H. El-gabalawy, L. C. Guenther, and C. N. Bernstein, Epidemiology of immune-mediated inflammatory diseases: incidence, prevalence, natural history, and comorbidities, J Rheumatol Suppl, vol.85, pp.2-10, 2010.

R. Banchereau, A. M. Cepika, J. Banchereau, and V. Pascual, Understanding human autoimmunity and autoinflammation through transcriptomics, Annu Rev Immunol, vol.35, pp.337-70, 2017.

A. N. Ananthakrishnan, Environmental risk factors for inflammatory bowel diseases: a review, Digest Dis Sci, vol.60, pp.290-298, 2015.

L. Belbasis, V. Bellou, E. Evangelou, J. P. Ioannidis, and I. Tzoulaki, Environmental risk factors and multiple sclerosis: an umbrella review of systematic reviews and meta-analyses, Lancet Neurol, vol.14, pp.263-73, 2015.

M. Parkes, A. Cortes, D. A. Van-heel, and M. A. Brown, Genetic insights into common pathways and complex relationships among immune-mediated diseases, Nat Rev Genet, vol.14, pp.661-73, 2013.

C. Cotsapas, B. F. Voight, E. Rossin, K. Lage, B. M. Neale et al., Pervasive sharing of genetic effects in autoimmune disease, PLoS Genet, vol.7, 2011.

D. Langlais, N. Fodil, and P. Gros, Genetics of infectious and inflammatory diseases: overlapping discoveries from association and exome-sequencing studies, Annu Rev Immunol, vol.35, pp.1-30, 2017.

X. Wu, H. Chen, and H. Xu, The genomic landscape of human immune-mediated diseases, J Hum Genet, vol.60, pp.675-81, 2015.

A. H. Beecham, N. A. Patsopoulos, D. K. Xifara, M. F. Davis, and A. Kemppinen, Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis, International Multiple Sclerosis Genetics Consortium, vol.45, pp.1353-60, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02565210

S. Sawcer, G. Hellenthal, M. Pirinen, and C. C. Spencer, Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis, Multiple Sclerosis Genetics Consortium, vol.476, pp.214-223, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00996686

L. Jostins, S. Ripke, R. K. Weersma, R. H. Duerr, D. P. Mcgovern et al., Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease, Nature, vol.491, pp.119-143, 2012.

S. Brophy, S. Pavy, P. Lewis, G. Taylor, L. Bradbury et al., Inflammatory eye, skin, and bowel disease in spondyloarthritis: genetic, phenotypic, and environmental factors, J Rheumatol, vol.28, pp.2667-73, 2001.

E. C. Somers, S. L. Thomas, L. Smeeth, and A. J. Hall, Autoimmune diseases cooccurring within individuals and within families: a systematic review, Epidemiology, vol.17, pp.202-219, 2006.

G. Schett, D. Elewaut, I. B. Mcinnes, J. M. Dayer, and M. F. Neurath, How cytokine networks fuel inflammation: toward a cytokine-based disease taxonomy, Nat Med, vol.19, pp.822-826, 2013.

D. Pennica, G. E. Nedwin, J. S. Hayflick, P. H. Seeburg, R. Derynck et al., Human tumour necrosis factor: precursor structure, expression and homology to lymphotoxin, Nature, vol.312, pp.724-733, 1984.

R. M. Locksley, N. Killeen, and M. J. Lenardo, The TNF and TNF receptor superfamilies: integrating mammalian biology, Cell, vol.104, pp.487-501, 2001.

B. B. Aggarwal, Signalling pathways of the TNF superfamily: a double-edged sword, Nat Rev Immunol, vol.3, pp.745-56, 2003.

M. A. Collart, P. Baeuerle, and P. Vassalli, Regulation of tumor necrosis factor alpha transcription in macrophages: involvement of four kappa B-like motifs and of constitutive and inducible forms of NF-kappa B, Mol Cell Biol, vol.10, pp.1498-506, 1990.

K. Haranaka, E. A. Carswell, B. D. Williamson, J. S. Prendergast, N. Satomi et al., Purification, characterization, and antitumor activity of nonrecombinant mouse tumor necrosis factor, Proc Natl Acad Sci, vol.83, pp.3949-53, 1986.

K. Takeda, S. Iwamoto, H. Sugimoto, T. Takuma, N. Kawatani et al., Identity of differentiation inducing factor and tumour necrosis factor, Nature, vol.323, pp.338-378, 1986.

G. Trinchieri, M. Kobayashi, M. Rosen, R. Loudon, M. Murphy et al., Tumor necrosis factor and lymphotoxin induce differentiation of human myeloid cell lines in synergy with immune interferon, J Exp Med, vol.164, pp.1206-1231, 1986.

D. F. Jelinek and P. E. Lipsky, Enhancement of human B cell proliferation and differentiation by tumor necrosis factor-alpha and interleukin 1, J Immunol, vol.139, pp.2970-2976, 1987.

J. H. Kehrl, A. Miller, and A. S. Fauci, Effect of tumor necrosis factor alpha on mitogen-activated human B cells, J Exp Med, vol.166, pp.786-91, 1987.

C. A. Dinarello, J. G. Cannon, S. M. Wolff, H. A. Bernheim, B. Beutler et al., Tumor necrosis factor (cachectin) is an endogenous pyrogen and induces production of interleukin 1, J Exp Med, vol.163, pp.1433-50, 1986.

J. L. Flynn, M. M. Goldstein, J. Chan, K. J. Triebold, K. Pfeffer et al., Tumor necrosis factor-? is required in the protective immune response against mycobacterium tuberculosis in mice, Immunity, vol.2, pp.561-72, 1995.

L. M. Sedger, M. F. Mcdermott, and . Tnf, TNF-receptors: from mediators of cell death and inflammation to therapeutic giants -past, present and future, Cytokine Growth Factor Rev, vol.25, pp.453-72, 2014.

M. Kriegler, C. Perez, K. Defay, A. I. Lu, and S. D. , A novel form of TNF/cachectin is a cell surface cytotoxic transmembrane protein: ramifications for the complex physiology of TNF, Cell, vol.53, pp.45-53, 1988.

M. L. Moss, S. L. Jin, M. E. Milla, D. M. Bickett, W. Burkhart et al., Cloning of a disintegrin metalloproteinase that processes precursor tumour-necrosis factor-alpha, Nature, vol.385, pp.733-739, 1997.

M. Grell, E. Douni, H. Wajant, M. Lohden, M. Clauss et al., The transmembrane form of tumor-necrosis-factor is the prime activating ligand of the 80 Kda tumor-necrosis-factor receptor, Cell, vol.83, pp.793-802, 1995.

A. Krippner-heidenreich, F. Tubing, S. Bryde, S. Willi, G. Zimmermann et al., Control of receptor-induced signaling complex formation by the kinetics of ligand/receptor interaction, J Biol Chem, vol.277, pp.44155-63, 2002.

A. Mukhopadhyay, J. Suttles, R. D. Stout, and B. B. Aggarwal, Genetic deletion of the tumor necrosis factor receptor p60 or p80 abrogates ligandmediated activation of nuclear factor-kappa B and of mitogen-activated protein kinases in macrophages, J Biol Chem, vol.276, pp.31906-31918, 2001.

T. Weiss, M. Grell, K. Siemienski, F. Muhlenbeck, H. Durkop et al., TNFR80-dependent enhancement of TNFR60-induced cell death is mediated by TNFR-associated factor 2 and is specific for TNFR60, J Immunol, vol.161, pp.3136-3178, 1998.

L. Cabal-hierro and P. S. Lazo, Signal transduction by tumor necrosis factor receptors, Cell Signal, vol.24, pp.1297-305, 2012.

G. D. Kalliolias and L. B. Ivashkiv, TNF biology, pathogenic mechanisms and emerging therapeutic strategies, Nat Rev Rheumatol, vol.12, pp.49-62, 2016.

D. Brenner, H. Blaser, and T. W. Mak, Regulation of tumour necrosis factor signalling: live or let die, Nat Rev Immunol, vol.15, pp.362-74, 2015.

C. Y. Wang, M. W. Mayo, R. G. Korneluk, D. V. Goeddel, and A. S. Baldwin, NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation, Science, vol.281, pp.1680-1683, 1998.

W. Schneider-brachert, V. Tchikov, J. Neumeyer, J. M. Winoto-morbach, S. Held-feindt et al., Compartmentalization of TNF receptor 1 signaling: internalized TNF receptosomes as death signaling vesicles, Immunity, vol.21, pp.415-443, 2004.

F. K. Chan, N. F. Luz, and K. Moriwaki, Programmed necrosis in the cross talk of cell death and inflammation, Annu Rev Immunol, vol.33, pp.79-106, 2015.

M. Pasparakis and P. Vandenabeele, Necroptosis and its role in inflammation, Nature, vol.517, pp.311-331, 2015.

A. P. Gregory, C. A. Dendrou, K. E. Attfield, A. Haghikia, D. K. Xifara et al., TNF receptor 1 genetic risk mirrors outcome of anti-TNF therapy in multiple sclerosis, Nature, vol.488, pp.508-519, 2012.

B. Thoma, M. Grell, K. Pfizenmaier, and P. Scheurich, Identification of a 60-kD tumor necrosis factor (TNF) receptor as the major signal transducing component in TNF responses, J Exp Med, vol.172, pp.1019-1042, 1990.

V. Haridas, B. G. Darnay, K. Natarajan, R. Heller, and B. B. Aggarwal, Overexpression of the p80 TNF receptor leads to TNF-dependent apoptosis, nuclear factor-kappa B activation, and c-Jun kinase activation, J Immunol, vol.160, pp.3152-62, 1998.

C. Reinhard, B. Shamoon, V. Shyamala, and L. T. Williams, Tumor necrosis factor alpha-induced activation of c-jun N-terminal kinase is mediated by TRAF2, EMBO J, vol.16, pp.1080-92, 1997.

C. J. Wu, D. B. Conze, X. Li, S. X. Ying, J. A. Hanover et al., TNFalpha induced c-IAP1/TRAF2 complex translocation to a Ubc6-containing compartment and TRAF2 ubiquitination, EMBO J, vol.24, pp.1886-98, 2005.

P. Pantelidis, P. A. Lympany, P. J. Foley, G. C. Fanning, K. I. Welsh et al., Polymorphic analysis of the high-affinity tumor necrosis factor receptor 2, Tissue Antigens, vol.54, pp.585-91, 1999.

A. Barton, S. John, W. E. Ollier, A. Silman, and J. Worthington, Association between rheumatoid arthritis and polymorphism of tumor necrosis factor receptor II, but not tumor necrosis factor receptor I, in Caucasians, Arthritis Rheumat, vol.44, pp.61-66, 2001.

P. Dieude, E. Petit, S. Cailleau-moindrault, J. Osorio, C. Pierlot et al., Association between tumor necrosis factor receptor II and familial, but not sporadic, rheumatoid arthritis: evidence for genetic heterogeneity, Arthritis Rheumat, vol.46, pp.2039-2083, 2002.
URL : https://hal.archives-ouvertes.fr/hal-02084357

A. Till, P. Rosenstiel, A. Krippner-heidenreich, S. Mascheretti-croucher, P. J. Croucher et al., The Met-196 -> Arg variation of human tumor necrosis factor receptor 2 (TNFR2) affects TNF-alpha-induced apoptosis by impaired NF-kappaB signaling and target gene expression, J Biol Chem, vol.280, pp.5994-6004, 2005.

H. Lenk, S. Tanneberger, U. Müller, J. Ebert, and T. Shiga, Phase II clinical trial of high-dose recombinant human tumor necrosis factor, Cancer Chemother Pharmacol, vol.24, pp.391-393, 1989.

B. Beutler, I. W. Milsark, and A. C. Cerami, Passive immunization against cachectin/tumor necrosis factor protects mice from lethal effect of endotoxin, Science, vol.229, pp.869-71, 1985.

K. J. Tracey, Y. Fong, D. G. Hesse, K. R. Manogue, A. T. Lee et al., Anticachectin/TNF monoclonal antibodies prevent septic shock during lethal bacteraemia, Nature, vol.330, pp.662-666, 1987.

M. Feldmann, Development of anti-TNF therapy for rheumatoid arthritis, Nat Rev Immunol, vol.2, pp.364-71, 2002.

J. Brandt, H. Haibel, D. Cornely, W. Golder, J. Gonzalez et al., Successful treatment of active ankylosing spondylitis with the anti-tumor necrosis factor alpha monoclonal antibody infliximab, Arthritis Rheum, vol.43, pp.1346-52, 2000.

J. Braun, J. Sieper, M. Breban, E. Collantes-estevez, R. I. Davis et al., Anti-tumour necrosis factor ? therapy for ankylosing spondylitis: international experience, Ann Rheum Dis, vol.61, pp.51-60, 2002.

J. D. Gorman, K. E. Sack, and J. Davis, Treatment of ankylosing spondylitis by inhibition of tumor necrosis factor alpha, N Engl J Med, vol.346, pp.1349-56, 2002.

M. M. Ward, A. Deodhar, E. A. Akl, A. Lui, J. Ermann et al., American College of Rheumatology/Spondylitis Association of America/spondyloarthritis research and treatment network 2015 recommendations for the treatment of ankylosing spondylitis and nonradiographic axial spondyloarthritis, Arthritis Rheumatol, vol.68, pp.282-98, 2016.

J. Sieper and D. Poddubnyy, New evidence on the management of spondyloarthritis, Nat Rev Rheumatol, vol.12, pp.282-95, 2016.

D. Baeten, E. Kruithof, F. Van-den-bosch, N. Van-den-bossche, A. Herssens et al., Systematic safety follow up in a cohort of 107 patients with spondyloarthropathy treated with infliximab: a new perspective on the role of host defence in the pathogenesis of the disease?, Ann Rheum Dis, vol.62, pp.829-863, 2003.

S. Assassi, Rheumatoid arthritis, TNF inhibitors, and non-melanoma skin cancer, Bmj, vol.352, p.472, 2016.

P. P. Van-lumig, S. P. Menting, J. M. Van-den-reek, P. I. Spuls, P. L. Van-riel et al., An increased risk of non-melanoma skin cancer during TNF-inhibitor treatment in psoriasis patients compared to rheumatoid arthritis patients probably relates to disease-related factors, J Eur Acad Dermatol Venereol, vol.29, pp.752-60, 2015.

L. De-rycke, D. Baeten, D. Foell, E. Kruithof, E. M. Veys et al., Differential expression and response to anti-TNF? treatment of infiltrating versus resident tissue macrophage subsets in autoimmune arthritis, J Pathol, vol.206, pp.17-27, 2005.

M. J. Elliott, R. N. Maini, M. Feldmann, J. R. Kalden, A. C. Smolen et al., Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis, Lancet, vol.344, pp.1105-1115, 1994.

D. M. Knight, H. Trinh, J. Le, S. Siegel, D. Shealy et al., Construction and initial characterization of a mouse-human chimeric anti-TNF antibody, Mol Immunol, vol.30, issue.93, p.90106, 1993.

S. Siegel, D. J. Shealy, M. T. Nakada, J. Le, D. S. Woulfe et al., The mouse/human chimeric monoclonal antibody cA2 neutralizes TNF in vitro and protects transgenic mice from cachexia and TNF lethality in vivo, Cytokine, vol.7, pp.15-25, 1995.

G. Hutas, Golimumab as the first monthly subcutaneous fully human anti-TNF-alpha antibody in the treatment of inflammatory arthropathies, Immunotherapy, vol.2, pp.453-60, 2010.

D. J. Shealy, A. Cai, K. Staquet, A. Baker, E. R. Lacy et al., A trial of etanercept, a recombinant tumor necrosis factor receptor:Fc fusion protein, in patients with rheumatoid arthritis receiving methotrexate, N Engl J Med, vol.2, pp.253-262, 1999.

A. Nesbitt, G. Fossati, M. Bergin, P. Stephens, S. Stephens et al., Mechanism of action of certolizumab pegol (CDP870): in vitro comparison with other anti-tumor necrosis factor ? agents, Inflamm Bowel Dis, vol.13, pp.1323-1355, 2007.

Z. Kaymakcalan, P. Sakorafas, S. Bose, S. Scesney, L. Xiong et al., Comparisons of affinities, avidities, and complement activation of adalimumab, infliximab, and etanercept in binding to soluble and membrane tumor necrosis factor, Clin Immunol, vol.131, pp.308-324, 2009.

H. Mitoma, T. Horiuchi, and H. Tsukamoto, Binding activities of infliximab and etanercept to transmembrane tumor necrosis factor-alpha, Gastroenterology, vol.126, pp.934-939, 2004.

J. M. Van-den-brande, H. Braat, G. R. Van-den-brink, H. H. Versteeg, C. A. Bauer et al., Infliximab but not etanercept induces apoptosis in lamina propria T-lymphocytes from patients with Crohn's disease, Gastroenterology, vol.124, pp.1774-85, 2003.

H. Mitoma, T. Horiuchi, H. Tsukamoto, Y. Tamimoto, Y. Kimoto et al., Mechanisms for cytotoxic effects of anti-tumor necrosis factor agents on transmembrane tumor necrosis factor ?-expressing cells: comparison among infliximab, etanercept, and adalimumab, Arthritis Rheum, vol.58, pp.1248-57, 2008.

J. D. Greenberg, G. Reed, D. Decktor, L. Harrold, D. Furst et al., A comparative effectiveness study of adalimumab, etanercept and infliximab in biologically naive and switched rheumatoid arthritis patients: results from the US CORRONA registry, Ann Rheum Dis, vol.71, pp.1134-1176, 2012.

W. P. Kennedy, J. A. Simon, C. Offutt, P. Horn, A. Herman et al., Efficacy and safety of pateclizumab (anti-lymphotoxin-alpha) compared to adalimumab in rheumatoid arthritis: a head-to-head phase 2 randomized controlled study (The ALTARA Study), Arthritis Res Ther, vol.16, p.467, 2014.

M. H. Buch, P. G. Conaghan, M. A. Quinn, S. J. Bingham, D. Veale et al., True infliximab resistance in rheumatoid arthritis: a role for lymphotoxin alpha?, Ann Rheum Dis, vol.63, pp.1344-1350, 2004.

S. R. Targan, S. B. Hanauer, S. J. Van-deventer, L. Mayer, D. H. Present et al., A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn's disease. Crohn's Disease cA2 Study Group, N Engl J Med, vol.337, pp.1029-1064, 1997.

W. J. Sandborn, S. B. Hanauer, S. Katz, M. Safdi, D. G. Wolf et al., Etanercept for active Crohn's disease: a randomized, doubleblind, placebo-controlled trial, Gastroenterology, vol.121, pp.1088-94, 2001.

D. Costanzo, L. Ayala, F. Megna, M. Gaudiello, F. Patri et al., The risk of herpes zoster in the anti-TNF-alpha era: a case report and review of the literature, J Dermatol Case Rep, vol.7, pp.1-4, 2013.

W. G. Dixon, K. L. Hyrich, K. D. Watson, M. Lunt, J. Galloway et al., Drug-specific risk of tuberculosis in patients with rheumatoid arthritis treated with anti-TNF therapy: results from the British Society for Rheumatology Biologics Register (BSRBR), Ann Rheum Dis, vol.69, pp.522-530, 2010.

F. Tubach, D. Salmon, P. Ravaud, Y. Allanore, P. Goupille et al., Risk of tuberculosis is higher with anti-tumor necrosis factor monoclonal antibody therapy than with soluble tumor necrosis factor receptor therapy: the three-year prospective French Research Axed on Tolerance of Biotherapies registry, Arthritis Rheum, vol.60, pp.1884-94, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00618680

L. C. Coates, L. S. Cawkwell, N. Ng, A. N. Bennett, D. J. Bryer et al., Real life experience confirms sustained response to long-term biologics and switching in ankylosing spondylitis, Rheumatology, vol.47, pp.897-900, 2008.

B. J. Scallon, M. A. Moore, H. Trinh, D. M. Knight, and J. Ghrayeb, Chimeric anti-TNF-alpha monoclonal antibody cA2 binds recombinant transmembrane TNF-alpha and activates immune effector functions, Cytokine, vol.7, pp.251-260, 1995.

D. Tracey, L. Klareskog, E. H. Sasso, J. G. Salfeld, and P. P. Tak, Tumor necrosis factor antagonist mechanisms of action: a comprehensive review, Pharmacol Ther, vol.117, pp.244-79, 2008.

N. J. Gullick, H. G. Evans, L. D. Church, D. M. Jayaraj, A. Filer et al., Linking power Doppler ultrasound to the presence of th17 cells in the rheumatoid arthritis joint, PLoS ONE, vol.5, p.12516, 2010.

N. E. Aerts, K. J. De-knop, J. Leysen, D. G. Ebo, C. H. Bridts et al., Increased IL-17 production by peripheral T helper cells after tumour necrosis factor blockade in rheumatoid arthritis is accompanied by inhibition of migration-associated chemokine receptor expression, Rheumatology, vol.49, pp.2264-72, 2010.

D. Chen, Y. Chen, H. Chen, C. Hsieh, C. Lin et al., Increasing levels of circulating Th17 cells and interleukin-17 in rheumatoid arthritis patients with an inadequate response to anti-TNF-? therapy, Arthritis Res Ther, vol.13, 2011.

S. Herman, N. Zurgil, S. Machlav, A. Shinberg, P. Langevitz et al., Distinct effects of anti-tumor necrosis factor combined therapy on TH1/TH2 balance in rheumatoid arthritis patients, Clin Vacc Immunol, vol.18, pp.1077-82, 2011.

C. Lina, W. Conghua, N. L. Ping, and Z. , Combined treatment of etanercept and MTX reverses Th1/Th2, Th17/Treg imbalance in patients with rheumatoid arthritis, J Clin Immunol, vol.31, pp.596-605, 2011.

L. Limón-camacho, M. I. Vargas-rojas, J. Vázquez-mellado, J. Casasola-vargas, J. F. Moctezuma et al., In vivo peripheral blood proinflammatory T cells in patients with ankylosing spondylitis, J Rheumatol, vol.39, pp.830-835, 2012.

S. Alzabin, S. M. Abraham, T. E. Taher, A. Palfreeman, D. Hull et al., Incomplete response of inflammatory arthritis to TNF? blockade is associated with the Th17 pathway, Ann Rheum Dis, vol.71, pp.1741-1749, 2012.

B. Szalay, G. Mészáros, Á. Cseh, L. Ács, M. Deák et al., Adaptive immunity in ankylosing spondylitis: phenotype and functional alterations of T-cells before and during infliximab therapy, Clin Dev Immunol, p.808724, 2012.

L. Xueyi, C. Lina, W. Zhenbiao, H. Qing, L. Qiang et al., Levels of circulating Th17 cells and regulatory T cells in ankylosing spondylitis patients with an inadequate response to anti-TNF-? therapy, J Clin Immunol, vol.33, pp.151-61, 2013.

B. Szalay, B. Vásárhelyi, A. Cseh, T. Tulassay, M. Deák et al., The impact of conventional DMARD and biological therapies on CD4+ cell subsets in rheumatoid arthritis: a follow-up study, Clin. Rheumatol, vol.33, pp.175-85, 2014.

H. G. Evans, U. Roostalu, G. J. Walter, N. J. Gullick, K. S. Frederiksen et al., TNF-alpha blockade induces IL-10 expression in human CD4+ T cells, Nat Commun, vol.5, p.3199, 2014.

D. N. Hull, R. O. Williams, E. Pathan, S. Alzabin, S. Abraham et al., Antitumour necrosis factor treatment increases circulating T helper type 17 cells similarly in different types of inflammatory arthritis, Clin Exp Immunol, vol.181, pp.401-407, 2015.

R. Talotta, A. Berzi, F. Atzeni, A. Batticciotto, M. Clerici et al., Paradoxical expansion of Th1 and Th17 lymphocytes in rheumatoid arthritis following infliximab treatment: a possible explanation for a lack of clinical response, J Clin Immunol, vol.35, pp.550-557, 2015.

D. N. Hull, H. Cooksley, S. Chokshi, R. O. Williams, S. Abraham et al., Increase in circulating Th17 cells during anti-TNF therapy is associated with ultrasonographic improvement of synovitis in rheumatoid arthritis, Arthritis Res Ther, vol.18, p.303, 2016.

M. R. Ehrenstein, J. G. Evans, A. Singh, S. Moore, G. Warnes et al., Compromised function of regulatory T cells in rheumatoid arthritis and reversal by anti-TNFalpha therapy, J Exp Med, vol.200, pp.277-85, 2004.

S. Nadkarni, C. Mauri, and M. R. Ehrenstein, Anti-TNF-alpha therapy induces a distinct regulatory T cell population in patients with rheumatoid arthritis via TGF-beta, J Exp Med, vol.204, pp.33-42, 2007.

Z. Huang, B. Yang, Y. Shi, B. Cai, Y. Li et al., Anti-TNF-alpha therapy improves Treg and suppresses Teff in patients with rheumatoid arthritis, Cell Immunol, vol.279, pp.25-34, 2012.

J. L. Mcgovern, D. X. Nguyen, C. A. Notley, C. Mauri, D. A. Isenberg et al., Th17 cells are restrained by Treg cells via the inhibition of interleukin-6 in patients with rheumatoid arthritis responding to anti-tumor necrosis factor antibody therapy, Arthritis Rheumat, vol.64, pp.3129-3167, 2012.

D. X. Nguyen and M. R. Ehrenstein, Anti-TNF drives regulatory T cell expansion by paradoxically promoting membrane TNF-TNF-RII binding in rheumatoid arthritis, J Exp Med, vol.213, pp.1241-53, 2016.

J. H. Anolik, R. Ravikumar, J. Barnard, T. Owen, A. Almudevar et al., Cutting edge: anti-tumor necrosis factor therapy in rheumatoid arthritis inhibits memory B lymphocytes via effects on lymphoid germinal centers and follicular dendritic cell networks, J Immunol, vol.180, pp.688-92, 2008.

M. M. Souto-carneiro, V. Mahadevan, K. Takada, R. Fritsch-stork, T. Nanki et al., Alterations in peripheral blood memory B cells in patients with active rheumatoid arthritis are dependent on the action of tumour necrosis factor, Arthritis Res Ther, vol.11, 2009.

J. J. Kobie, B. Zheng, P. Bryk, M. Barnes, C. T. Ritchlin et al., Decreased influenza-specific B cell responses in rheumatoid arthritis patients treated with anti-tumor necrosis factor, Arthritis Res Ther, vol.13, 2011.

G. F. Salinas, L. De-rycke, B. Barendregt, J. E. Paramarta, H. Hreggvidstdottir et al., Anti-TNF treatment blocks the induction of T cell-dependent humoral responses, Ann Rheum Dis, vol.72, pp.1037-1080, 2013.

P. Conigliaro, P. Triggianese, C. Perricone, M. S. Chimenti, D. Muzio et al., Restoration of peripheral blood natural killer and B cell levels in patients affected by rheumatoid and psoriatic arthritis during etanercept treatment, Clin Exp Immunol, vol.177, pp.234-277, 2014.

C. I. Daien, S. Gailhac, T. Mura, R. Audo, B. Combe et al., Regulatory B10 cells are decreased in patients with rheumatoid arthritis and are inversely correlated with disease activity, Arthritis Rheumatol, vol.66, pp.2037-2083, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02191567

S. Glaesener, T. D. Quach, N. Onken, F. Weller-heinemann, F. Dressler et al., Distinct effects of methotrexate and etanercept on the B cell compartment in patients with juvenile idiopathic arthritis, Arthritis Rheumatol, vol.66, pp.2590-600, 2014.

M. Bautista-caro, E. De-miguel, D. Peiteado, C. Plasencia-rodríguez, A. Villalba et al., Increased frequency of circulating CD19+CD24hiCD38hi B cells with regulatory capacity in patients with Ankylosing spondylitis (AS) naïve for biological agents, PLoS ONE, vol.12, p.180726, 2017.

H. M. Ingelman-sundberg, Å. Laestadius, C. Chrapkowska, K. Mördrup, B. Magnusson et al., Diverse effects on vaccine-specific serum IgG titres and memory B cells upon methotrexate and anti-TNF-? therapy in children with rheumatic diseases: a cross-sectional study, Vaccine, vol.34, pp.1304-1315, 2016.

M. Chen, L. Zhang, Y. Ren, K. Zhang, Y. Yang et al., Defective function of CD24(+)CD38(+) regulatory B cells in ankylosing spondylitis, DNA Cell Biol, vol.35, pp.88-95, 2016.

S. Salomon, C. Guignant, P. Morel, G. Flahaut, C. Brault et al., Th17 and CD24hiCD27+ regulatory B lymphocytes are biomarkers of response to biologics in rheumatoid arthritis, Arthritis Res Ther, vol.19, p.33, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02544267

Z. Banko, J. Pozsgay, T. Gati, B. Rojkovich, I. Ujfalussy et al., Regulatory B cells in rheumatoid arthritis: alterations in patients receiving anti-TNF therapy, Clin Immunol, vol.184, pp.63-72, 2017.

A. Mavropoulos, A. Varna, E. Zafiriou, C. Liaskos, I. Alexiou et al., IL-10 producing Bregs are impaired in psoriatic arthritis and psoriasis and inversely correlate with IL-17-and IFNgamma-producing T cells, Clin Immunol, vol.184, pp.33-41, 2017.

E. J. Dombrecht, N. E. Aerts, A. J. Schuerwegh, M. M. Hagendorens, D. G. Ebo et al., Influence of anti-tumor necrosis factor therapy (Adalimumab) on regulatory T cells and dendritic cells in rheumatoid arthritis, Clin Exp Rheumatol, vol.24, pp.31-38, 2006.

G. Nocturne, S. Boudaoud, B. Ly, J. Pascaud, A. Paoletti et al., Impact of anti-TNF therapy on NK cells function and on immunosurveillance against B-cell lymphomas, J Autoimmunity, vol.80, pp.56-64, 2017.

W. X. Mo, S. S. Yin, H. Chen, C. Zhou, J. X. Zhou et al., Chemotaxis of Vdelta2 T cells to the joints contributes to the pathogenesis of rheumatoid arthritis, Ann Rheum Dis, vol.76, pp.2075-84, 2017.

I. Blijdorp, S. Menegatti, L. Van-mens, M. Van-de-sand, S. Chen et al., IL-22-and GM-CSF-expressing but not IL-17A-expressing group 3 innate lymphoid cells are expanded in the inflamed spondyloarthritis joint, Arthritis Rheumatol, vol.71, pp.392-402, 2019.

N. Kawanaka, M. Yamamura, T. Aita, Y. Morita, A. Okamoto et al., CD16+ blood monocytes and joint inflammation in rheumatoid arthritis, Arthritis Rheumat, vol.46, pp.2578-86, 2002.

L. R. Coulthard, J. Geiler, R. J. Mathews, L. D. Church, L. J. Dickie et al., Differential effects of infliximab on absolute circulating blood leucocyte counts of innate immune cells in early and late rheumatoid arthritis patients, Clin Exp Immunol, vol.170, pp.36-46, 2012.

M. Krasselt, C. Baerwald, U. Wagner, and M. Rossol, CD56+ monocytes have a dysregulated cytokine response to lipopolysaccharide and accumulate in rheumatoid arthritis and immunosenescence, Arthritis Res Ther, vol.15, p.139, 2013.

N. Muller, F. Doring, M. Klapper, K. Neumann, D. M. Schulte et al., Interleukin-6 and tumour necrosis factor-alpha differentially regulate lincRNA transcripts in cells of the innate immune system in vivo in human subjects with rheumatoid arthritis, Cytokine, vol.68, pp.65-73, 2014.

U. Meusch, M. Krasselt, M. Rossol, C. Baerwald, M. Klingner et al., In vitro response pattern of monocytes after tmTNF reverse signaling predicts response to anti-TNF therapy in rheumatoid arthritis, J Transl Med, vol.13, p.256, 2015.

D. Aeberli, R. Kamgang, D. Balani, W. Hofstetter, P. M. Villiger et al., Regulation of peripheral classical and non-classical monocytes on infliximab treatment in patients with rheumatoid arthritis and ankylosing spondylitis, RMD Open, vol.2, p.79, 2016.

J. Zhao, W. Yuan, C. Tao, P. Sun, Z. Yang et al., M2 polarization of monocytes in ankylosing spondylitis and relationship with inflammation and structural damage, Acta Pathol Microbiol Immunol Scand, vol.125, pp.1070-1075, 2017.

M. M. Maurice, W. L. Van-der-graaff, A. Leow, F. C. Breedveld, R. A. Van-lier et al., Treatment with monoclonal anti-tumor necrosis factor alpha antibody results in an accumulation of Th1 CD4+ T cells in the peripheral blood of patients with rheumatoid arthritis, Arthritis Rheum, vol.42, pp.2166-73, 1999.

J. Zou, M. Rudwaleit, J. Brandt, A. Thiel, J. Braun et al., Down-regulation of the nonspecific and antigen-specific T cell cytokine response in ankylosing spondylitis during treatment with infliximab, Arthritis Rheum, vol.48, pp.780-90, 2003.

J. Zou, M. Rudwaleit, J. Brandt, A. Thiel, J. Braun et al., Up regulation of the production of tumour necrosis factor alpha and interferon gamma by T cells in ankylosing spondylitis during treatment with etanercept, Ann Rheumat Dis, vol.62, pp.561-565, 2003.

O. Aravena, B. Pesce, L. Soto, N. Orrego, F. Sabugo et al., Anti-TNF therapy in patients with rheumatoid arthritis decreases Th1 and Th17 cell populations and expands IFN-gamma-producing NK cell and regulatory T cell subsets, Immunobiology, vol.216, pp.1256-63, 2011.

S. Dulic, Z. Vasarhelyi, F. Sava, L. Berta, B. Szalay et al., Tcell subsets in rheumatoid arthritis patients on long-term anti-TNF or IL-6 receptor blocker therapy, Mediators Inflamm, p.6894374, 2017.

P. Charles, M. J. Elliott, D. Davis, A. Potter, J. R. Kalden et al., Regulation of cytokines, cytokine inhibitors, and acutephase proteins following anti-TNF-alpha therapy in rheumatoid arthritis, J Immunol, vol.163, pp.1521-1529, 1999.

P. C. Taylor, A. M. Peters, E. Paleolog, P. T. Chapman, M. J. Elliott et al., Reduction of chemokine levels and leukocyte traffic to joints by tumor necrosis factor alpha blockade in patients with rheumatoid arthritis, Arthritis Rheumat, vol.43, pp.38-47, 2000.

P. Barrera, L. A. Joosten, A. A. Den-broeder, L. B. Van-de-putte, P. L. Van-riel et al., Effects of treatment with a fully human anti-tumour necrosis factor alpha monoclonal antibody on the local and systemic homeostasis of interleukin 1 and TNFalpha in patients with rheumatoid arthritis, Ann Rheumat Dis, vol.60, pp.660-669, 2001.

P. A. Klimiuk, S. Sierakowski, I. Domyslawska, and J. Chwiecko, Regulation of serum chemokines following infliximab therapy in patients with rheumatoid arthritis, Clin Exp Rheumatol, vol.24, pp.529-562, 2006.

Y. Kageyama, T. Ichikawa, T. Nagafusa, E. Torikai, M. Shimazu et al., Etanercept reduces the serum levels of interleukin-23 and macrophage inflammatory protein-3 alpha in patients with rheumatoid arthritis, Rheumatol Int, vol.28, pp.137-180, 2007.

A. W. Van-lieshout, J. Fransen, M. Flendrie, A. M. Eijsbouts, F. H. Van-den-hoogen et al., Circulating levels of the chemokine CCL18 but not CXCL16 are elevated and correlate with disease activity in rheumatoid arthritis, Ann Rheum Dis, vol.66, pp.1334-1342, 2007.

S. Fabre, A. M. Dupuy, N. Dossat, C. Guisset, J. D. Cohen et al., Protein biochip array technology for cytokine profiling predicts etanercept responsiveness in rheumatoid arthritis, Clin Exp Immunol, vol.153, pp.188-95, 2008.

S. Kawashiri, A. Kawakami, N. Iwamoto, K. Fujikawa, T. Aramaki et al., Proinflammatory cytokines synergistically enhance the production of chemokine ligand 20 (CCL20) from rheumatoid fibroblastlike synovial cells in vitro and serum CCL20 is reduced in vivo by biologic disease-modifying antirheumatic drugs, J Rheumatol, vol.36, p.132, 2009.

T. Odai, M. Matsunawa, R. Takahashi, K. Wakabayashi, T. Isozaki et al., Correlation of CX3CL1 and CX3CR1 levels with response to infliximab therapy in patients with rheumatoid arthritis, J Rheumatol, vol.36, pp.1158-65, 2009.

C. Popa, P. Barrera, L. A. Joosten, P. L. Van-riel, B. J. Kullberg et al., Cytokine production from stimulated whole blood cultures in rheumatoid arthritis patients treated with various TNF blocking agents, Eur Cytokine Netw, vol.20, pp.88-93, 2009.

H. Akbulut, S. S. Koca, M. Ozgen, and A. Isik, Anti-tumor necrosis factor therapies reduce serum macrophage inflammatory protein-1alpha in ankylosing spondylitis, J Rheumatol, vol.37, pp.1073-1077, 2010.

C. Yue, X. You, L. Zhao, H. Wang, F. Tang et al., The effects of adalimumab and methotrexate treatment on peripheral Th17 cells and IL-17/IL-6 secretion in rheumatoid arthritis patients, Rheumatol Int, vol.30, pp.1553-1560, 2010.

F. Bosè, L. Raeli, C. Garutti, E. Frigerio, A. Cozzi et al., Dual role of anti-TNF therapy: enhancement of TCR-mediated T cell activation in peripheral blood and inhibition of inflammation in target tissues, Clin Immunol, vol.139, pp.164-76, 2011.

S. I. Taylan, B. Akinci, S. Bilge, D. Kozaci, and S. Akar, Biomarkers and cytokines of bone turnover: extensive evaluation in a cohort of patients with ankylosing spondylitis, BMC Musculoskelet Disorders, vol.13, p.191, 2012.

K. Kayakabe, T. Kuroiwa, N. Sakurai, H. Ikeuchi, T. Kadiombo et al., Interleukin-1beta measurement in stimulated whole blood cultures is useful to predict response to anti-TNF therapies in rheumatoid arthritis, Rheumatology, vol.51, pp.1639-1682, 2012.

Y. Tian, H. Shen, L. Xia, and J. Lu, Elevated serum and synovial fluid levels of interleukin-34 in rheumatoid arthritis: possible association with disease progression via interleukin-17 production, J Interfer Cytokine Res, vol.33, pp.398-401, 2013.

S. R. Greisen, K. K. Schelde, T. K. Rasmussen, T. W. Kragstrup, K. Stengaard-pedersen et al., CXCL13 predicts disease activity in early rheumatoid arthritis and could be an indicator of the therapeutic 'window of opportunity', Arthritis Res Ther, vol.16, p.434, 2014.

S. H. Chang, B. Y. Choi, J. Choi, J. J. Yoo, Y. Ha et al., Baseline serum interleukin-34 levels independently predict radiographic progression in patients with rheumatoid arthritis, Rheumatol Int, vol.35, pp.71-80, 2015.

F. Genre, R. Lopez-mejias, J. A. Miranda-filloy, B. Ubilla, V. Mijares et al., Anti-TNF-alpha therapy reduces endothelial cell activation in non-diabetic ankylosing spondylitis patients, Rheumatol Int, vol.35, pp.2069-78, 2015.

F. M. Milanez, C. G. Saad, V. T. Viana, J. C. Moraes, G. V. Perico et al., IL-23/Th17 axis is not influenced by TNF-blocking agents in ankylosing spondylitis patients, Arthritis Res Ther, vol.18, p.52, 2016.

B. K. Han, I. Kuzin, J. P. Gaughan, N. J. Olsen, and A. Bottaro, Baseline CXCL10 and CXCL13 levels are predictive biomarkers for tumor necrosis factor inhibitor therapy in patients with moderate to severe rheumatoid arthritis: a pilot, prospective study, Arthritis Res Ther, vol.10, pp.1-7, 2016.

H. M. Walters, N. Pan, T. J. Lehman, A. Adams, G. D. Kalliolias et al., The impact of disease activity and tumour necrosis factor-alpha inhibitor therapy on cytokine levels in juvenile idiopathic arthritis, Clin Exp Immunol, vol.184, pp.308-325, 2016.

T. Wampler-muskardin, P. Vashisht, J. M. Dorschner, M. A. Jensen, B. S. Chrabot et al., Increased pretreatment serum IFN-?/a ratio predicts nonresponse to tumour necrosis factor a inhibition in rheumatoid arthritis, Ann Rheumat Dis, vol.75, pp.1757-62, 2016.

J. Bystrom, F. I. Clanchy, T. E. Taher, M. M. Al-bogami, H. A. Muhammad et al., Response to treatment with TNFa inhibitors in rheumatoid arthritis is associated with high levels of GM-CSF and GM-CSF+ T lymphocytes, Clin Rev Allergy Immunol, vol.53, pp.265-76, 2017.

M. H. Al-mossawi, L. Chen, H. Fang, A. Ridley, D. Wit et al., Unique transcriptome signatures and GM-CSF expression in lymphocytes from patients with spondyloarthritis, Nat Commun, vol.8, pp.1-11, 2017.

A. Makris, S. Adamidi, C. Koutsianas, C. Tsalapaki, E. Hadziyannis et al., Increased frequency of peripheral B and T cells expressing granulocyte monocyte colony-stimulating factor in rheumatoid arthritis patients, Front Immunol, vol.8, 1967.

J. Bystrom, F. I. Clanchy, T. E. Taher, P. Mangat, A. S. Jawad et al., TNF? in the regulation of Treg and Th17 cells in rheumatoid arthritis and other autoimmune inflammatory diseases, Cytokine, vol.101, pp.4-13, 2018.

J. Davignon, B. Rauwel, Y. Degboé, A. Constantin, J. Boyer et al., Modulation of T-cell responses by anti-tumor necrosis factor treatments in rheumatoid arthritis: a review, Arthritis Res Ther, vol.20, p.229, 2018.

A. P. Cope, R. S. Liblau, X. Yang, M. Congia, C. Laudanna et al., Chronic tumor necrosis factor alters T cell responses by attenuating T cell receptor signaling, J Exp Med, vol.185, pp.1573-84, 1997.

Z. Zhang, C. L. Gorman, A. Vermi, C. Monaco, A. Foey et al., TCRzetadim lymphocytes define populations of circulating effector cells that migrate to inflamed tissues, Blood, vol.109, pp.4328-4335, 2007.

L. Steinman, A brief history of TH17, the first major revision in the TH1/TH2 hypothesis of T cell-mediated tissue damage, Nat Med, vol.13, pp.139-184, 2007.

G. Feng, W. Gao, T. B. Strom, M. Oukka, R. S. Francis et al., Exogenous IFN-gamma ex vivo shapes the alloreactive T-cell repertoire by inhibition of Th17 responses and generation of functional Foxp3+ regulatory T cells, Eur J Immunol, vol.38, pp.2512-2539, 2008.

L. E. Harrington, R. D. Hatton, P. R. Mangan, H. Turner, T. L. Murphy et al., Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages, Nat Immunol, vol.6, pp.1123-1155, 2005.

C. Chu, D. Swart, D. Alcorn, J. Tocker, and K. B. Elkon, Interferon-gamma regulates susceptibility to collagen-induced arthritis through suppression of interleukin-17, Arthritis Rheum, vol.56, pp.1145-51, 2007.

H. Kelchtermans, E. Schurgers, L. Geboes, T. Mitera, J. Van-damme et al., Effector mechanisms of interleukin-17 in collagen-induced arthritis in the absence of interferon-gamma and counteraction by interferon-gamma, Arthritis Res Ther, vol.11, 2009.

R. Nissinen, M. Leirisalo-repo, R. Peltomaa, T. Palosuo, and O. Vaarala, Cytokine and chemokine receptor profile of peripheral blood mononuclear cells during treatment with infliximab in patients with active rheumatoid arthritis, Ann Rheum. Dis, vol.63, pp.681-688, 2004.

A. J. Schuerwegh, J. F. Van-offel, W. J. Stevens, C. H. Bridts, D. Clerck et al., Influence of therapy with chimeric monoclonal tumour necrosis factoralpha antibodies on intracellular cytokine profiles of T lymphocytes and monocytes in rheumatoid arthritis patients, Rheumatology, vol.42, pp.541-549, 2003.

C. A. Notley, J. J. Inglis, S. Alzabin, F. E. Mccann, K. E. Mcnamee et al., Blockade of tumor necrosis factor in collagen-induced arthritis reveals a novel immunoregulatory pathway for Th1 and Th17 cells, J Exp Med, vol.205, pp.2491-2498, 2008.

J. A. Hamilton, Plasminogen activator/plasmin system in arthritis and inflammation: friend or foe?, Arthritis Rheum, vol.58, pp.645-653, 2008.

C. Worth, P. Bowness, H. Al-mossawi, and M. , Novel therapeutic targets in axial spondyloarthritis, Curr Treatm Opt Rheumatol, vol.4, pp.174-82, 2018.

J. D. Griffin, S. A. Cannistra, R. Sullivan, G. D. Demetri, T. J. Ernst et al., The biology of GM-CSF: regulation of production and interaction with its receptor, Int J Cell Cloning, vol.8, pp.35-44, 1990.

G. Reynolds, J. R. Gibbon, A. G. Pratt, M. J. Wood, D. Coady et al., Synovial CD4+ T-cell-derived GM-CSF supports the differentiation of an inflammatory dendritic cell population in rheumatoid arthritis, Ann Rheum Dis, vol.75, pp.899-907, 2016.

D. Greven, E. S. Cohen, D. M. Gerlag, J. Campbell, J. Woods et al., Preclinical characterisation of the GM-CSF receptor as a therapeutic target in rheumatoid arthritis, Ann Rheum Dis, vol.74, pp.1924-1954, 2015.

H. Yamada, A. Haraguchi, K. Sakuraba, K. Okazaki, J. Fukushi et al., Th1 is the predominant helper T cell subset that produces GM-CSF in the joint of rheumatoid arthritis. RMD Open, vol.3, p.487, 2017.

G. R. Burmester, I. B. Mcinnes, J. M. Kremer, P. Miranda, J. Vencovský et al., Mavrilimumab, a fully human granulocyte-macrophage colonystimulating factor receptor ? monoclonal antibody: long-term safety and efficacy in patients with rheumatoid arthritis, Arthritis Rheumatol, vol.70, pp.679-89, 2018.

P. P. Tak, P. C. Taylor, F. C. Breedveld, T. J. Smeets, M. R. Daha et al., Decrease in cellularity and expression of adhesion molecules by anti-tumor necrosis factor alpha monoclonal antibody treatment in patients with rheumatoid arthritis, Arthritis Rheum, vol.39, pp.1077-81, 1996.

R. E. Banks, Measurement of cytokines in clinical samples using immunoassays: problems and pitfalls, Crit Rev Clin Lab Sci, vol.37, pp.131-82, 2000.

D. Aeberli, M. Seitz, P. Jüni, and P. M. Villiger, Increase of peripheral CXCR3 positive T lymphocytes upon treatment of RA patients with TNF-alpha inhibitors, Rheumatology, vol.44, pp.172-177, 2005.

K. Hirota, H. Yoshitomi, M. Hashimoto, S. Maeda, S. Teradaira et al., Preferential recruitment of CCR6-expressing Th17 cells to inflamed joints via CCL20 in rheumatoid arthritis and its animal model, J Exp Med, vol.204, pp.2803-2815, 2007.

R. M. Strieter, M. D. Burdick, B. N. Gomperts, J. A. Belperio, and M. P. Keane, CXC chemokines in angiogenesis, Cytokine Growth Factor Rev, vol.16, pp.593-609, 2005.

H. Erdem, S. Pay, U. Musabak, I. Simsek, A. Dinc et al., Synovial angiostatic non-ELR CXC chemokines in inflammatory arthritides: does CXCL4 designate chronicity of synovitis?, Rheumatol Int, vol.27, pp.969-73, 2007.

A. Sucur, Z. Jajic, M. Artukovic, M. I. Matijasevic, B. Anic et al., Chemokine signals are crucial for enhanced homing and differentiation of circulating osteoclast progenitor cells, Arthritis Res Ther, vol.19, p.142, 2017.

A. Aggarwal, A. N. Sarangi, P. Gaur, A. Shukla, and R. Aggarwal, Gut microbiome in children with enthesitis-related arthritis in a developing country and the effect of probiotic administration, Clin Exp Immunol, vol.187, pp.480-489, 2017.

O. Pala, A. Diaz, B. B. Blomberg, and D. Frasca, B Lymphocytes in rheumatoid arthritis and the effects of anti-TNF-? agents on B lymphocytes: a review of the literature, Clin Ther, vol.40, pp.1034-1079, 2018.

D. A. Kaminski, C. Wei, Y. Qian, A. F. Rosenberg, and I. Sanz, Advances in human B cell phenotypic profiling, Front Immunol, vol.3, p.302, 2012.

M. Bautista-caro, I. Arroyo-villa, C. Castillo-gallego, E. De-miguel, D. Peiteado et al., Decreased frequencies of circulating follicular helper T cell counterparts and plasmablasts in ankylosing spondylitis patients Naïve for TNF blockers, PLoS ONE, vol.9, p.107086, 2014.

A. Fekete, L. Soos, Z. Szekanecz, Z. Szabo, P. Szodoray et al., Disturbances in B-and T-cell homeostasis in rheumatoid arthritis: suggested relationships with antigen-driven immune responses, J Autoimmun, vol.29, pp.154-63, 2007.

C. Mauri and M. Menon, The expanding family of regulatory B cells, Int Immunol, vol.27, pp.479-86, 2015.

F. Flores-borja, A. Bosma, D. Ng, V. Reddy, M. R. Ehrenstein et al., CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation, Sci Transl Med, vol.5, pp.173-196, 2013.

T. Arora, R. Padaki, L. Liu, A. E. Hamburger, A. R. Ellison et al., Differences in binding and effector functions between classes of TNF antagonists, Cytokine, vol.45, pp.124-155, 2009.

S. Salemi, A. Picchianti-diamanti, V. Germano, I. Donatelli, D. Martino et al., Influenza vaccine administration in rheumatoid arthritis patients under treatment with TNFalpha blockers: safety and immunogenicity, Clin Immunol, vol.134, pp.113-133, 2010.

M. Bombardieri, M. Lewis, and C. Pitzalis, Ectopic lymphoid neogenesis in rheumatic autoimmune diseases, Nature reviews. Rheumatology, vol.13, pp.141-54, 2017.

E. Corsiero, A. Nerviani, M. Bombardieri, and C. Pitzalis, Ectopic lymphoid structures: powerhouse of autoimmunity, Front Immunol, vol.7, p.430, 2016.

D. L. Drayton, X. Ying, J. Lee, W. Lesslauer, and N. H. Ruddle, Ectopic LT alpha beta directs lymphoid organ neogenesis with concomitant expression of peripheral node addressin and a HEV-restricted sulfotransferase, J Exp Med, vol.197, pp.1153-63, 2003.

G. C. Furtado, M. E. Pacer, G. Bongers, C. Benezech, Z. He et al., TNFalpha-dependent development of lymphoid tissue in the absence of RORgammat(+) lymphoid tissue inducer cells, Mucos Immunol, vol.7, pp.602-616, 2014.

A. Kratz, A. Campos-neto, M. S. Hanson, and N. H. Ruddle, Chronic inflammation caused by lymphotoxin is lymphoid neogenesis, J Exp Med, vol.183, pp.1461-72, 1996.

J. D. Canete, R. Celis, C. Moll, E. Izquierdo, S. Marsal et al., Clinical significance of synovial lymphoid neogenesis and its reversal after antitumour necrosis factor alpha therapy in rheumatoid arthritis, Ann Rheumat Dis, vol.68, pp.751-757, 2009.

T. Cantaert, J. Kolln, T. Timmer, T. C. Van-der-pouw-kraan, B. Vandooren et al., B lymphocyte autoimmunity in rheumatoid synovitis is independent of ectopic lymphoid neogenesis, J Immunol, vol.181, pp.785-94, 2008.

G. W. Jones and S. A. Jones, Ectopic lymphoid follicles: inducible centres for generating antigen-specific immune responses within tissues, Immunology, vol.147, pp.141-51, 2016.

F. Humby, M. Bombardieri, A. Manzo, S. Kelly, M. C. Blades et al., Ectopic lymphoid structures support ongoing production of classswitched autoantibodies in rheumatoid synovium, PLoS Med, vol.6, p.1, 2009.

X. Feng, X. Xu, Y. Wang, Z. Zheng, and G. Lin, Ectopic germinal centers and IgG4-producing plasmacytes observed in synovia of HLA-B27+ ankylosing spondylitis patients with advanced hip involvement, Int J Rheumatol, p.316421, 2015.

M. Rudwaleit, J. Listing, J. Brandt, J. Braun, and J. Sieper, Prediction of a major clinical response (BASDAI 50) to tumour necrosis factor alpha blockers in ankylosing spondylitis, Ann Rheum Dis, vol.63, pp.665-70, 2004.

S. Arends, E. Brouwer, E. Van-der-veer, H. Groen, M. K. Leijsma et al., Baseline predictors of response and discontinuation of tumor necrosis factor-alpha blocking therapy in ankylosing spondylitis: a prospective longitudinal observational cohort study, Arthritis Res Ther, vol.13, 2011.

M. K. De-vries, I. C. Van-eijk, I. E. Van-der-horst-bruinsma, M. J. Peters, M. T. Nurmohamed et al., Erythrocyte sedimentation rate, Creactive protein level, and serum amyloid a protein for patient selection and monitoring of anti-tumor necrosis factor treatment in ankylosing spondylitis, Arthritis Rheumat, vol.61, pp.1484-90, 2009.

N. Vastesaeger, B. V. Cruyssen, J. Mulero, G. Masmitja, J. Zarco et al., ASDAS high disease activity versus BASDAI elevation in patients with ankylosing spondylitis as selection criterion for anti-TNF therapy, Reumatol Clin, vol.10, pp.204-213, 2014.

E. Gremese, S. Bernardi, S. Bonazza, M. Nowik, G. Peluso et al., Body weight, gender and response to TNF-? blockers in axial spondyloarthritis, Rheumatology, vol.53, pp.875-81, 2014.

B. Glintborg, P. Højgaard, L. Hetland, M. , S. Krogh et al., Impact of tobacco smoking on response to tumour necrosis factor-alpha inhibitor treatment in patients with ankylosing spondylitis: results from the Danish nationwide DANBIO registry, Rheumatology, vol.55, pp.659-68, 2016.

S. J. Pedersen, I. J. Sorensen, P. Garnero, J. S. Johansen, O. R. Madsen et al., BASDAI and different treatment responses and their relation to biomarkers of inflammation, cartilage and bone turnover in patients with axial spondyloarthritis treated with TNFalpha inhibitors, Ann Rheumat Dis, vol.70, pp.1375-81, 2011.

M. C. Turina, N. Yeremenko, J. E. Paramarta, L. De-rycke, and D. Baeten, Calprotectin (S100A8/9) as serum biomarker for clinical response in proof-of-concept trials in axial and peripheral spondyloarthritis, Arthritis Res Ther, vol.16, p.413, 2014.

I. Y. Choi, D. M. Gerlag, M. J. Herenius, R. M. Thurlings, C. A. Wijbrandts et al., MRP8/14 serum levels as a strong predictor of response to biological treatments in patients with rheumatoid arthritis, Ann Rheum Dis, vol.74, pp.499-505, 2015.

L. G. Van-baarsen, C. A. Wijbrandts, F. Rustenburg, T. Cantaert, T. C. Van-der-pouw-kraan et al., Regulation of IFN response gene activity during infliximab treatment in rheumatoid arthritis is associated with clinical response to treatment, Arthritis Res Ther, vol.12, p.11, 2010.

D. Koczan, S. Drynda, M. Hecker, A. Drynda, R. Guthke et al., Molecular discrimination of responders and nonresponders to anti-TNF alpha therapy in rheumatoid arthritis by etanercept, Arthritis Res Ther, vol.10, p.50, 2008.

B. Stuhlmüller, T. Häupl, M. M. Hernandez, A. Grützkau, R. Kuban et al., CD11c as a transcriptional biomarker to predict response to anti-TNF monotherapy with adalimumab in patients with rheumatoid arthritis, Clin Pharmacol Ther, vol.87, pp.311-332, 2010.

R. Gaujoux, E. Starosvetsky, N. Maimon, F. Vallania, H. Bar-yoseph et al., Cell-centred meta-analysis reveals baseline predictors of anti-TNF? non-response in biopsy and blood of patients with IB, Gut, vol.2018, pp.1-11, 2018.

N. R. West, A. N. Hegazy, B. Owens, S. J. Bullers, B. Linggi et al., Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease, Nat Med, vol.23, pp.579-89, 2017.

R. Atreya, H. Neumann, C. Neufert, M. J. Waldner, U. Billmeier et al., In vivo imaging using fluorescent antibodies to tumor necrosis factor predicts therapeutic response in Crohn's disease, Nat Med, vol.20, pp.313-321, 2014.

T. Bazin, K. B. Hooks, T. Barnetche, M. E. Truchetet, R. Enaud et al., Microbiota composition may predict anti-Tnf alpha response in spondyloarthritis patients: an exploratory study, Sci Rep, vol.8, p.5446, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01760163

S. R. Kapoor, A. Filer, M. A. Fitzpatrick, B. A. Fisher, P. C. Taylor et al., Metabolic profiling predicts response to anti-tumor necrosis factor ? therapy in patients with rheumatoid arthritis, Arthritis Rheum, vol.65, pp.1448-56, 2013.

S. Garrett, T. Jenkinson, L. G. Kennedy, H. Whitelock, P. Gaisford et al., A new approach to defining disease status in ankylosing spondylitis: the Bath Ankylosing Spondylitis Disease Activity Index, J Rheumatol, vol.21, pp.2286-91, 1994.

P. Machado, R. Landewé, E. Lie, T. K. Kvien, J. Braun et al., Ankylosing Spondylitis Disease Activity Score (ASDAS): defining cut-off values for disease activity states and improvement scores, Ann Rheum Dis, vol.70, pp.47-53, 2011.

P. Machado and R. Landewe, Spondyloarthritis: is it time to replace BASDAI with ASDAS?, Nat Rev Rheumatol, vol.9, pp.388-90, 2013.

A. Spoorenberg, D. Van-der-heijde, E. De-klerk, M. Dougados, K. De-vlam et al., Relative value of erythrocyte sedimentation rate and Creactive protein in assessment of disease activity in ankylosing spondylitis, J Rheumatol, vol.26, pp.980-984, 1999.

C. Gabay and I. Kushner, Acute-phase proteins and other systemic responses to inflammation, N Engl J Med, vol.340, pp.448-54, 1999.

J. Ruof and G. Stucki, Validity aspects of erythrocyte sedimentation rate and Creactive protein in ankylosing spondylitis: a literature review, J Rheumatol, vol.26, pp.966-70, 1999.

N. Vastesaeger, D. Van-der-heijde, R. D. Inman, Y. Wang, A. Deodhar et al., Predicting the outcome of ankylosing spondylitis therapy, Ann Rheumat Dis, vol.70, pp.973-81, 2011.

K. L. Hyrich, K. D. Watson, A. J. Silman, and D. Symmons, Predictors of response to anti-TNF-? therapy among patients with rheumatoid arthritis: results from the British Society for, Rheumatology Biologics Register. Rheumatology, vol.45, pp.1558-65, 2006.

S. Arends, E. Van-der-veer, H. Groen, P. M. Houtman, T. L. Jansen et al., Serum MMP-3 level as a biomarker for monitoring and predicting response to etanercept treatment in ankylosing spondylitis, J Rheumatol, vol.38, pp.1644-50, 2011.

M. C. Turina, J. Sieper, N. Yeremenko, K. Conrad, H. Haibel et al., Calprotectin serum level is an independent marker for radiographic spinal progression in axial spondyloarthritis, Ann Rheumat Dis, vol.73, pp.1746-1754, 2014.

J. Hobbs, R. May, K. Tanousis, E. Mcneill, M. Mathies et al., Identification of fungi with potential micotoxigenic in fish meals used for preparation, Mol Cell Biol, vol.23, pp.2564-76, 2003.

N. Leukert, T. Vogl, K. Strupat, R. Reichelt, C. Sorg et al., Calcium-dependent tetramer formation of S100A8 and S100A9 is essential for biological activity, J Mol Biol, vol.359, pp.961-72, 2006.

D. Foell and J. Roth, Proinflammatory S100 proteins in arthritis and autoimmune disease, Arthritis Rheumat, vol.50, pp.3762-71, 2004.

P. L. Van-lent, L. Grevers, A. B. Blom, A. Sloetjes, J. S. Mort et al., Myeloid-related proteins S100A8/S100A9 regulate joint inflammation and cartilage destruction during antigen-induced arthritis, Ann rheumat Dis, vol.67, pp.1750-1758, 2008.

T. Vogl, K. Tenbrock, S. Ludwig, N. Leukert, C. Ehrhardt et al., Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock, Nat Med, vol.13, pp.1042-1051, 2007.

K. Loser, T. Vogl, M. Voskort, A. Lueken, V. Kupas et al., The tolllike receptor 4 ligands Mrp8 and Mrp14 are crucial in the development of autoreactive CD8+T cells, Nat Med, vol.16, pp.713-720, 2010.

J. S. Smolen, R. Landewe, J. Bijlsma, G. Burmester, K. Chatzidionysiou et al., EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheumat Dis, vol.76, pp.960-77, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01778213

P. Ranganathan, Rheumatoid arthritis: biomarkers of response to TNF inhibition in RA, Nat Rev Rheumatol, vol.11, pp.446-454, 2015.

J. Cui, S. Saevarsdottir, B. Thomson, L. Padyukov, A. H. Van-der-helm-van-mil et al., Rheumatoid arthritis risk allele PTPRC is also associated with response to anti-tumor necrosis factor alpha therapy, Arthritis Rheumat, vol.62, pp.1849-61, 2010.

D. Plant, R. Prajapati, K. L. Hyrich, A. W. Morgan, A. G. Wilson et al., Replication of association of the PTPRC gene with response to anti-tumor necrosis factor therapy in a large UK cohort, Arthritis Rheumat, vol.64, pp.665-70, 2012.

D. A. Pappas, C. Oh, R. M. Plenge, J. M. Kremer, and J. D. Greenberg, Association of rheumatoid arthritis risk alleles with response to anti-TNF biologics: results from the CORRONA registry and meta-analysis, Inflammation, vol.36, pp.279-84, 2013.

J. Cui, E. A. Stahl, S. Saevarsdottir, C. Miceli, D. Diogo et al.,

, Genome-wide association study and gene expression analysis identifies CD84 as a predictor of response to etanercept therapy in rheumatoid arthritis, PLoS Genet, vol.9, p.1003394, 2013.

S. K. Sieberts, F. Zhu, J. Garcia-garcia, E. Stahl, A. Pratap et al., Crowdsourced assessment of common genetic contribution to predicting anti-TNF treatment response in rheumatoid arthritis, Nat Commun, vol.7, p.12460, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01420921

S. Aggarwal, N. Ghilardi, M. H. Xie, F. J. De-sauvage, and A. L. Gurney, Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17, J Biol Chem, vol.278, pp.1910-1914, 2003.

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.744-752, 2003.

C. L. Langrish, Y. Chen, W. M. Blumenschein, J. Mattson, B. Basham et al., IL-23 drives a pathogenic T cell population that induces autoimmune inflammation, J Exp Med, vol.201, pp.233-273, 2005.

M. J. Mcgeachy, Y. Chen, C. M. Tato, A. Laurence, B. Joyce-shaikh et al., The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo, Nat Immunol, vol.10, pp.314-338, 2009.

S. Buonocore, P. P. Ahern, H. H. Uhlig, . Ivanov, . Ii et al., Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology, Nature, vol.464, pp.1371-1376, 2010.

A. Geremia, C. V. Arancibia-carcamo, M. P. Fleming, N. Rust, B. Singh et al., IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease, J Exp Med, vol.208, pp.1127-1160, 2011.

C. E. Sutton, S. J. Lalor, C. M. Sweeney, C. F. Brereton, E. C. Lavelle et al., Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity, Immunity, vol.31, pp.331-372, 2009.

T. J. Kenna, S. I. Davidson, R. Duan, L. A. Bradbury, J. Mcfarlane et al., Enrichment of circulating interleukin-17-secreting interleukin-23 receptorpositive gamma/delta T cells in patients with active ankylosing spondylitis, Arthritis Rheum, vol.64, pp.1420-1429, 2012.

J. P. Sherlock, B. Joyce-shaikh, S. P. Turner, C. C. Chao, M. Sathe et al., IL-23 induces spondyloarthropathy by acting on ROR-gammat(+)

, CD8(-) entheseal resident T cells, CD3(+)CD4(-), vol.18, pp.1069-76, 2012.

M. A. Brown, T. Kenna, and B. P. Wordsworth, Genetics of ankylosing spondylitisinsights into pathogenesis, Nat Rev Rheumatol, vol.12, pp.81-91, 2016.

M. Coffre, M. Roumier, M. Rybczynska, E. Sechet, H. K. Law et al., Combinatorial control of Th17 and Th1 cell functions by genetic variations in genes associated with the interleukin-23 signaling pathway in spondyloarthritis, Arthritis Rheum, vol.65, pp.1510-1531, 2013.

D. Meglio, P. , D. Cesare, A. Laggner, U. Chu et al., The IL23R R381Q gene variant protects against immune-mediated diseases by impairing IL-23-induced Th17 effector response in humans, PLoS ONE, vol.6, 2011.

R. Sarin, X. Wu, and C. Abraham, Inflammatory disease protective R381Q IL23 receptor polymorphism results in decreased primary CD4+ and CD8+ human T-cell functional responses, Proc Natl Acad Sci, vol.108, pp.9560-9565, 2011.

B. P. Fairfax, P. Humburg, S. Makino, V. Naranbhai, D. Wong et al., Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression, Science, vol.343, p.1246949, 2014.

J. C. Knight, Approaches for establishing the function of regulatory genetic variants involved in disease, Genome Med, vol.6, p.92, 2014.

B. Piasecka, D. Duffy, A. Urrutia, H. Quach, P. E. Posseme et al., Distinctive roles of age, sex, and genetics in shaping transcriptional variation of human immune responses to microbial challenges, Proc Natl Acad Sci USA, vol.115, pp.488-97, 2018.
URL : https://hal.archives-ouvertes.fr/pasteur-01721850

G. E. Fragoulis, S. Siebert, and I. B. Mcinnes, Therapeutic targeting of IL-17 and IL-23 cytokines in immune-mediated diseases, Annu Rev Med, vol.67, pp.337-53, 2016.

C. Jandus, G. Bioley, J. P. Rivals, J. Dudler, D. Speiser et al., Increased numbers of circulating polyfunctional Th17 memory cells in patients with seronegative spondylarthritides, Arthritis Rheum, vol.58, pp.2307-2324, 2008.

H. Shen, J. C. Goodall, H. Gaston, and J. S. , Frequency and phenotype of peripheral blood Th17 cells in ankylosing spondylitis and rheumatoid arthritis, Arthritis Rheum, vol.60, pp.1647-56, 2009.

P. Bowness, A. Ridley, J. Shaw, A. T. Chan, I. Wong-baeza et al., Th17 cells expressing KIR3DL2+ and responsive to HLA-B27 homodimers are increased in ankylosing spondylitis, J Immunol, vol.186, pp.2672-80, 2011.

D. Baeten, X. Baraliakos, J. Braun, J. Sieper, P. Emery et al., Anti-interleukin-17A monoclonal antibody secukinumab in treatment of ankylosing spondylitis: a randomised, double-blind, placebo-controlled trial, Lancet, vol.382, pp.1705-1718, 2013.

D. Baeten, J. Sieper, J. Braun, X. Baraliakos, M. Dougados et al., 2016 update of the ASAS-EULAR management recommendations for axial spondyloarthritis, N Engl J Med, vol.373, pp.978-91, 2015.

R. G. Langley, B. E. Elewski, M. Lebwohl, K. Reich, C. E. Griffiths et al., Secukinumab in plaque psoriasis-results of two phase 3 trials, N Engl J Med, vol.371, pp.326-364, 2014.

I. B. Mcinnes, P. J. Mease, B. Kirkham, A. Kavanaugh, C. T. Ritchlin et al., Secukinumab, a human anti-interleukin-17A monoclonal antibody, in patients with psoriatic arthritis (FUTURE 2): a randomised, doubleblind, placebo-controlled, phase 3 trial, Lancet, vol.386, pp.1137-1183, 2015.

P. J. Mease, I. B. Mcinnes, B. Kirkham, A. Kavanaugh, P. Rahman et al., Secukinumab inhibition of interleukin-17A in patients with psoriatic arthritis, N Engl J Med, vol.373, pp.1329-1368, 2015.

W. Hueber, B. E. Sands, S. Lewitzky, M. Vandemeulebroecke, W. Reinisch et al., Secukinumab in Crohn's Disease Study, Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial, Gut, vol.61, pp.1693-700, 2012.

S. R. Targan, B. Feagan, S. Vermeire, R. Panaccione, G. Y. Melmed et al., A randomized, Double-blind, placebo-controlled phase 2 study of brodalumab in patients with moderate-to-severe crohn's disease, Am J Gastroenterol, vol.111, pp.727-765, 2015.

L. Van-mens, M. Van-de-sande, S. Menegatti, S. Chen, I. Blijdorp et al., Brief report: interleukin-17 blockade with secukinumab in peripheral spondyloarthritis impacts synovial Immunopathology without compromising systemic immune responses, Arthritis Rheumatol, vol.70, 1994.

G. G. Krueger, R. G. Langley, C. Leonardi, N. Yeilding, C. Guzzo et al., A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis, N Engl J Med, vol.356, pp.580-92, 2007.

C. L. Leonardi, A. B. Kimball, K. A. Papp, N. Yeilding, C. Guzzo et al., Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1), Lancet, vol.371, pp.1665-74, 2008.

K. A. Papp, R. G. Langley, M. Lebwohl, G. G. Krueger, P. Szapary et al., Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2), Lancet, vol.371, pp.726-732, 2008.

C. E. Griffiths, B. E. Strober, P. Van-de-kerkhof, V. Ho, R. Fidelus-gort et al., Comparison of ustekinumab and etanercept for moderate-tosevere psoriasis, N Engl J Med, vol.362, pp.118-146, 2010.

K. A. Papp, A. Blauvelt, M. Bukhalo, M. Gooderham, J. G. Krueger et al., Risankizumab versus ustekinumab for moderate-to-severe plaque psoriasis, N Engl J Med, vol.376, pp.1551-60, 2017.

C. A. Murphy, C. L. Langrish, Y. Chen, W. Blumenschein, T. Mcclanahan et al., Divergent pro-and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation, J Exp Med, vol.198, pp.1951-1958, 2003.

S. Hue, P. Ahern, S. Buonocore, M. C. Kullberg, D. J. Cua et al., Interleukin-23 drives innate and T cell-mediated intestinal inflammation, J Exp Med, vol.203, pp.2473-83, 2006.

D. Yen, J. Cheung, H. Scheerens, F. Poulet, T. Mcclanahan et al., IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6, J Clin Invest, vol.116, pp.1310-1316, 2006.

B. G. Feagan, W. J. Sandborn, D. Haens, G. Panes, J. Kaser et al., Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn's disease: a randomised, doubleblind, placebo-controlled phase 2 study, Lancet, vol.389, issue.17, pp.30570-30576, 2017.

B. G. Feagan, W. J. Sandborn, C. Gasink, D. Jacobstein, Y. Lang et al., Ustekinumab as Induction and Maintenance Therapy for Crohn's Disease, N Engl J Med, vol.375, pp.1946-60, 2016.

D. Baeten, M. Ostergaard, J. C. Wei, J. Sieper, P. Jarvinen et al., Risankizumab, an IL-23 inhibitor, for ankylosing spondylitis: results of a randomised, double-blind, placebo-controlled, proof-of-concept, dose-finding phase 2 study, Ann Rheum Dis, vol.77, pp.1295-302, 2018.

P. R. Burton, D. G. Clayton, L. R. Cardon, N. Craddock, P. Deloukas et al., Association scan of 14,500 nonsynonymous SNPs in four diseases identifies autoimmunity variants, Nat Genet, vol.39, pp.1329-1366, 2007.

D. Ellinghaus, L. Jostins, S. L. Spain, A. Cortes, J. Bethune et al., Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci, Nat. Genet, vol.48, pp.510-518, 2016.

A. Cortes, J. Hadler, J. P. Pointon, P. C. Robinson, and T. Karaderi, Identification of multiple risk variants for ankylosing spondylitis through high-density genotyping of immune-related loci, International Genetics of Ankylosing Spondylitis Consortium, vol.45, pp.730-738, 2013.

E. Gracey, Z. Qaiyum, I. Almaghlouth, D. Lawson, S. Karki et al., IL-7 primes IL-17 in mucosal-associated invariant T (MAIT) cells, which contribute to the Th17-axis in ankylosing spondylitis, Ann Rheum Dis, vol.75, pp.2124-2156, 2016.

M. N. Van-tok, N. S. Lao, C. R. Alvi, M. Pots, D. Van-de-sande et al., The initiation, but not the persistence, of experimental spondyloarthritis is dependent on interleukin-23 signaling, Front Immunol, vol.9, p.1550, 2018.