G. G. Kaplan, The global burden of IBD: from 2015 to 2025, Nat Rev Gastroenterol Hepatol. déc, vol.12, issue.12, pp.720-727, 2015.

N. A. Molodecky, I. S. Soon, D. M. Rabi, W. A. Ghali, M. Ferris et al., Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review, Gastroenterology. janv, vol.142, issue.1, pp.46-54, 2012.

N. P. Thompson, R. Driscoll, R. E. Pounder, and A. J. Wakefield, Genetics versus environment in inflammatory bowel disease: results of a British twin study, BMJ. 13 janv, vol.312, issue.7023, pp.95-101, 1996.

J. Halfvarson, L. Bodin, C. Tysk, E. Lindberg, and G. Järnerot, Inflammatory bowel disease in a Swedish twin cohort: a long-term follow-up of concordance and clinical characteristics, Gastroenterology. juin, vol.124, issue.7, pp.1767-73, 2003.

U. Monsén, O. Bernell, C. Johansson, and G. Hellers, Prevalence of inflammatory bowel disease among relatives of patients with Crohn's disease, Scand J Gastroenterol. mars, vol.26, issue.3, pp.302-308, 1991.

G. C. Parkes, K. Whelan, and J. O. Lindsay, Smoking in inflammatory bowel disease: impact on disease course and insights into the aetiology of its effect, J Crohns Colitis. août, vol.8, issue.8, pp.717-742, 2014.

B. M. Calkins, A meta-analysis of the role of smoking in inflammatory bowel disease, Dig Dis Sci. déc, vol.34, issue.12, pp.1841-54, 1989.

J. L. Benjamin, C. Hedin, A. Koutsoumpas, S. C. Ng, N. E. Mccarthy et al., Smokers with active Crohn's disease have a clinically relevant dysbiosis of the gastrointestinal microbiota, Inflamm Bowel Dis. juin, vol.18, issue.6, pp.1092-100, 2012.

V. Bergeron, V. Grondin, S. Rajca, M. Maubert, B. Pigneur et al., Current smoking differentially affects blood mononuclear cells from patients with Crohn's disease and ulcerative colitis: relevance to its adverse role in the disease, Inflamm Bowel Dis. juin, vol.18, issue.6, pp.1101-1112, 2012.

D. T. Jones, M. T. Osterman, M. Bewtra, and J. D. Lewis, Passive smoking and inflammatory bowel disease: a meta-analysis, Am J Gastroenterol. sept, vol.103, issue.9, pp.2382-93, 2008.

R. E. Andersson, G. Olaison, C. Tysk, and A. Ekbom, Appendectomy and protection against ulcerative colitis, N Engl J Med. 15 mars, vol.344, issue.11, pp.808-822, 2001.

H. T. Jackson, E. F. Mongodin, K. P. Davenport, C. M. Fraser, A. D. Sandler et al., Cultureindependent evaluation of the appendix and rectum microbiomes in children with and without appendicitis, PloS One, vol.9, issue.4, p.95414, 2014.

D. C. Baumgart, S. Thomas, I. Przesdzing, D. Metzke, C. Bielecki et al., Exaggerated inflammatory response of primary human myeloid dendritic cells to lipopolysaccharide in patients with inflammatory bowel disease, Clin Exp Immunol. sept, vol.157, issue.3, pp.423-459, 2009.

A. Franke, D. Mcgovern, J. C. Barrett, K. Wang, G. L. Radford-smith et al., Genomewide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci, Nat Genet. déc, vol.42, issue.12, pp.1118-1143, 2010.

R. Cooney, J. Baker, O. Brain, B. Danis, T. Pichulik et al., NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation, Nat Med. janv, vol.16, issue.1, pp.90-97, 2010.

G. M. Cobrin and M. T. Abreu, Defects in mucosal immunity leading to Crohn's disease, Immunol Rev. août, vol.206, pp.277-95, 2005.

S. R. Targan and L. C. Karp, Defects in mucosal immunity leading to ulcerative colitis, Immunol Rev. août, vol.206, pp.296-305, 2005.

I. J. Fuss, F. Heller, M. Boirivant, F. Leon, M. Yoshida et al., Nonclassical CD1drestricted NK T cells that produce IL-13 characterize an atypical Th2 response in ulcerative colitis, J Clin Invest. mai, vol.113, issue.10, pp.1490-1497, 2004.

H. Sokol and P. Seksik, The intestinal microbiota in inflammatory bowel diseases: time to connect with the host, Curr Opin Gastroenterol. juill, vol.26, issue.4, pp.327-358, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00599080

B. D. Muegge, J. Kuczynski, D. Knights, J. C. Clemente, A. González et al., Diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans, Science. 20 mai, vol.332, issue.6032, pp.970-974, 2011.

L. Dethlefsen and D. A. Relman, Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation, Proc Natl Acad Sci U S A. 15 mars, vol.108, issue.1, pp.4554-61, 2011.

A. Darfeuille-michaud, C. Neut, N. Barnich, E. Lederman, D. Martino et al., Presence of adherent Escherichia coli strains in ileal mucosa of patients with Crohn's disease, Gastroenterology. déc, vol.115, issue.6, pp.1405-1418, 1998.

H. Sokol, V. Leducq, H. Aschard, H. Pham, S. Jegou et al., Fungal microbiota dysbiosis in IBD, Gut. juin, vol.66, issue.6, pp.1039-1087, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01270909

S. Paramsothy, M. A. Kamm, N. O. Kaakoush, A. J. Walsh, J. Van-den-bogaerde et al.,

, Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial, Lancet Lond Engl. 25 mars, vol.389, pp.1218-1246, 2017.

G. Van-assche, A. Dignass, J. Panes, L. Beaugerie, J. Karagiannis et al., The second European evidence-based Consensus on the diagnosis and management of Crohn's disease: Definitions and diagnosis, J Crohns Colitis. févr, vol.4, issue.1, pp.7-27, 2010.

J. Torres, S. Mehandru, J. Colombel, P. , and L. , Crohn's disease, Lancet Lond Engl. 29 avr, vol.389, pp.1741-55, 2017.

T. W. Eglinton, M. L. Barclay, R. B. Gearry, and F. A. Frizelle, The spectrum of perianal Crohn's disease in a population-based cohort, Dis Colon Rectum. juill, vol.55, issue.7, pp.773-780, 2012.

S. Vermeire, M. Peeters, R. Vlietinck, S. Joossens, D. Hond et al., AntiSaccharomyces cerevisiae antibodies (ASCA), phenotypes of IBD, and intestinal permeability: a study in IBD families, Inflamm Bowel Dis. févr, vol.7, issue.1, pp.8-15, 2001.

S. Vermeire, S. Joossens, M. Peeters, F. Monsuur, G. Marien et al., Comparative study of ASCA (Anti-Saccharomyces cerevisiae antibody) assays in inflammatory bowel disease, Gastroenterology. mars, vol.120, issue.4, pp.827-860, 2001.

G. E. Reese, V. A. Constantinides, C. Simillis, A. W. Darzi, T. R. Orchard et al., Diagnostic precision of anti-Saccharomyces cerevisiae antibodies and perinuclear antineutrophil cytoplasmic antibodies in inflammatory bowel disease, Am J Gastroenterol. oct, vol.101, issue.10, pp.2410-2432, 2006.

S. B. Menees, C. Powell, J. Kurlander, A. Goel, and W. D. Chey, A meta-analysis of the utility of Creactive protein, erythrocyte sedimentation rate, fecal calprotectin, and fecal lactoferrin to exclude inflammatory bowel disease in adults with IBS, Am J Gastroenterol. mars, vol.110, issue.3, pp.444-54, 2015.

V. Kristensen and B. Moum, Correspondence: fecal calprotectin and cut-off levels in inflammatory bowel disease, Scand J Gastroenterol, vol.50, issue.9, pp.1183-1187, 2015.

D. 'haens, G. Ferrante, M. Vermeire, S. Baert, F. Noman et al., Fecal calprotectin is a surrogate marker for endoscopic lesions in inflammatory bowel disease, Inflamm Bowel Dis. déc, vol.18, issue.12, pp.2218-2242, 2012.

C. A. Rubio, A. Orrego, G. Nesi, and Y. Finkel, Frequency of epithelioid granulomas in colonoscopic biopsy specimens from paediatric and adult patients with Crohn's colitis, J Clin Pathol, vol.60, issue.11, pp.1268-72, 2007.

J. Panés, R. Bouzas, M. Chaparro, V. García-sánchez, J. P. Gisbert et al., Systematic review: the use of ultrasonography, computed tomography and magnetic resonance imaging for the diagnosis, assessment of activity and abdominal complications of Crohn's disease, Aliment Pharmacol Ther. juill, vol.34, issue.2, pp.125-170, 2011.

J. Cosnes, S. Cattan, A. Blain, L. Beaugerie, F. Carbonnel et al., Long-term evolution of disease behavior of Crohn's disease, Inflamm Bowel Dis. juill, vol.8, issue.4, pp.244-50, 2002.

L. Peyrin-biroulet, E. V. Loftus, J. Colombel, and W. J. Sandborn, The natural history of adult Crohn's disease in population-based cohorts, Am J Gastroenterol. févr, vol.105, issue.2, pp.289-97, 2010.

S. Ghosh and R. Mitchell, Impact of inflammatory bowel disease on quality of life: Results of the

, European Federation of Crohn's and Ulcerative Colitis Associations (EFCCA) patient survey, J Crohns Colitis. sept, vol.1, issue.1, pp.10-20, 2007.

E. Louis, A. Collard, A. F. Oger, E. Degroote, A. Yafi et al., Behaviour of Crohn's disease according to the Vienna classification: changing pattern over the course of the disease, Gut. déc, vol.49, issue.6, pp.777-82, 2001.

D. A. Schwartz, E. V. Loftus, W. J. Tremaine, R. Panaccione, W. S. Harmsen et al., The natural history of fistulizing Crohn's disease in Olmsted County, Minnesota. Gastroenterology. avr, vol.122, issue.4, pp.875-80, 2002.

C. Zallot and L. Peyrin-biroulet, Clinical risk factors for complicated disease: how reliable are they? Dig Dis Basel Switz, vol.30, pp.67-72, 2012.

J. Satsangi, M. S. Silverberg, S. Vermeire, and J. Colombel, The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications, Gut. juin, vol.55, issue.6, pp.749-53, 2006.

M. S. Silverberg, J. Satsangi, T. Ahmad, I. Arnott, C. N. Bernstein et al., Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a Working Party of the 2005 Montreal World Congress of Gastroenterology, Can J Gastroenterol J Can Gastroenterol. sept, vol.19, 2005.

J. Cosnes, C. Gower-rousseau, P. Seksik, and A. Cortot, Epidemiology and natural history of inflammatory bowel diseases, Gastroenterology. mai, vol.140, issue.6, pp.1785-94, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00631553

A. Dignass, R. Eliakim, F. Magro, C. Maaser, Y. Chowers et al., Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 1: definitions and diagnosis, J Crohns Colitis. déc, vol.6, issue.10, pp.965-90, 2012.

S. Vermeire, G. Van-assche, and P. Rutgeerts, The role of C-reactive protein as an inflammatory marker in gastrointestinal diseases, Nat Clin Pract Gastroenterol Hepatol. déc, vol.2, issue.12, pp.580-586, 2005.

C. Prantera, M. Davoli, R. Lorenzetti, F. Pallone, A. Marcheggiano et al., Clinical and laboratory indicators of extent of ulcerative colitis. Serum C-reactive protein helps the most, J Clin Gastroenterol. févr, vol.10, issue.1, pp.41-46, 1988.

G. Suarez and B. , Histological predictors of relapse in ulcerative colitis, Abstract OP056, 2017.

M. J. Etchevers, M. Aceituno, O. García-bosch, I. Ordás, M. Sans et al., Risk factors and characteristics of extent progression in ulcerative colitis, Inflamm Bowel Dis. sept, vol.15, issue.9, pp.1320-1325, 2009.

A. D. Levin, M. E. Wildenberg, and G. R. Van-den-brink, Mechanism of Action of Anti-TNF Therapy in Inflammatory Bowel Disease, J Crohns Colitis. août, vol.10, issue.8, pp.989-97, 2016.

S. R. Targan, S. B. Hanauer, S. J. Van-deventer, L. Mayer, D. H. Present et al., A shortterm 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, issue.15, pp.1029-1064, 1997.

S. B. Hanauer, B. G. Feagan, G. R. Lichtenstein, L. F. Mayer, S. Schreiber et al.,

, Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial, Lancet Lond Engl. 4 mai, vol.359, issue.9317, pp.1541-1550, 2002.

P. Rutgeerts, W. J. Sandborn, B. G. Feagan, W. Reinisch, A. Olson et al., Infliximab for induction and maintenance therapy for ulcerative colitis, N Engl J Med. 8 déc, vol.353, issue.23, pp.2462-76, 2005.

W. J. Sandborn, B. G. Feagan, C. Marano, H. Zhang, R. Strauss et al., Subcutaneous golimumab induces clinical response and remission in patients with moderate-to-severe ulcerative colitis, Gastroenterology. janv, vol.146, issue.1, pp.85-95, 2014.

L. Peyrin-biroulet, P. Deltenre, N. De-suray, J. Branche, W. J. Sandborn et al., Efficacy and safety of tumor necrosis factor antagonists in Crohn's disease: meta-analysis of placebo-controlled trials, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc. juin, vol.6, issue.6, pp.644-53, 2008.

G. R. Lichtenstein, B. G. Feagan, R. D. Cohen, B. A. Salzberg, R. H. Diamond et al., Serious infection and mortality in patients with Crohn's disease: more than 5 years of follow-up in the TREAT TM registry, Am J Gastroenterol. sept, vol.107, issue.9, pp.1409-1431, 2012.

A. C. Ford and L. Peyrin-biroulet, Opportunistic infections with anti-tumor necrosis factor-? therapy in inflammatory bowel disease: meta-analysis of randomized controlled trials, Am J Gastroenterol. août, vol.108, issue.8, pp.1268-76, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01706703

J. Kirchgesner, M. Lemaitre, A. Rudnichi, A. Racine, M. Zureik et al., Therapeutic management of inflammatory bowel disease in real-life practice in the current era of anti-TNF agents: analysis of the French administrative health databases, Aliment Pharmacol Ther, vol.45, issue.1, pp.37-49, 2009.

L. Peyrin-biroulet, Y. Bouhnik, X. Roblin, G. Bonnaud, H. Hagège et al., Algorithmes de prise en charge de la rectocolite hémorragique 2015 : Consensus National Français, Hépato Gastro, vol.23, pp.33-42, 2016.

L. Peyrin-biroulet, Y. Bouhnik, X. Roblin, G. Bonnaud, H. Hagege et al., Algorithmes de prise en charge de la maladie de Crohn en 2016 : Consensus National Français, Hépato Gastro, vol.23, pp.619-633, 2016.

D. 'haens, G. Baert, F. Van-assche, G. Caenepeel, P. Vergauwe et al., Early combined immunosuppression or conventional management in patients with newly diagnosed Crohn's disease: an open randomised trial, Lancet Lond Engl. 23 févr, vol.371, issue.9613, pp.660-667, 2008.

J. F. Colombel, W. J. Sandborn, W. Reinisch, G. J. Mantzaris, A. Kornbluth et al.,

, Infliximab, azathioprine, or combination therapy for Crohn's disease, N Engl J Med. 15 avr, vol.362, issue.15, pp.1383-95, 2010.

L. Beaugerie and S. H. Itzkowitz, Cancers Complicating Inflammatory Bowel Disease, N Engl J Med. 9 juill, vol.373, issue.2, p.195, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01301711

L. Peyrin-biroulet, W. Sandborn, B. E. Sands, W. Reinisch, W. Bemelman et al.,

, Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE): Determining Therapeutic Goals for Treat-to-Target, Am J Gastroenterol. sept, vol.110, issue.9, pp.1324-1362, 2015.

L. Peyrin-biroulet, M. Ferrante, F. Magro, S. Campbell, D. Franchimont et al., Results from the 2nd Scientific Workshop of the ECCO. I: Impact of mucosal healing on the course of inflammatory bowel disease, J Crohns Colitis, vol.5, issue.5, pp.477-83, 2011.

J. Colombel, R. Panaccione, P. Bossuyt, M. Lukas, F. Baert et al., Effect of tight control management on Crohn's disease (CALM): a multicentre, randomised, controlled phase 3 trial, Lancet Lond Engl, vol.23, pp.2779-89, 2018.

R. Panaccione, J. Colombel, P. Bossuyt, F. Baert, and T. Vanasek, Long-term costeffectiveness of tight control for Crohn, Abstract DOP065, 2018.

M. Allez, K. Karmiris, L. E. Van-assche, G. Ben-horin, S. Klein et al., Report of the ECCO pathogenesis workshop on anti-TNF therapy failures in inflammatory bowel diseases: definitions, frequency and pharmacological aspects, J Crohns Colitis, vol.4, issue.4, pp.355-66, 2010.

S. Ben-horin, U. Kopylov, and Y. Chowers, Optimizing anti-TNF treatments in inflammatory bowel disease, Autoimmun Rev. janv, vol.13, issue.1, pp.24-30, 2014.

J. P. Gisbert and J. Panés, Loss of response and requirement of infliximab dose intensification in Crohn's disease: a review, Am J Gastroenterol. mars, vol.104, issue.3, pp.760-767, 2009.

V. Billioud, W. J. Sandborn, and L. Peyrin-biroulet, Loss of response and need for adalimumab dose intensification in Crohn's disease: a systematic review, Am J Gastroenterol. avr, vol.106, issue.4, pp.674-84, 2011.

V. Billioud, D. Laharie, J. Filippi, X. Roblin, A. Oussalah et al., Adherence to adalimumab therapy in Crohn's disease: a French multicenter experience, Inflamm Bowel Dis. janv, vol.17, issue.1, pp.152-161, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01732435

E. A. Maser, R. Villela, M. S. Silverberg, and G. R. Greenberg, Association of trough serum infliximab to clinical outcome after scheduled maintenance treatment for Crohn's disease, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc, vol.4, issue.10, pp.1248-54, 2006.

O. J. Adedokun, W. J. Sandborn, B. G. Feagan, P. Rutgeerts, Z. Xu et al., Association between serum concentration of infliximab and efficacy in adult patients with ulcerative colitis, Gastroenterology. déc, vol.147, issue.6, pp.1296-1307, 2014.

X. Roblin, M. Rinaudo, D. Tedesco, E. Phelip, J. M. Genin et al., Development of an algorithm incorporating pharmacokinetics of adalimumab in inflammatory bowel diseases, Am J Gastroenterol. août, vol.109, issue.8, pp.1250-1256, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01688079

N. Vande-casteele, R. Khanna, B. G. Levesque, L. Stitt, G. Y. Zou et al., The relationship between infliximab concentrations, antibodies to infliximab and disease activity in Crohn's disease. Gut, vol.64, pp.1539-1584, 2015.

I. Ordás, B. G. Feagan, and W. J. Sandborn, Therapeutic drug monitoring of tumor necrosis factor antagonists in inflammatory bowel disease, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc, vol.10, issue.10, pp.85-86, 2012.

X. Roblin, H. Marotte, M. Rinaudo, D. Tedesco, E. Moreau et al., Association between pharmacokinetics of adalimumab and mucosal healing in patients with inflammatory bowel diseases, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc. janv, vol.12, issue.1, pp.80-84, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01687260

W. Afif, E. V. Loftus, W. A. Faubion, S. V. Kane, D. H. Bruining et al., Clinical utility of measuring infliximab and human anti-chimeric antibody concentrations in patients with inflammatory bowel disease, Am J Gastroenterol. mai, vol.105, issue.5, pp.1133-1142, 2010.

J. F. Colombel, W. J. Sandborn, and P. Rutgeerts, Adalimumab for maintenance of clinical response and remission in patients with Crohn's disease: the CHARM trial, J Pediatr Gastroenterol Nutr. févr, vol.132, issue.2, pp.226-233, 2007.

S. Ben-horin and Y. Chowers, Review article: loss of response to anti-TNF treatments in Crohn's disease, Aliment Pharmacol Ther. mai, vol.33, issue.9, pp.987-95, 2011.

M. Regueiro, B. Siemanowski, K. E. Kip, and S. Plevy, Infliximab dose intensification in Crohn's disease, Inflamm Bowel Dis. sept, vol.13, issue.9, pp.1093-1102, 2007.

B. Pariente, P. De-chambrun, G. Krzysiek, R. Desroches, M. Louis et al., Trough levels and antibodies to infliximab may not predict response to intensification of infliximab therapy in patients with inflammatory bowel disease, Inflamm Bowel Dis. juill, vol.18, issue.7, pp.1199-206, 2012.

F. Baert, M. Noman, S. Vermeire, G. Van-assche, D. '-haens et al., Influence of immunogenicity on the long-term efficacy of infliximab in Crohn's disease, N Engl J Med. 13 févr, vol.348, issue.7, pp.601-609, 2003.

N. Vande-casteele, A. Gils, S. Singh, L. Ohrmund, S. Hauenstein et al., Antibody response to infliximab and its impact on pharmacokinetics can be transient, Am J Gastroenterol. juin, vol.108, issue.6, pp.962-71, 2013.

B. Ungar, Y. Chowers, M. Yavzori, O. Picard, E. Fudim et al., The temporal evolution of antidrug antibodies in patients with inflammatory bowel disease treated with infliximab, Gut. août, vol.63, issue.8, pp.1258-64, 2014.

F. Baert, M. Noman, S. Vermeire, G. Van-assche, D. '-haens et al., Influence of immunogenicity on the long-term efficacy of infliximab in Crohn's disease, N Engl J Med. 13 févr, vol.348, issue.7, pp.601-609, 2003.

A. S. Strik, G. R. Van-den-brink, C. Ponsioen, R. Mathot, M. Löwenberg et al., Suppression of anti-drug antibodies to infliximab or adalimumab with the addition of an immunomodulator in patients with inflammatory bowel disease, Aliment Pharmacol Ther, vol.45, issue.8, pp.1128-1162, 2017.

L. Peyrin-biroulet, J. Salleron, J. Filippi, C. Reenaers, O. Antunes et al., Anti-TNF Monotherapy for Crohn's Disease: a 13-year Multicentre Experience, J Crohns Colitis. mai, vol.10, issue.5, pp.516-540, 2016.

S. Ben-horin, M. Waterman, U. Kopylov, M. Yavzori, O. Picard et al., Addition of an immunomodulator to infliximab therapy eliminates antidrug antibodies in serum and restores clinical response of patients with inflammatory bowel disease, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc. avr, vol.11, issue.4, pp.444-451, 2013.

K. Papamichael, P. Karatzas, and G. J. Mantzaris, Addition of an immunomodulator as a rescue therapy for loss of response to adalimumab dose escalation in patients with Crohn's disease, J Crohns Colitis. juill, vol.9, issue.7, pp.589-90, 2015.

C. Ma, R. Panaccione, S. J. Heitman, S. M. Devlin, S. Ghosh et al., Systematic review: the short-term and long-term efficacy of adalimumab following discontinuation of infliximab, Aliment Pharmacol Ther, vol.30, issue.10, pp.977-86, 2009.

B. G. Feagan, G. R. Greenberg, G. Wild, R. N. Fedorak, P. Paré et al., Treatment of ulcerative colitis with a humanized antibody to the alpha4beta7 integrin, N Engl J Med. 16 juin, vol.352, issue.24, pp.2499-507, 2005.

M. Briskin, D. Winsor-hines, A. Shyjan, N. Cochran, S. Bloom et al., Human mucosal addressin cell adhesion molecule-1 is preferentially expressed in intestinal tract and associated lymphoid tissue, Am J Pathol. juill, vol.151, issue.1, pp.97-110, 1997.

W. J. Sandborn, B. G. Feagan, P. Rutgeerts, S. Hanauer, J. Colombel et al., Vedolizumab as induction and maintenance therapy for Crohn's disease, N Engl J Med. 22 août, vol.369, issue.8, pp.711-732, 2013.

B. G. Feagan, P. Rutgeerts, B. E. Sands, S. Hanauer, J. Colombel et al., Vedolizumab as induction and maintenance therapy for ulcerative colitis, N Engl J Med. 22 août, vol.369, issue.8, pp.699-710, 2013.

B. E. Sands, B. G. Feagan, P. Rutgeerts, J. Colombel, W. J. Sandborn et al., Effects of vedolizumab induction therapy for patients with Crohn's disease in whom tumor necrosis factor antagonist treatment failed, Gastroenterology. sept, vol.147, issue.3, pp.618-627, 2014.

S. Danese, B. Feagan, W. Sandborn, J. Colombel, S. Vermeire et al., A phase 3b open-label multicentre study (VERSIFY) of the efficacy of vedolizumab on endoscopic healing in moderately to severely active Crohn, Abstract OP023, 2018.

M. Noman, M. Ferrante, R. Bisschops, D. Hertogh, G. Van-den-broeck et al.,

, Vedolizumab Induces Long-term Mucosal Healing in Patients With Crohn's Disease and Ulcerative Colitis, J Crohns Colitis. 1 sept, vol.11, issue.9, pp.1085-1094, 2017.

A. Amiot, J. Grimaud, L. Peyrin-biroulet, J. Filippi, B. Pariente et al., Effectiveness and Safety of Vedolizumab Induction Therapy for Patients With Inflammatory Bowel Disease, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc, vol.14, issue.11, pp.1593-1601, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01278611

W. J. Sandborn, J. F. Colombel, R. Enns, B. G. Feagan, S. B. Hanauer et al., Natalizumab induction and maintenance therapy for Crohn's disease, N Engl J Med, vol.353, issue.18, pp.1912-1937, 2005.

J. Colombel, B. E. Sands, P. Rutgeerts, W. Sandborn, S. Danese et al., The safety of vedolizumab for ulcerative colitis and Crohn's disease, Gut. mai, vol.66, issue.5, pp.839-51, 2017.

E. V. Loftus, J. Colombel, B. G. Feagan, S. Vermeire, W. J. Sandborn et al., Long-term Efficacy of Vedolizumab for Ulcerative Colitis, J Crohns Colitis. 1 avr, vol.11, issue.4, pp.400-411, 2017.

S. Vermeire, E. V. Loftus, J. Colombel, B. G. Feagan, W. J. Sandborn et al., Long-term Efficacy of Vedolizumab for Crohn's Disease, J Crohns Colitis. 1 avr, vol.11, issue.4, pp.412-436, 2017.

M. Rosario, N. L. Dirks, M. R. Gastonguay, A. A. Fasanmade, T. Wyant et al., Population pharmacokinetics-pharmacodynamics of vedolizumab in patients with ulcerative colitis and Crohn's disease, Aliment Pharmacol Ther. juill, vol.42, issue.2, pp.188-202, 2015.

M. Rosario, J. L. French, N. L. Dirks, S. Sankoh, A. Parikh et al., Exposure-efficacy Relationships for Vedolizumab Induction Therapy in Patients with Ulcerative Colitis or Crohn's Disease, J Crohns Colitis. 1 août, vol.11, issue.8, pp.921-930, 2017.

N. Williet, G. Boschetti, M. Fovet, D. Bernado, T. Claudez et al., Association Between Low Trough Levels of Vedolizumab During Induction Therapy for Inflammatory Bowel Diseases and Need for Additional Doses Within 6 Months, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc, vol.15, issue.11, pp.1750-1757, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01908856

W. Yacoub, N. Williet, L. Pouillon, T. Di-bernado, D. Carvalho-bittencourt et al., Early vedolizumab trough levels predict mucosal healing in inflammatory bowel disease: a multicentre prospective observational study, Aliment Pharmacol Ther. 31 janv, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01720723

J. Earp, L. Fang, L. Ma, Y. Wang, and M. Rosario, Assessing Labeling Claims for Drug Interactions using a Population PK Approach: Vedolizumab. ACOP5 Abstract, 2017.

D. Hudesman, S. Chang, P. Shashi, A. Winters, and S. Chablaney, Impact of concomitant immunomodulator use on vedolizumab effectiveness: a multicentre consortium propensity scorematched analysis. DOP053 Abstract, 2018.

A. Parikh, T. Leach, T. Wyant, C. Scholz, S. Sankoh et al., Vedolizumab for the treatment of active ulcerative colitis: a randomized controlled phase 2 dose-ranging study, Inflamm Bowel Dis. août, vol.18, issue.8, pp.1470-1479, 2012.

B. Ungar, U. Kopylov, M. Yavzori, E. Fudim, O. Picard et al., Association of Vedolizumab Level, Anti-drug Antibodies, and ?4?7 Occupancy With Response in Patients With Inflammatory Bowel Diseases, Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc. 6 déc, 2017.

J. Buchanan, S. Wordsworth, T. Ahmad, A. Perrin, S. Vermeire et al., Managing the long term care of inflammatory bowel disease patients: The cost to European health care providers, J Crohns Colitis. août, vol.5, issue.4, pp.301-317, 2011.

S. Danese, L. Vuitton, and L. Peyrin-biroulet, Biologic agents for IBD: practical insights, Nat Rev Gastroenterol Hepatol. sept, vol.12, issue.9, pp.537-582, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01667747

M. E. Van-der-valk, M. Mangen, M. Leenders, G. Dijkstra, A. A. Van-bodegraven et al., Healthcare costs of inflammatory bowel disease have shifted from hospitalisation and surgery towards anti-TNF? therapy: results from the COIN study, Gut. janv, vol.63, issue.1, pp.72-81, 2014.

M. R. Wilson, A. Bergman, H. Chevrou-severac, R. Selby, M. Smyth et al., Costeffectiveness of vedolizumab compared with infliximab, adalimumab, and golimumab in patients with ulcerative colitis in the United Kingdom, Eur J Health Econ HEPAC Health Econ Prev Care. 8 mars, 2017.

L. Yokomizo, B. Limketkai, and K. T. Park, Cost-effectiveness of adalimumab, infliximab or vedolizumab as first-line biological therapy in moderate-to-severe ulcerative colitis, BMJ Open Gastroenterol, vol.3, issue.1, p.93, 2016.

W. J. Sandborn, C. Gasink, L. Gao, M. A. Blank, J. Johanns et al., Ustekinumab induction and maintenance therapy in refractory Crohn's disease, N Engl J Med, vol.367, issue.16, pp.1519-1547, 2012.

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.17, issue.20, pp.1946-60, 2016.

W. J. Sandborn, C. Su, B. E. Sands, D. Haens, G. R. Vermeire et al., Tofacitinib as Induction and Maintenance Therapy for Ulcerative Colitis, N Engl J Med, vol.04, issue.18, pp.1723-1759, 2017.

?. Letter, addition of methotrexate neither restores clinical response nor improve the pharmacokinetic profile of vedolizumab

A. S. Strik, G. R. Van-den-brink, and C. Ponsioen, Suppression of anti-drug antibodies to infliximab or adalimumab with the addition of an immunomodulator in patients with inflammatory bowel disease, Aliment Pharmacol Ther, vol.45, pp.1128-1134, 2017.

L. Peyrin-biroulet, J. Salleron, and J. Filippi, Anti TNF monotherapy for Crohn's disease : a 13year Multicentre Experience, J Crohns Colitis, vol.10, pp.516-540, 2016.

S. Ben-horin, M. Waterman, and U. Kopylov, Addition of an immunomodulator to infliximab therapy eliminates antidrug antibodies in serum and restores clinical response of patients with inflammatory bowel disease, Clin Gastroenterol Hepatol, vol.11, pp.444-447, 2013.

K. Papamichael, P. Karatzas, and G. J. Mantzaris, Addition of an immunomodulator as a rescue therapy for loss of response to adalimumab dose escalation in patients with Crohn's disease, J Crohn's Colitis, vol.10, pp.589-590, 2015.

W. Sandborn and B. G. Feagan, Vedolizumab as induction and maintenance therapy for Crohn's disease, N Engl J Med, vol.369, pp.711-732, 2013.

?. , Early changes in the pharmacokinetic profile of vedolizumab-treated patients with inflammatory bowel disease may predict response after dose optimization

J. Colombel, B. E. Sands, and P. Rutgeerts, The safety of vedolizumab for ulcerative colitis and Crohn's disease, Gut, vol.66, pp.839-51, 2017.

M. Rosario, J. L. French, and N. L. Dirks, Exposure-efficacy Relationships for Vedolizumab Induction Therapy in Patients with Ulcerative Colitis or Crohn's Disease, J Crohns Colitis, vol.11, pp.921-930, 2017.
DOI : 10.1093/ecco-jcc/jjx021

E. Loftus, J. Colombel, and B. G. Feagan, Long-term Efficacy of Vedolizumab for Ulcerative Colitis, J Crohns Colitis, vol.11, pp.400-411, 2017.
DOI : 10.1093/ecco-jcc/jjw177

URL : https://academic.oup.com/ecco-jcc/article-pdf/11/4/400/17682347/jjw177.pdf

S. Vermeire, E. Loftus, and J. Colombel, Long-term Efficacy of Vedolizumab for Crohn's Disease, J Crohns Colitis, vol.11, pp.412-436, 2017.
DOI : 10.1093/ecco-jcc/jjw176

URL : https://academic.oup.com/ecco-jcc/article-pdf/11/4/412/17682337/jjw176.pdf