N. Bossard, M. Velten, and L. Remontet, Survival of cancer patients in France: A population-based study from The Association of the French Cancer Registries (FRANCIM), European Journal of Cancer, vol.43, issue.1, 2007.
DOI : 10.1016/j.ejca.2006.07.021

URL : https://hal.archives-ouvertes.fr/hal-00434567

V. Jooste, L. Remontet, and M. Colonna, Trends in the incidence of digestive cancers in France between 1980 and 2005 and projections for the year 2010, European Journal of Cancer Prevention, vol.20, issue.5, pp.375-380, 2011.
DOI : 10.1097/CEJ.0b013e3283465426

URL : https://hal.archives-ouvertes.fr/hal-00697965

P. Favoriti, G. Carbone, M. Greco, F. Pirozzi, R. Pirozzi et al., Worldwide burden of colorectal cancer: a review, Updates in Surgery, vol.45, issue.8, 2016.
DOI : 10.1016/j.ejca.2008.11.018

E. Sanz-garcia, J. Grasselli, G. Argiles, M. Elez, and J. Tabernero, Current and advancing treatments for metastatic colorectal cancer, Expert Opinion on Biological Therapy, vol.25, issue.4, pp.93-110, 2016.
DOI : 10.1016/j.molonc.2014.05.003

T. Boehm, J. Folkman, T. Browder, O. Reilly, and M. , Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance, Nature, vol.92, issue.6658, pp.404-40710, 1997.
DOI : 10.1073/pnas.92.10.4562

M. Kieran, R. Kalluri, and Y. Cho, The VEGF pathway in cancer and disease: responses, resistance, and the path forward. Cold Spring Harb Perspect Med, 2012.

G. Mirone, A. Shukla, and G. Marfe, Signaling mechanisms of resistance to EGFR-and

F. Fan, S. Samuel, and P. Gaur, Chronic exposure of colorectal cancer cells to bevacizumab promotes compensatory pathways that mediate tumour cell migration, British Journal of Cancer, vol.59, issue.8, pp.1270-1277, 2011.
DOI : 10.1002/cncr.21145

N. Yamagishi, S. Teshima-kondo, and K. Masuda, Chronic inhibition of tumor cell-derived VEGF enhances the malignant phenotype of colorectal cancer cells, BMC Cancer, vol.8, issue.10, pp.229-239, 2013.
DOI : 10.1586/14737140.8.10.1545

P. Mésange, V. Poindessous, M. Sabbah, A. Escargueil, A. De-gramont et al., Intrinsic bevacizumab resistance is associated with prolonged activation of autocrine VEGF signaling and hypoxia tolerance in colorectal cancer cells and can be overcome by nintedanib, a small molecule angiokinase inhibitor., Oncotarget, vol.5, issue.13, pp.4709-4721, 2014.
DOI : 10.18632/oncotarget.1671

S. Gaffen, R. Jain, A. V. Garg, and D. Cua, The IL-23???IL-17 immune axis: from mechanisms to therapeutic testing, Nature Reviews Immunology, vol.24, issue.9, pp.585-600, 2014.
DOI : 10.1093/cid/cit722

W. Ouyang, J. Kolls, and Y. Zheng, The Biological Functions of T Helper 17 Cell Effector Cytokines in Inflammation, Immunity, vol.28, issue.4, pp.454-467, 2008.
DOI : 10.1016/j.immuni.2008.03.004

L. Steinman, A brief history of TH17, the first major revision in the TH1/TH2 hypothesis of T cell???mediated tissue damage, Nature Medicine, vol.59, issue.2, pp.139-145, 1551.
DOI : 10.4049/jimmunol.165.11.6107

B. Yang, H. Kang, A. Fung, H. Zhao, T. Wang et al., The Role of Interleukin 17 in Tumour Proliferation, Angiogenesis, and Metastasis, Mediators of Inflammation, vol.31, issue.1, article 795, pp.623759-623769, 2014.
DOI : 10.1016/j.leukres.2013.06.022

W. Zou and N. Restifo, TH17 cells in tumour immunity and immunotherapy, Nature Reviews Immunology, vol.8, issue.4, pp.248-256, 2010.
DOI : 10.4049/jimmunol.174.5.2591

P. Miossec and J. Kolls, Targeting IL-17 and TH17 cells in chronic inflammation, Nature Reviews Drug Discovery, vol.56, issue.10, pp.763-776, 2012.
DOI : 10.1002/art.22733

G. Murugaiyan and B. Saha, Protumor vs Antitumor Functions of IL-17, The Journal of Immunology, vol.183, issue.7, pp.4169-4175, 2009.
DOI : 10.4049/jimmunol.0901017

Y. Iwakura, H. Ishigame, S. Saijo, and S. Nakae, Functional Specialization of Interleukin-17 Family Members, Immunity, vol.34, issue.2, pp.149-162, 2011.
DOI : 10.1016/j.immuni.2011.02.012

K. Hirota, H. Ahlfors, J. Duarte, and B. Stockinger, Regulation and function of innate and adaptive interleukin-17-producing cells, EMBO reports, vol.207, issue.2, 2011.
DOI : 10.1084/jem.20100703

S. Chang and C. Dong, IL-17F: Regulation, signaling and function in inflammation, Cytokine, vol.46, issue.1, pp.7-11, 2009.
DOI : 10.1016/j.cyto.2008.12.024

L. Gouvello, S. Bastuji-garin, S. Aloulou, and N. , High prevalence of Foxp3 and IL17 in MMR-proficient colorectal carcinomas, Gut, vol.57, issue.6, pp.772-779, 2008.
DOI : 10.1136/gut.2007.123794

URL : https://hal.archives-ouvertes.fr/inserm-00284889

I. Georgiades, L. Curtis, R. Morris, C. Bird, and A. Wyllie, Heterogeneity studies identify a subset of sporadic colorectal cancers without evidence for chromosomal or microsatellite instability, Oncogene, vol.18, issue.56, pp.7933-7940, 2000.
DOI : 10.1002/(SICI)1097-0215(19990301)80:5<667::AID-IJC6>3.0.CO;2-0

R. Tang, C. Changchien, and M. Wu, Colorectal cancer without high microsatellite instability and chromosomal instability--an alternative genetic pathway to human colorectal cancer, Carcinogenesis, vol.25, issue.5, pp.841-846, 2003.
DOI : 10.1093/carcin/bgh074

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, 2012.
DOI : 10.1371/journal.pone.0034959

J. Liu, Y. Duan, and X. Cheng, IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma, Biochemical and Biophysical Research Communications, vol.407, issue.2, pp.348-354, 2011.
DOI : 10.1016/j.bbrc.2011.03.021

G. Radosavljevic, B. Ljujic, and I. Jovanovic, Interleukin-17 may be a valuable serum tumor marker in patients with colorectal carcinoma, Neoplasma, vol.57, issue.2, pp.135-144, 2010.
DOI : 10.4149/neo_2010_02_135

J. Wang, K. Xu, and J. Wu, The changes of Th17 cells and the related cytokines in the progression of human colorectal cancers, BMC Cancer, vol.117, issue.1, pp.1471-2407, 2012.
DOI : 10.1182/blood-2010-11-317321

E. Lereclus, M. Tout, and A. Girault, A possible association of baseline serum IL-17A concentrations with progression-free survival of metastatic colorectal cancer patients treated with a bevacizumab-based regimen, BMC Cancer, vol.4, issue.2
DOI : 10.4161/2162402X.2014.984547

M. Tosolini, A. Kirilovsky, and B. Mlecnik, Clinical Impact of Different Classes of Infiltrating T Cytotoxic and Helper Cells (Th1, Th2, Treg, Th17) in Patients with Colorectal Cancer, Cancer Research, vol.71, issue.4, pp.1263-1271, 2011.
DOI : 10.1158/0008-5472.CAN-10-2907

J. Tseng, C. Yang, and S. Liang, Interleukin-17A Modulates Circulating Tumor Cells in Tumor Draining Vein of Colorectal Cancers and Affects Metastases, Clinical Cancer Research, vol.20, issue.11, pp.2885-2897, 2014.
DOI : 10.1158/1078-0432.CCR-13-2162

K. Wang, M. Kim, D. Caro, and G. , Interleukin-17 Receptor A Signaling in Transformed Enterocytes Promotes Early Colorectal Tumorigenesis, Immunity, vol.41, issue.6, 2014.
DOI : 10.1016/j.immuni.2014.11.009

A. Chung, X. Wu, and G. Zhuang, An interleukin-17???mediated paracrine network promotes tumor resistance to anti-angiogenic therapy, Nature Medicine, vol.59, issue.9, pp.1114-1123, 2013.
DOI : 10.1177/002215540305100503

Y. Zhao, T. Wu, S. Shao, B. Shi, and Y. Zhao, Phenotype, development, and biological function of myeloid-derived suppressor cells, OncoImmunology, vol.120, issue.2, 2016.
DOI : 10.4049/jimmunol.0804253

M. Manjili, X. Wang, and S. Abrams, Evolution of Our Understanding of Myeloid Regulatory Cells: From MDSCs to Mregs, Frontiers in Immunology, vol.1, issue.4, 2014.
DOI : 10.3892/mco.2013.134

N. Obermajer, J. Wong, and R. Edwards, Induction and stability of human Th17 cells require endogenous NOS2 and cGMP-dependent NO signaling, The Journal of Experimental Medicine, vol.65, issue.7, pp.1433-1445, 2013.
DOI : 10.1038/nri2742

Z. Xie, Y. Qu, and Y. Leng, Human colon carcinogenesis is associated with increased interleukin-17-driven inflammatory responses, Drug Design, Development and Therapy, issue.9, pp.1679-1689, 2015.
DOI : 10.2147/DDDT.S79431

L. Wang, T. Yi, M. Kortylewski, D. 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

M. Numasaki, J. Fukushi, and M. Ono, Interleukin-17 promotes angiogenesis and tumor growth, Blood, vol.101, issue.7, pp.2620-262710, 2003.
DOI : 10.1182/blood-2002-05-1461

D. Anh, . Van, S. Park, and H. Lee, The Membrane-Bound Form of IL-17A Promotes the Growth and Tumorigenicity of Colon Cancer Cells, Molecules and Cells, vol.39, issue.7, pp.536-542, 2016.
DOI : 10.14348/molcells.2016.0048

J. Chen, J. Xia, and X. Liang, Intratumoral Expression of IL-17 and Its Prognostic Role in Gastric Adenocarcinoma Patients, International Journal of Biological Sciences, vol.7, issue.1, pp.53-60, 2011.
DOI : 10.7150/ijbs.7.53

I. Kryczek, S. Wei, W. Szeliga, L. Vatan, and W. Zou, Endogenous IL-17 contributes to reduced tumor growth and metastasis, Blood, vol.114, issue.2, pp.357-359, 2009.
DOI : 10.1182/blood-2008-09-177360

X. Qian, H. Chen, X. Wu, L. Hu, Q. Huang et al., Interleukin-17 acts as double-edged sword in antitumor immunity and tumorigenesis

D. Simone, V. Pallone, F. Monteleone, G. Stolfi, and C. , 17 cytokines in the control of colorectal cancer, OncoImmunology, vol.29, issue.12, pp.26617-26627, 2013.
DOI : 10.1016/j.ccr.2013.06.017

J. Ye, R. Livergood, and G. Peng, The Role and Regulation of Human Th17 Cells in Tumor Immunity, The American Journal of Pathology, vol.182, issue.1
DOI : 10.1016/j.ajpath.2012.08.041

D. Mcgovern, J. Rotter, and L. Mei, Genetic epistasis of IL23/IL17 pathway genes in Crohn??s disease, Inflammatory Bowel Diseases, vol.15, issue.6, pp.883-889, 2009.
DOI : 10.1002/ibd.20855

R. Hayashi, T. Tahara, and H. Shiroeda, Influence of IL17A polymorphisms (rs2275913 and rs3748067) on the susceptibility to ulcerative colitis, Clinical and Experimental Medicine, vol.39, issue.4, pp.239-244, 2013.
DOI : 10.1038/ng2135

J. Eaden, K. Abrams, and J. Mayberry, The risk of colorectal cancer in ulcerative colitis: a meta-analysis, Gut, vol.48, issue.4, pp.526-535, 2001.
DOI : 10.1136/gut.48.4.526

X. Liu, X. Lin, and S. Gaffen, Crucial Role for Nuclear Factor of Activated T Cells in T Cell Receptor-mediated Regulation of Human Interleukin-17, Journal of Biological Chemistry, vol.12, issue.50, pp.52762-52771, 2004.
DOI : 10.1016/S1471-4906(01)01973-1

M. Kawaguchi, D. Takahashi, and N. Hizawa, IL-17F sequence variant (His161Arg) is associated with protection against asthma and antagonizes wild-type IL-17F activity, Journal of Allergy and Clinical Immunology, vol.117, issue.4, pp.795-801, 2006.
DOI : 10.1016/j.jaci.2005.12.1346

Y. Duan, J. Shi, C. Pan, H. Chen, and S. Zhang, Association between the Interleukin-17A -197G>A (rs2275913) Polymorphism and Risk of Digestive Cancer, Asian Pacific Journal of Cancer Prevention, vol.15, issue.21, pp.9295-9300, 2014.
DOI : 10.7314/APJCP.2014.15.21.9295

A. Rafiei, V. Hosseini, and G. Janbabai, Polymorphism in the interleukin-17A promoter contributes to gastric cancer, World Journal of Gastroenterology, vol.19, issue.34, pp.5693-5699, 2013.
DOI : 10.3748/wjg.v19.i34.5693

X. Wu, Z. Zeng, and B. Chen, Association between polymorphisms in interleukin-17A and interleukin-17F genes and risks of gastric cancer, International Journal of Cancer, vol.69, issue.1, pp.86-92, 2010.
DOI : 10.1158/0008-5472.CAN-09-0596

X. Zhang, L. Zheng, Y. Sun, and X. Zhang, Analysis of the association of interleukin-17 gene polymorphisms with gastric cancer risk and interaction with Helicobacter pylori infection in a Chinese population, Tumor Biology, vol.124, issue.7, pp.1575-1580, 2014.
DOI : 10.1016/S0016-5085(03)00157-4

Z. Dai, T. Zhang, and S. Lin, Role of IL-17A rs2275913 and IL-17F rs763780 polymorphisms in risk of cancer development: an updated meta-analysis, Scientific Reports, vol.21, issue.1, 2016.
DOI : 10.1002/sim.1186

K. Nemati, H. Golmoghaddam, S. V. Hosseini, A. Ghaderi, and M. Doroudchi, Interleukin-17FT7488 allele is associated with a decreased risk of colorectal cancer and tumor progression, Gene, vol.561, issue.1, pp.88-94, 2015.
DOI : 10.1016/j.gene.2015.02.014

I. Omrane, R. Marrakchi, and O. Baroudi, Significant association between interleukin-17A polymorphism and colorectal cancer, Tumor Biology, vol.7, issue.4, pp.6627-6632, 2014.
DOI : 10.1371/journal.pone.0034959

Y. Lu, J. Gu, and H. Lu, Association Between IL-17A +197 G/A Polymorphism and Cancer Risk: A Meta-Analysis, Genetic Testing and Molecular Biomarkers, vol.20, issue.1, pp.24-30, 2016.
DOI : 10.1089/gtmb.2015.0143

M. Lowes, A. Bowcock, and J. Krueger, Pathogenesis and therapy of psoriasis, Nature, vol.126, issue.7130, pp.866-873, 2007.
DOI : 10.4049/jimmunol.177.7.4917

C. Chao, S. Chen, and I. Adamopoulos, Anti-IL-17A therapy protects against bone erosion in experimental models of rheumatoid arthritis, Autoimmunity, vol.8, issue.3, pp.243-252, 2011.
DOI : 10.1186/ar1872

W. Hueber, D. Patel, and T. Dryja, Effects of AIN457, a Fully Human Antibody to Interleukin-17A, on Psoriasis, Rheumatoid Arthritis, and Uveitis, Science Translational Medicine, vol.94, issue.1, 2010.
DOI : 10.1016/0002-9394(82)90197-0

K. Gordon, A. Blauvelt, and K. Papp, Phase 3 Trials of Ixekizumab in Moderate-to-Severe Plaque Psoriasis, New England Journal of Medicine, vol.375, issue.4, pp.345-35610, 2016.
DOI : 10.1056/NEJMoa1512711

M. Lebwohl, B. Strober, and A. Menter, Phase 3 Studies Comparing Brodalumab with Ustekinumab in Psoriasis, New England Journal of Medicine, vol.373, issue.14, pp.1318-1328, 2015.
DOI : 10.1056/NEJMoa1503824

R. Prabhala, M. Fulciniti, and D. Pelluru, Targeting IL-17A in multiple myeloma: a potential novel therapeutic approach in myeloma, Leukemia, vol.45, issue.2, pp.379-389, 2016.
DOI : 10.1056/NEJMoa1109017

Y. Ma, C. Chen, and D. Li, Targeting of interleukin (IL)-17A inhibits PDL1 expression in tumor cells and induces anticancer immunity in an estrogen receptor-negative murine model of breast cancer, Oncotarget, 2016.
DOI : 10.18632/oncotarget.13819