J. Ferlay, M. Colombet, I. Soerjomataram, T. Dyba, G. Randi et al., Cancer incidence and mortality patterns in europe: Estimates for 40 countries and 25 major cancers in 2018, Eur. J. Cancer, vol.103, pp.356-387, 2018.

E. Senkus, S. Kyriakides, S. Ohno, F. Penault-llorca, P. Poortmans et al., Primary breast cancer: Esmo clinical practice guidelines for diagnosis, treatment and follow-up, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol./ESMO, vol.26, 2015.

K. Altundag, Have all triple-negative breast cancer patients worse breast cancer-specific survival?, Breast, vol.36, 2017.

Y. Feng, M. Spezia, S. Huang, C. Yuan, Z. Zeng et al., Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis, Genes Dis, vol.5, pp.77-106, 2018.

S. Lal, A. E. Mccart-reed, X. M. De-luca, and P. T. Simpson, Molecular signatures in breast cancer, vol.131, pp.135-146, 2017.

T. Sorlie, C. M. Perou, R. Tibshirani, T. Aas, S. Geisler et al., Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications, Proc. Natl. Acad. Sci, vol.98, pp.10869-10874, 2001.

C. M. Perou, T. Sorlie, M. B. Eisen, M. Van-de-rijn, S. S. Jeffrey et al., Molecular portraits of human breast tumours, Nature, vol.406, pp.747-752, 2000.

H. G. Russnes, O. C. Lingjaerde, A. L. Borresen-dale, and C. Caldas, Breast cancer molecular stratification: From intrinsic subtypes to integrative clusters, Am. J. Pathol, vol.187, pp.2152-2162, 2017.

F. Cardoso, E. Senkus, A. Costa, E. Papadopoulos, M. Aapro et al., 4th eso-esmo international consensus guidelines for advanced breast cancer (abc 4)dagger, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol./ESMO, vol.29, pp.1634-1657, 2018.

T. J. Ballinger, J. B. Meier, and V. M. Jansen, Current landscape of targeted therapies for hormone-receptor positive, her2 negative metastatic breast cancer, Front. Oncol, vol.8, 2018.

T. Westphal, S. P. Gampenrieder, G. Rinnerthaler, and R. Greil, Cure in metastatic breast cancer, vol.11, pp.172-179, 2018.

L. G. Eng, S. Dawood, V. Sopik, B. Haaland, P. S. Tan et al., Ten-year survival in women with primary stage iv breast cancer, Breast Cancer Res. Treat, vol.160, pp.145-152, 2016.

L. Y. Dirix, I. Takacs, G. Jerusalem, P. Nikolinakos, H. T. Arkenau et al., Avelumab, an anti-pd-l1 antibody, in patients with locally advanced or metastatic breast cancer: A phase 1b javelin solid tumor study, Breast Cancer Res. Treat, vol.167, pp.671-686, 2018.

L. Wein, S. J. Luen, P. Savas, R. Salgado, and S. Loi, Checkpoint blockade in the treatment of breast cancer: Current status and future directions, Br. J. Cancer, vol.119, pp.4-11, 2018.

L. Hui and Y. Chen, Tumor microenvironment: Sanctuary of the devil, Cancer Lett, vol.368, pp.7-13, 2015.

C. Genova, G. Rossi, E. Rijavec, F. Biello, G. Barletta et al., Releasing the brake: Safety profile of immune check-point inhibitors in non-small cell lung cancer, Expert Opin. Drug Saf, vol.16, pp.573-585, 2017.

P. A. Ascierto and G. A. Mcarthur, Checkpoint inhibitors in melanoma and early phase development in solid tumors: What's the future?, J. Transl. Med, vol.15, 2017.

P. Samadi, S. Saki, F. K. Dermani, M. Pourjafar, and M. Saidijam, Emerging ways to treat breast cancer: Will promises be met?, Cell. Oncol. (Dordr.), vol.41, pp.605-621, 2018.

F. Balkwill and A. Mantovani, Inflammation and cancer: Back to virchow?, Lancet, vol.357, pp.539-545, 2001.

C. I. Diakos, K. A. Charles, D. C. Mcmillan, and S. J. Clarke, Cancer-related inflammation and treatment effectiveness, Lancet Oncol, vol.15, pp.493-503, 2014.

D. G. Denardo and L. M. Coussens, Balancing immune response: Crosstalk between adaptive and innate immune cells during breast cancer progression, Breast Cancer Res, vol.9, 2007.

S. Setrerrahmane and H. Xu, Tumor-related interleukins: Old validated targets for new anti-cancer drug development, Mol. Cancer, vol.16, 2017.

Y. J. Sloot, J. W. Smit, L. A. Joosten, and R. T. Netea-maier, Insights into the role of il-32 in cancer, Semin. Immunol, 2018.

I. Holen, D. V. Lefley, S. E. Francis, S. Rennicks, S. Bradbury et al., Il-1 drives breast cancer growth and bone metastasis in vivo, Oncotarget, vol.7, pp.75571-75584, 2016.

T. H. Heo, J. Wahler, and N. Suh, Potential therapeutic implications of il-6/il-6r/gp130-targeting agents in breast cancer, Oncotarget, vol.7, pp.15460-15473, 2016.

N. Amatya, A. V. Garg, and S. L. Gaffen, Il-17 signaling: The yin and the yang, Trends Immunol, vol.38, pp.310-322, 2017.

Y. Song and J. M. Yang, Role of interleukin (il)-17 and t-helper (th)17 cells in cancer, Biochem. Biophys. Res. Commun, vol.493, pp.1-8, 2017.

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.

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.

S. L. Gaffen, Life before seventeen: Cloning of the il-17 receptor, J. Immunol, vol.187, pp.4389-4391, 2011.

N. Isailovic, K. Daigo, A. Mantovani, and C. Selmi, Interleukin-17 and innate immunity in infections and chronic inflammation, J. Autoimmun, vol.60, pp.1-11, 2015.

J. Fabre, J. Giustiniani, C. Garbar, F. Antonicelli, Y. Merrouche et al., Targeting the tumor microenvironment: The protumor effects of il-17 related to cancer type, Int. J. Mol. Sci, vol.17, 1433.
URL : https://hal.archives-ouvertes.fr/hal-01841211

J. Fabre, J. Giustiniani, F. Antonicelli, Y. Merrouche, M. Bagot et al., A focus on il-17 targeting treatments in psoriasis, J. Dermatol. Res, vol.2, pp.1-5, 2016.

D. Giusti, S. Le-jan, G. Gatouillat, P. Bernard, B. N. Pham et al., Biomarkers related to bullous pemphigoid activity and outcome, Exp. Dermatol, vol.26, pp.1240-1247, 2017.

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.34, pp.341-350, 2013.

S. Le-jan, J. Plee, D. Vallerand, A. Dupont, E. Delanez et al., Innate immune cell-produced il-17 sustains inflammation in bullous pemphigoid, J. Investig. Dermatol, vol.134, pp.2908-2917, 2014.

J. Plee, S. Le-jan, J. Giustiniani, C. Barbe, P. Joly et al., Integrating longitudinal serum il-17 and il-23 follow-up, along with autoantibodies variation, contributes to predict bullous pemphigoid outcome

Z. Fan, J. Yang, C. Yang, and X. Guo, Il-17: A promising therapeutic target for atherosclerosis, Int. J. Cardiol, vol.202, pp.930-931, 2016.

F. Fossiez, O. Djossou, P. Chomarat, L. Flores-romo, S. Ait-yahia et al., T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines, J. Exp. Med, vol.183, pp.2593-2603, 1996.

S. G. Hymowitz, E. H. Filvaroff, J. P. Yin, J. Lee, L. Cai et al., Il-17s adopt a cystine knot fold: Structure and activity of a novel cytokine, il-17f, and implications for receptor binding, EMBO J, vol.20, pp.5332-5341, 2001.

N. Q. Mcdonald and W. A. Hendrickson, A structural superfamily of growth factors containing a cystine knot motif, Cell, vol.73, pp.421-424, 1993.

T. A. Moseley, D. R. Haudenschild, L. Rose, and A. H. Reddi, Interleukin-17 family and il-17 receptors, Cytokine Growth Factor Rev, vol.14, pp.155-174, 2003.

S. H. Chang and C. Dong, A novel heterodimeric cytokine consisting of il-17 and il-17f regulates inflammatory responses, Cell Res, vol.17, pp.435-440, 2007.

J. F. Wright, Y. Guo, A. Quazi, D. P. Luxenberg, F. Bennett et al., Identification of an interleukin 17f/17a heterodimer in activated human cd4+ t cells, J. Biol. Chem, vol.282, pp.13447-13455, 2007.

X. D. Huang, H. Zhang, and M. X. He, Comparative and evolutionary analysis of the interleukin 17 gene family in invertebrates, PLoS ONE, vol.10, 2015.

J. K. Kolls and A. Linden, Interleukin-17 family members and inflammation, Immunity, vol.21, pp.467-476, 2004.

Y. Iwakura, H. Ishigame, S. Saijo, and S. Nakae, Functional specialization of interleukin-17 family members, Immunity, vol.34, pp.149-162, 2011.

Q. Bie, C. Jin, B. Zhang, and H. Dong, Il-17b: A new area of study in the il-17 family, Mol. Immunol, vol.90, pp.50-56, 2017.

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 alpha-helix for act1 binding and il-17 signaling, Acta Crystallogr. D Biol. Crystallogr, vol.70, pp.1476-1483, 2014.

D. D. Patel, D. M. Lee, F. Kolbinger, and C. Antoni, Effect of il-17a blockade with secukinumab in autoimmune diseases, Ann. Rheum. Dis, vol.72, pp.116-123, 2013.

J. F. Wright, F. Bennett, B. Li, J. Brooks, D. P. Luxenberg et al., The human il-17f/il-17a heterodimeric cytokine signals through the il-17ra/il-17rc receptor complex, J. Immunol, vol.181, pp.2799-2805, 2008.

S. Zrioual, M. L. Toh, A. Tournadre, Y. Zhou, M. A. Cazalis et al., Il-17ra and il-17rc receptors are essential for il-17a-induced elr+ cxc chemokine expression in synoviocytes and are overexpressed in rheumatoid blood, J. Immunol, vol.180, pp.655-663, 2008.

R. E. Kuestner, D. W. Taft, A. Haran, C. S. Brandt, T. Brender et al., Identification of the il-17 receptor related molecule il-17rc as the receptor for il-17f, J. Immunol, vol.179, pp.5462-5473, 2007.

L. Monin and S. L. Gaffen, Interleukin 17 family cytokines: Signaling mechanisms, biological activities, and therapeutic implications, Cold Spring Harb. Perspect. Biol, vol.10, 2018.

T. R. Mosmann, H. Cherwinski, M. W. Bond, M. A. Giedlin, and R. L. Coffman, Two types of murine helper t cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins, J. Immunol, vol.136, pp.2348-2357, 1986.

T. Aarvak, M. Chabaud, P. Miossec, and J. Natvig, Il-17 is produced by some proinflammatory th1/th0 cells but not by th2 cells, J. Immunol, vol.162, pp.1246-1251, 1999.

L. Steinman, A brief history of t(h)17, the first major revision in the t(h)1/t(h)2 hypothesis of t cell-mediated tissue damage, Nat. Med, vol.13, pp.139-145, 2007.

H. Park, Z. Li, X. O. Yang, S. H. Chang, R. Nurieva et al., A distinct lineage of cd4 t cells regulates tissue inflammation by producing interleukin 17, Nat. Immunol, vol.6, pp.1133-1141, 2005.

T. Korn, M. Oukka, V. Kuchroo, and E. Bettelli, Th17 cells: Effector t cells with inflammatory properties, Semin. Immunol, vol.19, pp.362-371, 2007.

S. C. Liang, X. Y. Tan, D. P. Luxenberg, R. Karim, K. Dunussi-joannopoulos et al., Interleukin (il)-22 and il-17 are coexpressed by th17 cells and cooperatively enhance expression of antimicrobial peptides, J. Exp. Med, vol.203, pp.2271-2279, 2006.

R. Nurieva, X. O. Yang, G. Martinez, Y. Zhang, A. D. Panopoulos et al., Essential autocrine regulation by il-21 in the generation of inflammatory t cells, Nature, vol.448, pp.480-483, 2007.

J. A. Smith, The bench-to-bedside story of il-17 and the therapeutic efficacy of its targeting in spondyloarthritis, Curr. Rheumatol. Rep, vol.18, p.33, 2016.

M. Veldhoen, R. J. Hocking, C. J. Atkins, R. M. Locksley, and B. Stockinger, Tgfbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of il-17-producing t cells, Immunity, vol.24, pp.179-189, 2006.

J. Bystrom, T. E. Taher, M. S. Muhyaddin, F. I. Clanchy, P. Mangat et al., Harnessing the therapeutic potential of th17 cells, Med. Inflamm, 2015.

M. Ciofani, A. Madar, C. Galan, M. Sellars, K. Mace et al., A validated regulatory network for th17 cell specification, Cell, vol.151, pp.289-303, 2012.

M. Huber, S. Heink, A. Pagenstecher, K. Reinhard, J. Ritter et al., Il-17a secretion by cd8+ t cells supports th17-mediated autoimmune encephalomyelitis, J. Clin. Investig, vol.123, pp.247-260, 2013.

M. Kronenberg, Toward an understanding of nkt cell biology: Progress and paradoxes, Annu. Rev. Immunol, vol.23, pp.877-900, 2005.

B. R. Marks, H. N. Nowyhed, J. Y. Choi, A. C. Poholek, J. M. Odegard et al., Thymic self-reactivity selects natural interleukin 17-producing t cells that can regulate peripheral inflammation, Nat. Immunol, vol.10, pp.1125-1132, 2009.

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.421, pp.744-748, 2003.

T. Noordenbos, I. Blijdorp, S. Chen, J. Stap, E. Mul et al., Human mast cells capture, store, and release bioactive, exogenous il-17a, J. Leukoc. Biol, vol.100, pp.453-462, 2016.

A. M. Lin, C. J. Rubin, R. Khandpur, J. Y. Wang, M. Riblett et al., Mast cells and neutrophils release il-17 through extracellular trap formation in psoriasis, J. Immunol, vol.187, pp.490-500, 2011.

K. Wang and M. Karin, The il-23 to il-17 cascade inflammation-related cancers, Clin. Exp. Rheumatol, vol.33, pp.87-90, 2015.

H. Morita, K. Arae, H. Unno, S. Toyama, K. Motomura et al., Il-25 and il-33 contribute to development of eosinophilic airway inflammation in epicutaneously antigen-sensitized mice, PLoS ONE, vol.10, 2015.

Y. H. Wang, P. Angkasekwinai, N. Lu, K. S. Voo, K. Arima et al., Il-25 augments type 2 immune responses by enhancing the expansion and functions of tslp-dc-activated th2 memory cells, J. Exp. Med, vol.204, pp.1837-1847, 2007.

M. L. Manni, K. M. Robinson, and J. F. Alcorn, A tale of two cytokines: Il-17 and il-22 in asthma and infection, Expert Rev. Respir. Med, vol.8, pp.25-42, 2014.

C. J. Corrigan, W. Wang, Q. Meng, C. Fang, G. Eid et al., Allergen-induced expression of il-25 and il-25 receptor in atopic asthmatic airways and late-phase cutaneous responses, J. Allergy Clin. Immunol, vol.128, pp.116-124, 2011.

M. A. Kleinschek, A. M. Owyang, B. Joyce-shaikh, C. L. Langrish, Y. Chen et al., Il-25 regulates th17 function in autoimmune inflammation, J. Exp. Med, vol.204, pp.161-170, 2007.

V. Ramirez-carrozzi, A. Sambandam, E. Luis, Z. Lin, S. Jeet et al., Il-17c regulates the innate immune function of epithelial cells in an autocrine manner, Nat. Immunol, vol.12, pp.1159-1166, 2011.

S. Y. Hwang and H. Y. Kim, Expression of il-17 homologs and their receptors in the synovial cells of rheumatoid arthritis patients, Mol. Cells, vol.19, pp.180-184, 2005.

H. Li, J. Chen, A. Huang, J. Stinson, S. Heldens et al., Cloning and characterization of il-17b and il-17c, two new members of the il-17 cytokine family, Proc. Natl. Acad. Sci, vol.97, pp.773-778, 2000.

T. Starnes, H. E. Broxmeyer, M. J. Robertson, and R. Hromas, Cutting edge: Il-17d, a novel member of the il-17 family, stimulates cytokine production and inhibits hemopoiesis, J. Immunol, vol.169, pp.642-646, 2002.

J. A. Salvator-fabre, The multifaceted immune system, MOJ Immunol, vol.3, 2016.

M. C. Bonnet, M. Bagot, and A. Bensussan, Monoclonal antibodies targeting il-17a or its receptor in psoriasis: A new therapeutic approach?, Med. Sci, vol.28, pp.1035-1037, 2012.

D. J. Van-uden, H. W. Van-laarhoven, A. H. Westenberg, J. H. De-wilt, and C. F. Blanken-peeters, Inflammatory breast cancer: An overview, Crit. Rev. Oncol./Hematol, vol.93, pp.116-126, 2015.

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 Res, vol.10, 2008.

S. Cochaud, J. Giustiniani, C. Thomas, E. Laprevotte, C. Garbar et al., Il-17a is produced by breast cancer tils and promotes chemoresistance and proliferation through erk1/2. Sci

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, Mol. Med. Rep, vol.6, pp.1099-1102, 2012.

N. Eiro, B. Fernandez-garcia, L. O. Gonzalez, and F. J. Vizoso, Cytokines related to mmp-11 expression by inflammatory cells and breast cancer metastasis

Y. Merrouche, J. Fabre, H. Cure, C. Garbar, C. Fuselier et al., Il-17e synergizes with egf and confers in vitro resistance to egfr-targeted therapies in tnbc cells, Oncotarget, vol.7, pp.53350-53361, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01841201

J. S. Nam, M. Terabe, M. J. Kang, H. Chae, N. Voong et al., Transforming growth factor beta subverts the immune system into directly promoting tumor growth through interleukin-17, Cancer Res, vol.68, pp.3915-3923, 2008.

L. D. Roy, S. Ghosh, L. B. Pathangey, T. L. Tinder, H. E. Gruber et al., Collagen induced arthritis increases secondary metastasis in mmtv-pyv mt mouse model of mammary cancer, BMC Cancer, vol.11, 2011.

L. D. Roy, M. Sahraei, J. L. Schettini, H. E. Gruber, D. M. Besmer et al., Systemic neutralization of il-17a significantly reduces breast cancer associated metastasis in arthritic mice by reducing cxcl12/sdf-1 expression in the metastatic niches, BMC Cancer, vol.14, 2014.

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

E. Laprevotte, S. Cochaud, S. Du-manoir, M. Lapierre, C. Dejou et al., The il-17b-il-17 receptor b pathway promotes resistance to paclitaxel in breast tumors through activation of the erk1/2 pathway, Oncotarget, vol.8, pp.113360-113372, 2017.

K. Keyomarsi, S. L. Tucker, T. A. Buchholz, M. Callister, Y. Ding et al., Cyclin e and survival in patients with breast cancer, The N. Engl. J. Med, vol.347, pp.1566-1575, 2002.

J. A. Bonner, N. J. Maihle, B. R. Folven, T. J. Christianson, and K. Spain, The interaction of epidermal growth factor and radiation in human head and neck squamous cell carcinoma cell lines with vastly different radiosensitivities, Int. J. Radiat. Oncol. Biol. Phys, vol.29, pp.243-247, 1994.

C. K. Huang, C. Y. Yang, Y. M. Jeng, C. L. Chen, H. H. Wu et al., Autocrine/paracrine mechanism of interleukin-17b receptor promotes breast tumorigenesis through nf-kappab-mediated antiapoptotic pathway, Oncogene, vol.33, pp.2968-2977, 2014.

S. B. Coffelt, K. Kersten, C. W. Doornebal, J. Weiden, K. Vrijland et al., Il-17-producing gammadelta t cells and neutrophils conspire to promote breast cancer metastasis, Nature, vol.522, pp.345-348, 2015.

Z. Jiang, J. Chen, X. Du, H. Cheng, X. Wang et al., Il-25 blockade inhibits metastasis in breast cancer, Protein Cell, vol.8, pp.191-201, 2017.

N. Eiro, L. Gonzalez, L. O. Gonzalez, B. Fernandez-garcia, M. L. Lamelas et al., Relationship between the inflammatory molecular profile of breast carcinomas and distant metastasis development, PLoS ONE, vol.7, 2012.

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 Immunol. Immunother, vol.59, pp.805-817, 2010.

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, Sci. Transl. Med, 2011.

M. Ma, W. Huang, and D. Kong, Il-17 inhibits the accumulation of myeloid-derived suppressor cells in breast cancer via activating stat3, Int. Immunopharmacol, vol.59, pp.148-156, 2018.

J. S. Nam, M. Terabe, M. Mamura, M. J. Kang, H. Chae et al., An anti-transforming growth factor beta antibody suppresses metastasis via cooperative effects on multiple cell compartments, Cancer Res, vol.68, pp.3835-3843, 2008.

B. Yang, H. Kang, A. Fung, H. Zhao, T. Wang et al., The role of interleukin 17 in tumour proliferation, angiogenesis, and metastasis, Mediat. Inflamm, 2014.

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, pp.34-44, 2015.

M. C. Honorati, S. Neri, L. Cattini, and A. Facchini, Il-17 enhances the susceptibility of u-2 os osteosarcoma cells to nk cell lysis, Clin. Exp. Immunol, vol.133, pp.344-349, 2003.

T. O'sullivan, R. Saddawi-konefka, E. Gross, M. Tran, S. P. Mayfield et al., Interleukin-17d mediates tumor rejection through recruitment of natural killer cells, Cell Rep, vol.7, pp.989-998, 2014.

P. Parajuli and S. Mittal, Role of il-17 in glioma progression, J. Spine Neurosurg, pp.1-4, 2013.

F. Wu, J. Xu, Q. Huang, J. Han, L. Duan et al., The role of interleukin-17 in lung cancer, Mediat. Inflamm, vol.8494079, 2016.

K. Gu, M. M. Li, J. Shen, F. Liu, J. Y. Cao et al., Interleukin-17-induced emt promotes lung cancer cell migration and invasion via nf-kappab/zeb1 signal pathway, Am. J. Cancer Res, vol.5, pp.1169-1179, 2015.

M. Feng, Y. Wang, K. Chen, Z. Bian, W. Jinfang et al., Il-17a promotes the migration and invasiveness of cervical cancer cells by coordinately activating mmps expression via the p38/nf-kappab signal pathway, PLoS ONE, vol.9, 2014.

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.

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

T. Proverbs-singh, J. L. Feldman, M. J. Morris, K. A. Autio, and T. A. Traina, Targeting the androgen receptor in prostate and breast cancer: Several new agents in development, Endocr. Relat. Cancer, vol.22, pp.87-106, 2015.

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 Res, vol.72, pp.2589-2599, 2012.

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.36, pp.687-699, 2016.

C. Chen, Q. Zhang, S. Liu, K. R. Parajuli, Y. Qu et al., You, Z. Il-17 and insulin/igf1 enhance adhesion of prostate cancer cells to vascular endothelial cells through cd44-vcam-1 interaction, Prostate, vol.75, pp.883-895, 2015.

T. Welte and X. H. Zhang, Interleukin-17 could promote breast cancer progression at several stages of the disease, Mediat. Inflamm, vol.804347, 2015.

J. Loget, J. Plee, F. Antonicelli, and P. Bernard, A successful treatment with ustekinumab in a case of relapsing bullous pemphigoid associated with psoriasis, J. Eur. Acad. Dermatol. Venereol, vol.31, pp.228-230, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01825897