T. F. Burns et al., NF-??B1 p105 suppresses lung tumorigenesis through the Tpl2 kinase but independently of its NF-??B function, Oncogene, vol.6, issue.18, pp.2299-2310, 2016.
DOI : 10.1158/1078-0432.CCR-10-0992

URL : http://europepmc.org/articles/pmc4548811?pdf=render

J. Fabre, J. Giustinniani, F. Antonicelli, Y. Merrouche, M. Bagot et al., A Focus on IL-17 Targeting Treatments in Psoriasis, Journal of Dermatological Research, vol.1, issue.2, pp.1-5, 2016.
DOI : 10.17554/j.issn.2413-8223.2016.01.10

URL : http://www.ghrnet.org/index.php/jdr/article/download/1677/2052

D. Haudenschild, T. Moseley, L. Rose, and A. H. Reddi, Soluble and Transmembrane Isoforms of Novel Interleukin-17 Receptor-like Protein by RNA Splicing and Expression in Prostate Cancer, Journal of Biological Chemistry, vol.162, issue.6, pp.4309-4316, 2002.
DOI : 10.1126/science.1058040

G. E. Steiner, M. E. Newman, D. Paikl, U. Stix, N. Memaran-dagda et al., Expression and function of pro-inflammatory interleukin IL-17 and IL-17 receptor in normal, benign hyperplastic, and malignant prostate, The Prostate, vol.53, issue.3, pp.171-182, 2003.
DOI : 10.1016/S0090-4295(98)00455-5

K. S. Sfanos, T. C. Bruno, C. H. Maris, L. Xu, and C. J. Thoburn, W.B

C. G. Drake, Phenotypic analysis of prostate-infiltrating lymphocytes reveals Th17 and Treg skewing, Clin. Cancer Res, vol.14, pp.3254-3261, 2008.

B. Zhang, G. Rong, H. Wei, M. Zhang, J. Bi et al., The prevalence of Th17 cells in patients with gastric cancer, Biochemical and Biophysical Research Communications, vol.374, issue.3, pp.533-537, 2008.
DOI : 10.1016/j.bbrc.2008.07.060

E. Derhovanessian, V. Adams, K. Hahnel, A. Groeger, H. Pandha et al., T-cells, but not Tregs, correlates with clinical response to whole-cell vaccination in prostate cancer patients, International Journal of Cancer, vol.116, issue.6, pp.1372-1379, 2009.
DOI : 10.4049/jimmunol.180.1.214

C. Horlock, B. Stott, P. J. Dyson, M. Morishita, R. C. Coombes et al., The effects of trastuzumab on the CD4+CD25+FoxP3+ and CD4+IL17A+ T-cell axis in patients with breast cancer, British Journal of Cancer, vol.61, issue.7, pp.1061-1067, 2009.
DOI : 10.1038/nature06878

X. Qian, H. Chen, X. Wu, L. Hu, Q. Huang et al., Interleukin-17 acts as double-edged sword in anti-tumor immunity and tumorigenesis, Cytokine, vol.89
DOI : 10.1016/j.cyto.2015.09.011

E. Tartour, F. Fossiez, I. Joyeux, A. Galinha, A. Gey et al., Interleukin 17, a T-cell-derived cytokine, promotes tumorigenicity of human cervical tumors in nude mice, Cancer Res, vol.59, pp.3698-3704, 1999.

S. Punt, G. J. Fleuren, E. Kritikou, E. Lubberts, J. B. Trimbos et al., Angels and demons: Th17 cells represent a beneficial response, while neutrophil IL-17 is associated with poor prognosis in squamous cervical cancer, OncoImmunology, vol.59, issue.1
DOI : 10.1002/ijc.2910570608

URL : http://europepmc.org/articles/pmc4368129?pdf=render

D. E. Lyon, N. L. Mccain, J. Walter, and C. Schubert, Cytokine Comparisons Between Women With Breast Cancer and Women With a Negative Breast Biopsy, Nursing Research, vol.57, issue.1, pp.51-58, 2008.
DOI : 10.1097/01.NNR.0000280655.58266.6c

URL : http://europepmc.org/articles/pmc2234268?pdf=render

X. Zhu, L. A. Mulcahy, R. A. Mohammed, A. H. Lee, H. A. Franks et al., IL-17 expression by breast-cancer-associated macrophages: IL-17 promotes invasiveness of breast cancer cell lines, Breast Cancer Research, vol.67, issue.6, pp.95-106, 2008.
DOI : 10.1159/000028045

J. W. Du, K. Y. Xu, L. Y. Fang, and X. L. Qi, Interleukin-17, produced by lymphocytes, promotes tumor growth and angiogenesis in a mouse model of breast cancer, Molecular Medicine Reports, vol.6, issue.5, pp.1099-1102
DOI : 10.3892/mmr.2012.1036

S. Furuta, Y. M. Jeng, L. Zhou, L. Huang, I. Kuhn et al., IL-25 Causes Apoptosis of IL-25R-Expressing Breast Cancer Cells Without Toxicity to Nonmalignant Cells, Science Translational Medicine, vol.357, issue.1, 2011.
DOI : 10.1056/NEJMra043186

G. Kim, P. Khanal, S. C. Lim, H. J. Yun, S. G. Ahn et al., Interleukin-17 induces AP-1 activity and cellular transformation via upregulation of tumor progression locus 2 activity, Carcinogenesis, vol.70, issue.24, pp.341-350, 2013.
DOI : 10.1158/0008-5472.CAN-10-0775

S. Cochaud, J. Giustiniani, C. Thomas, E. Laprevotte, and C. Garbar, C

G. Alberici and N. Bonnefoy, IL-17A is produced by breast cancer TILs and promotes chemoresistance and proliferation through ERK1, Sci. Rep, vol.2013, issue.3, pp.3456-3466

S. Mombelli, S. Cochaud, Y. Merrouche, C. Garbar, F. Antonicelli et al., IL-17A and its homologs IL-25/IL-17E recruit the c-RAF/S6 kinase pathway and the generation of pro-oncogenic LMW-E in breast cancer cells, Scientific Reports, vol.1, issue.1, pp.11874-11884
DOI : 10.1016/S1568-7864(01)00019-2

URL : https://hal.archives-ouvertes.fr/hal-01841216

T. Benatar, M. Y. Cao, Y. Lee, J. Lightfoot, N. Feng et al., IL-17E, a proinflammatory cytokine, has antitumor efficacy against several tumor types in vivo, Cancer Immunology, Immunotherapy, vol.116, issue.6, pp.805-817, 2010.
DOI : 10.4049/jimmunol.176.2.1013

T. Benatar, M. Y. Cao, Y. Lee, H. Li, N. Feng et al., Virulizin?? induces production of IL-17E to enhance antitumor activity by recruitment of eosinophils into tumors, Cancer Immunology, Immunotherapy, vol.272, issue.12, pp.1757-1769, 2008.
DOI : 10.4049/jimmunol.178.7.4222

M. T. Duong, S. Akli, C. Wei, H. F. Wingate, W. Liu et al., LMW-E/CDK2 Deregulates Acinar Morphogenesis, Induces Tumorigenesis, and Associates with the Activated b-Raf-ERK1/2-mTOR Pathway in Breast Cancer Patients, PLoS Genetics, vol.34, issue.3, p.1002538, 2012.
DOI : 10.1371/journal.pgen.1002538.s012

D. Marzo, A. M. Marchi, V. L. Epstein, J. I. Nelson, and W. G. , Proliferative Inflammatory Atrophy of the Prostate, The American Journal of Pathology, vol.155, issue.6, pp.1985-1992, 1999.
DOI : 10.1016/S0002-9440(10)65517-4

Z. You, Y. Dong, X. Kong, Y. Zhang, R. L. Vessella et al., Differential Expression of IL-17RC Isoforms in Androgen-Dependent and Androgen-Independent Prostate Cancers, Neoplasia, vol.9, issue.6, pp.464-470, 2007.
DOI : 10.1593/neo.07109

Z. You, D. Ge, S. Liu, Q. Zhang, A. D. Borowsky et al., Interleukin-17 induces expression of chemokines and cytokines in prostatic epithelial cells but does not stimulate cell growth in vitro, Int. J. Med. Biol. Front, vol.18, pp.629-644, 2012.

S. Liu, Q. Zhang, C. Chen, D. Ge, Y. Qu et al., Hyperinsulinemia enhances interleukin-17-induced inflammation to promote prostate cancer development in obese mice through inhibiting glycogen synthase kinase 3-mediated phosphorylation and degradation of interleukin-17 receptor, Oncotarget, vol.7, issue.12, pp.13651-13666, 2016.
DOI : 10.18632/oncotarget.7296

Q. Zhang, S. Liu, D. Ge, Q. Zhang, Y. Xue et al., Interleukin-17 Promotes Formation and Growth of Prostate Adenocarcinoma in Mouse Models, Cancer Research, vol.72, issue.10, pp.2589-2599, 2012.
DOI : 10.1158/0008-5472.CAN-11-3795

Q. Zhang, S. Liu, K. R. Parajuli, W. Zhang, K. Zhang et al., Interleukin-17 promotes prostate cancer via MMP7-induced epithelial-to-mesenchymal transition, Oncogene, vol.26, issue.5, 2016.
DOI : 10.1093/bioinformatics/btp692

Y. Zhou, M. L. Toh, S. Zrioual, and P. Miossec, IL-17A versus IL-17F induced intracellular signal transduction pathways and modulation by IL-17RA and IL-17RC RNA interference in AGS gastric adenocarcinoma cells, Cytokine, vol.38, issue.3, pp.157-164, 2007.
DOI : 10.1016/j.cyto.2007.06.002

Q. Bie, C. Sun, A. Gong, C. Li, Z. Su et al., Non-tumor tissue derived interleukin-17B activates IL-17RB/AKT/??-catenin pathway to enhance the stemness of gastric cancer, Scientific Reports, vol.104, issue.1, pp.25447-25459
DOI : 10.1073/pnas.0611589104

B. Li, X. Y. Shi, D. X. Liao, B. R. Cao, C. H. Luo et al., Advanced colorectal adenoma related gene expression signature may predict prognostic for colorectal cancer patients with adenoma-carcinoma sequence, Int. J. Clin. Exp. Med, vol.8, pp.4883-4898, 2015.

G. Cui, A. Yuan, R. Goll, and J. Florholmen, IL-17A in the tumor microenvironment of the human colorectal adenoma???carcinoma sequence, Scandinavian Journal of Gastroenterology, vol.32, issue.4, pp.1304-1312
DOI : 10.1371/journal.pone.0034959

A. Al-samadi, S. Moossavi, A. Salem, M. Sotoudeh, S. M. Tuovinen et al., Distinctive expression pattern of interleukin-17 cytokine family members in colorectal cancer, Tumor Biology, vol.179, issue.10, pp.1609-1615, 2016.
DOI : 10.4049/jimmunol.179.10.7128

F. Housseau, S. Wu, E. C. Wick, H. Fan, X. Wu et al., Redundant Innate and Adaptive Sources of IL17 Production Drive Colon Tumorigenesis, Cancer Research, vol.76, issue.8, pp.2115-2124, 2016.
DOI : 10.1158/0008-5472.CAN-15-0749

B. Pan, D. Che, J. Cao, J. Shen, S. Jin et al., Interleukin-17 levels correlate with poor prognosis and vascular endothelial growth factor concentration in the serum of patients with non-small cell lung cancer, Biomarkers, vol.2014, issue.4, pp.232-239, 2015.
DOI : 10.1186/bcr2195

B. Pan, J. Shen, J. Cao, Y. Zhou, L. Shang et al., Interleukin-17 promotes angiogenesis by stimulating VEGF production of cancer cells via the STAT3/GIV signaling pathway in non-small-cell lung cancer, Scientific Reports, vol.292, issue.1, pp.16053-16066, 2015.
DOI : 10.1016/j.canlet.2009.11.013

M. Numasaki, M. Watanabe, T. Suzuki, H. Takahashi, A. Nakamura et al., IL-17 Enhances the Net Angiogenic Activity and In Vivo Growth of Human Non-Small Cell Lung Cancer in SCID Mice through Promoting CXCR-2-Dependent Angiogenesis, The Journal of Immunology, vol.175, issue.9, pp.6177-6189, 2005.
DOI : 10.4049/jimmunol.175.9.6177

D. He, H. Li, N. Yusuf, C. A. Elmets, M. Athar et al., IL-17 Mediated Inflammation Promotes Tumor Growth and Progression in the Skin, PLoS ONE, vol.10, issue.2, p.32126, 2012.
DOI : 10.1371/journal.pone.0032126.g005

URL : https://doi.org/10.1371/journal.pone.0032126

F. Mcallister and J. Kolls, Th17 cytokines in non-melanoma skin cancer, European Journal of Immunology, vol.41, issue.3, pp.692-694
DOI : 10.1016/j.immuni.2014.11.009

L. Wu, X. Chen, J. Zhao, B. Martin, J. A. Zepp et al., A novel IL-17 signaling pathway controlling keratinocyte proliferation and tumorigenesis via the TRAF4???ERK5 axis, The Journal of Experimental Medicine, vol.212, issue.10, pp.1571-1587, 2015.
DOI : 10.1038/nm1720

L. Wang, T. Yi, M. Kortylewski, D. M. Pardoll, D. Zeng et al., IL-17 can promote tumor growth through an IL-6???Stat3 signaling pathway, The Journal of Experimental Medicine, vol.206, issue.7, pp.1457-1464, 2009.
DOI : 10.1016/j.jhep.2008.12.033

URL : http://jem.rupress.org/content/jem/206/7/1457.full.pdf

M. Doroudchi, Z. G. Pishe, M. Malekzadeh, H. Golmoghaddam, M. Taghipour et al., Elevated serum IL-17A but not IL-6 in glioma versus meningioma and schwannoma. Asian Pac, J. Cancer Prev, vol.14, pp.5225-5230, 2013.
DOI : 10.7314/apjcp.2013.14.9.5225

URL : http://ocean.kisti.re.kr/downfile/volume/apocp/POCPA9/2013/v14n9/POCPA9_2013_v14n9_5225.pdf

A. Kehlen, K. Thiele, D. Riemann, N. Rainov, and J. Langner, Interleukin-17 stimulates the expression of I??B?? mRNA and the secretion of IL-6 and IL-8 in glioblastoma cell lines, Journal of Neuroimmunology, vol.101, issue.1, pp.1-6, 1999.
DOI : 10.1016/S0165-5728(99)00111-3

D. A. Wainwright, S. Sengupta, Y. Han, I. V. Ulasov, and M. S. Lesniak, The Presence of IL-17A and T Helper 17 Cells in Experimental Mouse Brain Tumors and Human Glioma, PLoS ONE, vol.12, issue.10, p.15390, 2010.
DOI : 10.1371/journal.pone.0015390.t002

G. Cantini, F. Pisati, A. Mastropietro, V. Frattini, Y. Iwakura et al., A critical role for regulatory T cells in driving cytokine profiles of Th17 cells and their modulation of glioma microenvironment, Cancer Immunology, Immunotherapy, vol.125, issue.12, pp.1739-1750, 2011.
DOI : 10.1002/ijc.24497

J. Hu, H. Ye, D. Zhang, W. Liu, M. Li et al., U87MG glioma cells overexpressing IL-17 acclerate early-stage growth in vivo and cause a higher level of CD31 mRNA expression in tumor tissues, Oncology Letters, vol.6, issue.4, pp.993-999
DOI : 10.3892/ol.2013.1518

H. Liang, L. Yi, X. Wang, C. Zhou, and L. Xu, Interleukin-17 Facilitates the Immune Suppressor Capacity of High-Grade Glioma-Derived CD4 (+) CD25 (+) Foxp3 (+) T Cells Via Releasing Transforming Growth Factor Beta, Scandinavian Journal of Immunology, vol.72, issue.2, pp.144-150, 2014.
DOI : 10.1136/annrheumdis-2012-202201

URL : http://onlinelibrary.wiley.com/doi/10.1111/sji.12185/pdf

P. Parajuli, R. Anand, C. Mandalaparty, R. Suryadevara, P. U. Sriranga et al., Preferential expression of functional IL-17R in glioma stem cells: potential role in self-renewal, Oncotarget, vol.7, issue.5, pp.6121-6135, 2016.
DOI : 10.18632/oncotarget.6847

J. P. Zhang, J. Yan, J. Xu, X. H. Pang, M. S. Chen et al., Increased intratumoral IL-17-producing cells correlate with poor survival in hepatocellular carcinoma patients, Journal of Hepatology, vol.50, issue.5, pp.980-989, 2009.
DOI : 10.1016/j.jhep.2008.12.033

D. M. Kuang, C. Peng, Q. Zhao, and Y. Wu,

, J. Immunol, vol.185, pp.1544-1549, 2010.

M. D. Sternlicht, A. Lochter, C. J. Sympson, B. Huey, J. P. Rougier et al., The Stromal Proteinase MMP3/Stromelysin-1 Promotes Mammary Carcinogenesis, Cell, vol.98, issue.2, pp.137-146, 1999.
DOI : 10.1016/S0092-8674(00)81009-0

M. L. Stallings-mann, J. Waldmann, Y. Zhang, E. Miller, M. L. Gauthier et al., Matrix Metalloproteinase Induction of Rac1b, a Key Effector of Lung Cancer Progression, Science Translational Medicine, vol.283, issue.2, 2012.
DOI : 10.1152/ajplung.00302.2001

Y. Ma, L. Aymeric, C. Locher, S. R. Mattarollo, N. F. Delahaye et al., 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

O. Sullivan, T. Saddawi-konefka, R. Gross, E. Tran, M. Mayfield et al., Interleukin-17D mediates tumor rejection through recruitment of natural killer cells. Cell Rep, pp.989-998, 2014.