P. Agostinis, K. Berg, and K. Cengel, Photodynamic therapy of cancer: An update, CA: A Cancer Journal for Clinicians, vol.4, issue.1, pp.250-81, 2011.
DOI : 10.3322/caac.20114

L. Ratkay, J. Waterfield, and D. Hunt, Photodynamic Therapy in Immune (Non-Oncological) Disorders, BioDrugs, vol.3914, issue.2, pp.127-162, 2000.
DOI : 10.2165/00063030-200014020-00006

H. Lassalle, D. Dumas, S. Grafe, D. Hallewin, M. Guillemin et al., Correlation between in vivo pharmacokinetics, intratumoral distribution and photodynamic efficiency of liposomal mTHPC, Journal of Controlled Release, vol.134, issue.2, pp.118-142, 2009.
DOI : 10.1016/j.jconrel.2008.11.016

URL : https://hal.archives-ouvertes.fr/hal-00338449

M. Bovis, J. Woodhams, and M. Loizidou, Improved in vivo delivery of m-THPC via pegylated liposomes for use in photodynamic therapy, Journal of Controlled Release, vol.157, issue.2, pp.196-205, 2012.
DOI : 10.1016/j.jconrel.2011.09.085

M. Senge and J. Brandt, (m-hydroxyphenyl)chlorin)--a secondgeneration photosensitizer, Temoporfin (Foscan(R) Photochem Photobiol, vol.5101587, pp.20-1240, 2011.

V. Reshetov, H. Lassalle, and A. Francois, Photodynamic therapy with conventional and PEGylated liposomal formulations of mTHPC (temoporfin): comparison of treatment efficacy and distribution characteristics in vivo, International Journal of Nanomedicine, vol.8, pp.3817-3848, 2013.
DOI : 10.2147/IJN.S51002

URL : https://hal.archives-ouvertes.fr/hal-00875936

J. Garrier, L. Bezdetnaya, and C. Barlier, Foslip??-based photodynamic therapy as a means to improve wound healing, Photodiagnosis and Photodynamic Therapy, vol.8, issue.4, pp.321-328, 2011.
DOI : 10.1016/j.pdpdt.2011.06.003

URL : https://hal.archives-ouvertes.fr/hal-00598501

A. Hansch, O. Frey, M. Gajda, G. Susanna, J. Boettcher et al., Photodynamic treatment as a novel approach in the therapy of arthritic joints, Lasers in Surgery and Medicine, vol.50, issue.4, pp.265-72, 2008.
DOI : 10.1002/lsm.20620

V. Garcia, M. De-lima, and T. Okamoto, Effect of photodynamic therapy on the healing of cutaneous third-degree-burn: histological study in rats, Lasers in Medical Science, vol.21, issue.Suppl, pp.221-229, 2010.
DOI : 10.1007/s10103-009-0694-z

G. Jori, C. Fabris, M. Soncin, S. Ferro, O. Coppellotti et al., Photodynamic therapy in the treatment of microbial infections: Basic principles and perspective applications, Lasers in Surgery and Medicine, vol.48, issue.5, pp.468-81, 2006.
DOI : 10.1002/lsm.20361

D. King, H. Jiang, G. Simkin, M. Obochi, J. Levy et al., Photodynamic Alteration of the Surface Receptor Expression Pattern of Murine Splenic Dendritic Cells, Scandinavian Journal of Immunology, vol.49, issue.2, pp.184-92, 1999.
DOI : 10.1046/j.1365-3083.1999.00498.x

G. Simkin, J. Tao, J. Levy, and D. Hunt, IL-10 Contributes to the Inhibition of Contact Hypersensitivity in Mice Treated with Photodynamic Therapy, The Journal of Immunology, vol.164, issue.5, pp.2457-62, 2000.
DOI : 10.4049/jimmunol.164.5.2457

J. Byun, G. Lee, and H. Choi, and IL-10 in Cultured Fibroblasts after ALA-IPL Photodynamic Treatment, Annals of Dermatology, vol.23, issue.1, pp.19-22, 2011.
DOI : 10.5021/ad.2011.23.1.19

W. Boehncke, T. Elshorst-schmidt, and R. Kaufmann, Systemic Photodynamic Therapy Is a Safe and Effective Treatment for Psoriasis, Archives of Dermatology, vol.136, issue.2, pp.271-273, 2000.
DOI : 10.1001/archderm.136.2.271

L. Favre, F. Borle, and D. Velin, Low dose endoluminal photodynamic therapy improves murine T cell-mediated colitis, Endoscopy, vol.43, issue.07, pp.604-620, 2011.
DOI : 10.1055/s-0030-1256382

T. Jess, C. Rungoe, and L. Peyrin-biroulet, Risk of Colorectal Cancer in Patients With Ulcerative Colitis: A Meta-analysis of Population-Based Cohort Studies, Clinical Gastroenterology and Hepatology, vol.10, issue.6, pp.639-684, 2012.
DOI : 10.1016/j.cgh.2012.01.010

S. Wirtz, C. Neufert, and B. Weigmann, Chemically induced mouse models of intestinal inflammation Neurath MF, Fuss I, Kelsall BL, et al. Antibodies to interleukin 12 abrogate established experimental colitis in mice, Nat Protocols.J Exp Med, vol.2182, issue.18, pp.541-61281, 1995.

P. Lepage, P. Seksik, and M. Sutren, Biodiversity of the Mucosa-Associated Microbiota Is Stable Along the Distal Digestive Tract in Healthy Individuals and Patients With Ibd, Inflammatory Bowel Diseases, vol.11, issue.5, pp.473-80, 2005.
DOI : 10.1097/01.MIB.0000159662.62651.06

P. Seksik, L. Rigottier-gois, and G. Gramet, Alterations of the dominant faecal bacterial groups in patients with Crohn's disease of the colon, Gut, vol.52, issue.2, pp.237-279, 2003.
DOI : 10.1136/gut.52.2.237

C. Maynard, L. Harrington, and K. Janowski, Regulatory T cells expressing interleukin 10 develop from Foxp3+ and Foxp3??? precursor cells in the absence of interleukin 10, Nature Immunology, vol.171, issue.9, pp.931-972, 2007.
DOI : 10.1016/j.immuni.2006.09.013

D. Colussi, G. Brandi, and F. Bazzoli, Molecular Pathways Involved in Colorectal Cancer: Implications for Disease Behavior and Prevention, International Journal of Molecular Sciences, vol.14, issue.8, pp.16365-85, 2013.
DOI : 10.3390/ijms140816365

M. Suganuma, S. Okabe, and M. Kurusu, Discrete roles of cytokines, TNF-alpha, IL-1, IL-6 in tumor promotion and cell transformation, Int J Oncol, vol.20, pp.131-137, 2002.

D. Seril, J. Liao, and G. Yang, Oxidative stress and ulcerative colitis-associated carcinogenesis: studies in humans and animal models, Carcinogenesis, vol.24, issue.3, pp.353-62, 2003.
DOI : 10.1093/carcin/24.3.353

A. Walker and T. Lawley, Therapeutic modulation of intestinal dysbiosis, Pharmacological Research, vol.69, issue.1, pp.75-86, 2013.
DOI : 10.1016/j.phrs.2012.09.008

A. Couturier-maillard, T. Secher, and A. Rehman, NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer, Journal of Clinical Investigation, vol.123, pp.700-711, 2013.
DOI : 10.1172/JCI62236DS1

URL : https://hal.archives-ouvertes.fr/hal-00944194

J. Triantafillidis, E. Merikas, and F. Georgopoulos, Current and emerging drugs for the treatment of inflammatory bowel disease, Drug Design, Development and Therapy, vol.5, pp.185-210, 2011.
DOI : 10.2147/DDDT.S11290

Y. Meissner and A. Lamprecht, Alternative Drug Delivery Approaches for the Therapy of Inflammatory Bowel Disease, Journal of Pharmaceutical Sciences, vol.97, issue.8, pp.2878-91, 2008.
DOI : 10.1002/jps.21216

W. Ulbrich and A. Lamprecht, Targeted drug-delivery approaches by nanoparticulate carriers in the therapy of inflammatory diseases, Journal of The Royal Society Interface, vol.90, issue.12, pp.55-66, 2010.
DOI : 10.1016/S1474-4422(04)00937-8

R. Jayasree, A. Gupta, and K. Rathinam, The Influence of Photodynamic Therapy on the Wound Healing Process in Rats, Journal of Biomaterials Applications, vol.15, issue.3, pp.176-86, 2001.
DOI : 10.1106/9335-Q0NC-5XCQ-KBYK

J. Silva, Z. Lacava, and S. Kuckelhaus, Evaluation of the use of low level laser and photosensitizer drugs in healing, Lasers in Surgery and Medicine, vol.7, issue.5, pp.451-458, 2004.
DOI : 10.1002/lsm.20062

S. Coutier, L. Bezdetnaya, and S. Marchal, Foscan?? (mTHPC) photosensitized macrophage activation: enhancement of phagocytosis, nitric oxide release and tumour necrosis factor-??-mediated cytolytic activity, British Journal of Cancer, vol.81, issue.1, pp.37-42, 1999.
DOI : 10.1038/sj.bjc.6690648

S. Gollnick, B. Lee, and L. Vaughan, Activation of the IL-10 Gene Promoter Following Photodynamic Therapy of Murine Keratinocytes??, Photochemistry and Photobiology, vol.160, issue.2, pp.170-177, 2001.
DOI : 10.1562/0031-8655(2001)073<0170:AOTIGP>2.0.CO;2

Y. Li, L. Cai, and H. Wang, Pleiotropic regulation of macrophage polarization and tumorigenesis by formyl peptide receptor-2, Oncogene, vol.830, pp.3887-99

J. Garrier, V. Reshetov, and V. Zorin, Contrasting Facets of nanoparticules-based phototherapy: photodamage and photo-regeneration. Mohamed L. Elsaie Photodynamic Therapy: New Research, Medical Procedures, Testing and Technology, pp.3-38, 2013.

W. Zhao, L. Qi, and Y. Qin, Functional comparison between genes dysregulated in ulcerative colitis and colorectal carcinoma Chronic inflammation in cancer development, PloS one. Front Immunol, vol.82, issue.8, p.98, 2011.

D. Rubin, A. Shaker, M. Levin, J. Boshuizen, and D. Van-nispen, Chronic intestinal inflammation: inflammatory bowel disease and colitisassociated colon cancer Alterations in Muc2 biosynthesis and secretion during dextran sulfate sodium-induced colitis, Front Immunol. Am J physiol Gastrointest Liver Physiol, vol.3282, pp.382-391, 2002.

L. Poritz, K. Garver, and C. Green, Loss of the Tight Junction Protein ZO-1 in Dextran Sulfate Sodium Induced Colitis, Journal of Surgical Research, vol.140, issue.1, pp.12-21, 2007.
DOI : 10.1016/j.jss.2006.07.050

R. Marcon, R. Claudino, and R. Dutra, Exacerbation of DSS-induced colitis in mice lacking kinin B(1) receptors through compensatory up-regulation of kinin B(2) receptors: the role of tight junctions and intestinal homeostasis, Br J Pharmacol, vol.16, pp.389-402, 2013.

I. Sobhani, A. Amiot, L. Baleur, and Y. , Microbial dysbiosis and colon carcinogenesis: could colon cancer be considered a bacteria-related disease? Development of novel oral formulations prepared via hot melt extrusion for targeted delivery of photosensitizer to the colon, Therap Adv Gastroenterol. Photochem Photobiol, vol.687, issue.43, pp.215-29867, 2011.

W. Fabry, 1560-1624)-the other fabricius, JAMA, p.933, 0190.

B. Burril, L. G. Crohn, and G. D. Landmark-article, Regional ileitis. A pathological and clinical entity, JAMA, vol.251, pp.73-79, 1932.

J. Cosnes, Epidemiology and Natural History of Inflammatory Bowel Diseases, Gastroenterology, vol.140, issue.6, pp.1785-94
DOI : 10.1053/j.gastro.2011.01.055

URL : https://hal.archives-ouvertes.fr/hal-00631553

M. Gasparetto and G. Guariso, Highlights in IBD Epidemiology and Its Natural History in the Paediatric Age, Gastroenterology Research and Practice, vol.57, issue.3, p.829040, 2013.
DOI : 10.1038/ajg.2009.177

E. V. Loftus and . Jr, Clinical epidemiology of inflammatory bowel disease: incidence, prevalence, and environmental influences, Gastroenterology, vol.126, issue.6, pp.1504-1521, 2004.
DOI : 10.1053/j.gastro.2004.01.063

M. D. Kappelman, Recent Trends in the Prevalence of Crohn???s Disease and Ulcerative Colitis in a Commercially Insured US Population, Digestive Diseases and Sciences, vol.132, issue.2, pp.519-544, 2013.
DOI : 10.1007/s10620-012-2371-5

F. T. Veloso, Extraintestinal manifestations of inflammatory bowel disease: Do they influence treatment and outcome?, World Journal of Gastroenterology, vol.17, issue.22, pp.2702-2709
DOI : 10.3748/wjg.v17.i22.2702

S. Ardizzone, Extraintestinal manifestations of inflammatory bowel disease. Dig Liver Dis, pp.253-262, 2008.

R. Caprilli, European evidence based consensus on the diagnosis and management of Crohn's disease: special situations, Gut, vol.55, issue.suppl_1, pp.36-58, 2006.
DOI : 10.1136/gut.2005.081950c

M. E. Spehlmann, Epidemiology of inflammatory bowel disease in a German twin cohort: Results of a nationwide study, Inflammatory Bowel Diseases, vol.14, issue.7, pp.14-968, 2008.
DOI : 10.1002/ibd.20380

P. Hruz and L. Eckmann, Innate immune defence: NOD2 and autophagy in the pathogenesis of Crohn???s disease, Swiss Medical Weekly, vol.140, p.13135
DOI : 10.4414/smw.2010.13135

T. Watanabe, NOD2 is a negative regulator of Toll-like receptor 2???mediated T helper type 1 responses, Nature Immunology, vol.52, issue.8, pp.800-808, 2004.
DOI : 10.1016/S0006-291X(02)02807-3

B. Khor, A. Gardet, R. J. Xavier-lees, and C. W. , Genetics and pathogenesis of inflammatory bowel disease New IBD genetics: common pathways with other diseases Geographical variations of inflammatory bowel disease in France: a study based on national health insurance data, Nature. Gut Inflamm Bowel Dis, vol.474, issue.163, pp.307-324, 2006.

V. Pinsk, Inflammatory Bowel Disease in the South Asian Pediatric Population of British Columbia, The American Journal of Gastroenterology, vol.100, issue.5, pp.1077-83, 2007.
DOI : 10.1136/adc.88.11.995

F. Carbonnel, Environmental risk factors in Crohn's disease and ulcerative colitis: an update Smoking cessation and the course of Crohn's disease: an intervention study, Gastroenterol Clin Biol Gastroenterology, vol.33, issue.1205, pp.1093-1102, 2001.

B. M. Calkins and H. S. Odes, A meta-analysis of the role of smoking in inflammatory bowel disease Effects of current cigarette smoking on clinical course of Crohn's disease and ulcerative colitis Ulcerative colitis in smokers, non-smokers and ex-smokers, Dig Dis Sci Dig Dis Sci World J Gastroenterol, vol.34, issue.2222, pp.1841-54, 1989.

R. E. Andersson, Appendectomy and Protection against Ulcerative Colitis, New England Journal of Medicine, vol.344, issue.11, pp.808-822, 2001.
DOI : 10.1056/NEJM200103153441104

M. G. Russel, Appendectomy and the risk of developing ulcerative colitis or Crohn's disease: Results of a large case-control study. South Limburg Inflammatory Bowel Disease Study Group, Gastroenterology, vol.113, issue.2, pp.377-82, 1997.
DOI : 10.1053/gast.1997.v113.pm9247453

G. Pineton-de-chambrun, Pathogenic agents in inflammatory bowel diseases, Current Opinion in Gastroenterology, vol.24, issue.4, pp.440-447, 2008.
DOI : 10.1097/MOG.0b013e3283023be5

R. B. Sartor, Does Mycobacterium avium subspecies paratuberculosis cause Crohn's disease? Gut, pp.896-904, 2005.
DOI : 10.1136/gut.2004.055889

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1774604

I. Abubakar, A Case-Control Study of Drinking Water and Dairy Products in Crohn's Disease--Further Investigation of the Possible Role of Mycobacterium avium paratuberculosis, American Journal of Epidemiology, vol.165, issue.7, pp.165-776, 2007.
DOI : 10.1093/aje/kwk067

A. Patel, N. Shah, and N. , Mycobacterium avium subsp paratuberculosis???Incidences in milk and milk products, their isolation, enumeration, characterization, and role in human health, Journal of Microbiology, Immunology and Infection, vol.44, issue.6, pp.473-482, 2007.
DOI : 10.1016/j.jmii.2011.04.009

C. Abraham and J. H. Cho, Bugging of the Intestinal Mucosa, New England Journal of Medicine, vol.357, issue.7, pp.708-718, 2007.
DOI : 10.1056/NEJMcibr073420

N. A. Koloski, L. Bret, and G. Radford-smith, Hygiene hypothesis in inflammatory bowel disease: A critical review of the literature, World Journal of Gastroenterology, vol.14, issue.2, pp.165-73, 2008.
DOI : 10.3748/wjg.14.165

F. Scaldaferri and C. Fiocchi, Inflammatory bowel disease: Progress and current concepts of etiopathogenesis, Journal of Digestive Diseases, vol.293, issue.4, pp.171-179, 2007.
DOI : 10.1053/j.gastro.2006.03.054

J. P. Hugot, Crohn's disease: the cold chain hypothesis, The Lancet, vol.362, issue.9400, pp.2012-2017, 2003.
DOI : 10.1016/S0140-6736(03)15024-6

A. Forbes, T. Kalantzis-malekzadeh, and F. , Crohn's disease: the cold chain hypothesis, International Journal of Colorectal Disease, vol.2, issue.5, pp.399-401, 2006.
DOI : 10.1007/s00384-005-0003-7

M. A. Mcguckin, Intestinal barrier dysfunction in inflammatory bowel diseases, Inflammatory Bowel Diseases, vol.15, issue.1, pp.100-113, 2009.
DOI : 10.1002/ibd.20539

G. Roda, Intestinal epithelial cells in inflammatory bowel diseases, World Journal of Gastroenterology, vol.16, issue.34, pp.4264-71
DOI : 10.3748/wjg.v16.i34.4264

A. Swidsinski, Viscosity gradient within the mucus layer determines the mucosal barrier function and the spatial organization of the intestinal microbiota, Inflammatory Bowel Diseases, vol.13, issue.8, pp.13-963, 2007.
DOI : 10.1002/ibd.20163

K. J. Maloy and F. Powrie, Intestinal homeostasis and its breakdown in inflammatory bowel disease The role of the immune system in governing hostmicrobe interactions in the intestine, Nature. Nat Immunol, vol.474, issue.73517, pp.298-306, 2013.

M. Sasaki, J. M. Klapproth-tannock, and G. W. , The role of bacteria in the pathogenesis of ulcerative colitis 2012: p. 704953. 43 The bowel microbiota and inflammatory bowel diseases Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach, J Signal Transduct Int J Inflam. Gut, issue.2, pp.55-205, 2006.

A. Swidsinski, Mucosal flora in inflammatory bowel disease High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients, Gastroenterology BMC Microbiol Proc Natl Acad Sci, vol.122, issue.143, pp.44-54, 2002.

S. Kang, Dysbiosis of fecal microbiota in Crohn??s disease patients as revealed by a custom phylogenetic microarray, Inflammatory Bowel Diseases, vol.16, issue.12, pp.2034-2076, 1996.
DOI : 10.1002/ibd.21319

T. Jess, Risk of Intestinal Cancer in Inflammatory Bowel Disease: A Population-Based Study From Olmsted County, Minnesota, Gastroenterology, vol.130, issue.4, pp.1039-1085, 2006.
DOI : 10.1053/j.gastro.2005.12.037

N. N. Andersen and T. Jess, Has the risk of colorectal cancer in inflammatory bowel disease decreased?, World Journal of Gastroenterology, vol.19, issue.43, pp.7561-7569, 2013.
DOI : 10.3748/wjg.v19.i43.7561

T. Jess, Increased Risk of Intestinal Cancer in Crohn's Disease: A Meta-Analysis of Population-Based Cohort Studies, The American Journal of Gastroenterology, vol.107, issue.2, pp.2724-2733, 2005.
DOI : 10.1111/j.1572-0241.2003.08671.x

T. Jess, C. Rungoe, and L. Peyrin-biroulet, Risk of Colorectal Cancer in Patients With Ulcerative Colitis: A Meta-analysis of Population-Based Cohort Studies, Clinical Gastroenterology and Hepatology, vol.10, issue.6, pp.639-684, 2012.
DOI : 10.1016/j.cgh.2012.01.010

M. Lukas, Inflammatory Bowel Disease as a Risk Factor for Colorectal Cancer, Digestive Diseases, vol.28, issue.4-5, pp.619-643
DOI : 10.1159/000320276

A. Rizzo, Intestinal inflammation and colorectal cancer: A double-edged sword?, World J Gastroenterol, vol.17, issue.26, pp.3092-100

J. Xie and S. H. Itzkowitz, Cancer in inflammatory bowel disease, World Journal of Gastroenterology, vol.14, issue.3, pp.378-89, 2008.
DOI : 10.3748/wjg.14.378

T. A. Ullman, S. H. Itzkowitz-nieminen, and U. , Intestinal inflammation and cancer Inflammation and disease duration have a cumulative effect on the risk of dysplasia and carcinoma in IBD: A case-control observational study based on registry data Age at diagnosis of inflammatory bowel disease influences early development of colorectal cancer in inflammatory bowel disease patients: a nationwide, long-term survey, Gastroenterology. Int J Cancer J Gastroenterol, vol.140, issue.5812, pp.1807-1823, 2012.

J. A. Eaden, K. R. Abrams, and J. F. Mayberry, The risk of colorectal cancer in ulcerative colitis: a meta-analysis, Gut, vol.48, issue.4, pp.526-561, 2001.
DOI : 10.1136/gut.48.4.526

G. Multhoff, M. Molls, J. Radons, and J. , Chronic Inflammation in Cancer Development, Frontiers in Immunology, vol.2, issue.6, pp.2101-2114
DOI : 10.3389/fimmu.2011.00098

T. A. Ullman and S. H. Itzkowitz, Intestinal Inflammation and Cancer, Gastroenterology, vol.140, issue.6, pp.1807-1823, 2011.
DOI : 10.1053/j.gastro.2011.01.057

J. P. Zackular, The Gut Microbiome Modulates Colon Tumorigenesis, mBio, vol.4, issue.6, pp.692-705, 2013.
DOI : 10.1128/mBio.00692-13

A. C. Goodwin, Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis Enterotoxigenic Bacteroides fragilis: a rogue among symbiotes, Proc Natl Acad Sci U S A Clin Microbiol Rev, vol.108, issue.222, pp.349-69, 2009.

M. Gueimonde, Qualitative and quantitative analyses of the bifidobacterial microbiota in the colonic mucosa of patients with colorectal cancer, diverticulitis and inflammatory bowel disease, World Journal of Gastroenterology, vol.13, issue.29, pp.13-3985, 2007.
DOI : 10.3748/wjg.v13.i29.3985

J. M. Uronis, Modulation of the intestinal microbiota alters colitis-associated colorectal cancer susceptibility The microbiome and cancer, PLoS One Nat Rev Cancer, issue.4611, pp.13-800, 2009.

K. J. Rhee, Induction of Persistent Colitis by a Human Commensal, Enterotoxigenic Bacteroides fragilis, in Wild-Type C57BL/6 Mice, Infection and Immunity, vol.77, issue.4, pp.1708-1726, 2009.
DOI : 10.1128/IAI.00814-08

S. Wu, A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses, Nature Medicine, vol.124, issue.9, pp.1016-1038, 2009.
DOI : 10.1038/nm.2015

T. Jess, Decreasing risk of colorectal cancer in patients with inflammatory bowel disease over 30 years e1 Effect of 5-aminosalicylate use on colorectal cancer and dysplasia risk: a systematic review and metaanalysis of observational studies, Gastroenterology. Am J Gastroenterol, vol.143, issue.26, pp.375-81, 2005.

C. Stolfi, F. Pallone, G. Monteleone-baars, and J. E. , Colorectal cancer chemoprevention by mesalazine and its derivatives 2012: p. 980458. 76 The risk of inflammatory bowel disease-related colorectal carcinoma is limited: results from a nationwide nested case-control study Thiopurines prevent advanced colorectal neoplasia in patients with inflammatory bowel disease, J Biomed Biotechnol Am J Gastroenterol. Gut, vol.106, issue.22, pp.61-235

M. D. Rutter, Thirty-Year Analysis of a Colonoscopic Surveillance Program for Neoplasia in Ulcerative Colitis, Gastroenterology, vol.130, issue.4, pp.1030-1038, 2006.
DOI : 10.1053/j.gastro.2005.12.035

J. K. Triantafillidis, E. Merikas, and F. Georgopoulos, Current and emerging drugs for the treatment of inflammatory bowel disease Treatment of inflammatory bowel disease (IBD), Drug Des Devel Ther Pharmacol Rep, issue.53, pp.185-210, 2011.

D. Burger and S. Travis, Conventional Medical Management of Inflammatory Bowel Disease, Gastroenterology, vol.140, issue.6, pp.1827-1837
DOI : 10.1053/j.gastro.2011.02.045

I. Curkovic, M. Egbring, and G. A. , Kullak-Ublick, Risks of inflammatory bowel disease treatment with glucocorticosteroids and aminosalicylates, Dig Dis, pp.31-34, 2013.

W. J. Sandborn, Azathioprine: State of the Art in Inflammatory Bowel Disease, Scandinavian Journal of Gastroenterology, vol.91, issue.8097, pp.92-101, 1998.
DOI : 10.1136/gut.35.3.360

C. A. Siegel, Risk of Lymphoma Associated With Combination Anti???Tumor Necrosis Factor and Immunomodulator Therapy for the Treatment of Crohn's Disease: A Meta-Analysis, Clinical Gastroenterology and Hepatology, vol.7, issue.8, pp.874-81, 2009.
DOI : 10.1016/j.cgh.2009.01.004

L. Beaugerie, Lymphoproliferative disorders in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study, The Lancet, vol.374, issue.9701, pp.374-1617, 2009.
DOI : 10.1016/S0140-6736(09)61302-7

URL : https://hal.archives-ouvertes.fr/hal-00466312

A. Lopez, Increased Risk of Acute Myeloid Leukemias and Myelodysplastic Syndromes in Patients Who Received Thiopurine Treatment for Inflammatory Bowel Disease Increased risk for nonmelanoma skin cancers in patients who receive thiopurines for inflammatory bowel disease, Clin Gastroenterol Hepatol. Gastroenterology, vol.87, issue.1415, pp.1621-1649

Y. Mazor, Risk Factors for Serious Adverse Effects of Thiopurines in Patients with Crohn&#8217;s Disease, Current Drug Safety, vol.8, issue.3, pp.181-186
DOI : 10.2174/15748863113089990033

A. A. Alfadhli, J. W. Mcdonald, B. G. Feagan-talley, and N. J. , Methotrexate for induction of remission in refractory Crohn's disease): p. CD003459. 90 An evidence-based systematic review on medical therapies for inflammatory bowel disease Use of biological molecules in the treatment of inflammatory bowel disease, Cochrane Database Syst Rev Am J Gastroenterol. J Intern Med, vol.106, issue.2701, pp.15-28, 2005.

G. R. Lichtenstein, Serious Infection and Mortality in Patients With Crohn's Disease: More Than 5 Years of Follow-Up in the TREAT??? Registry, The American Journal of Gastroenterology, vol.31, issue.9, pp.1409-1431
DOI : 10.3899/jrheum.100347

S. B. Hanauer, Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial, The Lancet, vol.359, issue.9317, pp.359-1541, 2002.
DOI : 10.1016/S0140-6736(02)08512-4

H. Yanai and S. B. Hanauer, Assessing Response and Loss of Response to Biological Therapies in IBD, The American Journal of Gastroenterology, vol.102, issue.4, pp.685-98
DOI : 10.3109/00365521.2010.510572

T. Yamamoto and T. Watanabe, Surgery for luminal Crohn???s disease, World Journal of Gastroenterology, vol.20, issue.1, pp.78-90, 2014.
DOI : 10.3748/wjg.v20.i1.78

L. Maggiori and Y. Panis, Surgical management of IBD???from an open to a laparoscopic approach, Nature Reviews Gastroenterology & Hepatology, vol.240, issue.5, pp.297-306, 2013.
DOI : 10.1007/s00464-011-2117-z

C. Charpentier, Natural history of elderly-onset inflammatory bowel disease: a population-based cohort study, Gut, vol.139, issue.Suppl 2, pp.423-455
DOI : 10.1136/gutjnl-2012-303864

J. F. Colombel, Adalimumab for Maintenance of Clinical Response and Remission in Patients With Crohn???s Disease: The CHARM Trial, Gastroenterology, vol.132, issue.1, pp.52-65, 2007.
DOI : 10.1053/j.gastro.2006.11.041

G. Pineton-de-chambrun, Clinical implications of mucosal healing for the management of IBD, Nature Reviews Gastroenterology & Hepatology, vol.50, issue.1, pp.15-29
DOI : 10.1038/nrgastro.2009.203

M. Dave, E. V. Loftus, and J. , Mucosal healing in inflammatory bowel disease-a true paradigm of success?, Gastroenterol Hepatol, vol.8, issue.1, pp.29-38

L. Pastorelli, Central Role of the Gut Epithelial Barrier in the Pathogenesis of Chronic Intestinal Inflammation: Lessons Learned from Animal Models and Human Genetics Animal models of inflammatory bowel disease, J Pharmacol Toxicol Methods, issue.2, pp.50-81, 2004.

C. O. Elson and Y. Cong, Understanding Immune-Microbial Homeostasis in Intestine, Immunologic Research, vol.26, issue.1-3, pp.87-94, 2002.
DOI : 10.1385/IR:26:1-3:087

S. Wirtz and M. F. Neurath, Mouse models of inflammatory bowel disease???, Advanced Drug Delivery Reviews, vol.59, issue.11, pp.1073-83, 2007.
DOI : 10.1016/j.addr.2007.07.003

S. Wirtz, Chemically induced mouse models of intestinal inflammation, Nature Protocols, vol.47, issue.3, pp.541-547, 2007.
DOI : 10.1038/nprot.2007.41

D. Tardieu, J. J. Petit, and C. R. , Le modèle animal d'inflammation au Dextran Soduim Sulfate : études des mécanismes d'action proinflammatoire et génotoxique chez le rat, Revue Méd. Vét, issue.11, pp.152-811, 2001.

S. Wirtz and M. F. Neurath, Animal models of intestinal inflammation: new insights into the molecular pathogenesis and immunotherapy of inflammatory bowel disease, International Journal of Colorectal Disease, vol.15, issue.3, pp.144-60, 2000.
DOI : 10.1007/s003840000227

D. Andrea and A. , IL-10) inhibits human lymphocyte interferon gamma-production by suppressing natural killer cell stimulatory factor/IL-12 synthesis in accessory cells, Interleukin J Exp Med, vol.10, issue.1783, pp.1041-1049, 1993.

R. De-waal-malefyt, Interleukin 10 (IL-10) and viral IL-10 strongly reduce antigen-specific human T cell proliferation by diminishing the antigen-presenting capacity of monocytes via downregulation of class II major histocompatibility complex expression, Journal of Experimental Medicine, vol.174, issue.4, pp.915-939, 1991.
DOI : 10.1084/jem.174.4.915

F. Koch, High level IL-12 production by murine dendritic cells: upregulation via MHC class II and CD40 molecules and downregulation by IL-4 and IL-10 [published erratum appears in J Exp Med 1996 Oct 1;184(4):following 1590], Journal of Experimental Medicine, vol.184, issue.2, pp.741-747, 1996.
DOI : 10.1084/jem.184.2.741

T. B. Fitzpatrick and M. A. Pathak, Historical aspects of methoxsalen and other furocoumarins, J Invest Dermatol 112. O. Raab, Z., Biol. A.J., Muench. Med. Wochenschr, vol.32, issue.50, pp.229-260, 1900.

T. J. Dougherty, Photoradiation therapy for the treatment of malignant tumors, Cancer Res, vol.38, issue.8, pp.2628-2663, 1978.

T. J. Dougherty, A brief history of clinical photodynamic therapy development at Roswell Park Cancer Institute, J Clin Laser Med Surg, vol.14, issue.5, pp.219-240, 1996.

P. Agostinis, Photodynamic therapy of cancer: An update, CA: A Cancer Journal for Clinicians, vol.4, issue.1, pp.250-81, 2011.
DOI : 10.3322/caac.20114

C. Hopper, C. Niziol, and M. Sidhu, The cost-effectiveness of Foscan mediated photodynamic therapy (Foscan-PDT) compared with extensive palliative surgery and palliative chemotherapy for patients with advanced head and neck cancer in the UK, Oral Oncology, vol.40, issue.4, pp.372-82, 2004.
DOI : 10.1016/j.oraloncology.2003.09.003

B. Pegaz, Photothrombic activity of m-THPC-loaded liposomal formulations: Pre-clinical assessment on chick chorioallantoic membrane model, European Journal of Pharmaceutical Sciences, vol.28, issue.1-2, pp.134-174, 2006.
DOI : 10.1016/j.ejps.2006.01.008

Y. N. Konan, R. Gurny, and E. Allemann, State of the art in the delivery of photosensitizers for photodynamic therapy, Journal of Photochemistry and Photobiology B: Biology, vol.66, issue.2, pp.89-106, 2002.
DOI : 10.1016/S1011-1344(01)00267-6

B. Chen, B. W. Pogue, and T. Hasan, Liposomal delivery of photosensitising agents, Expert Opinion on Drug Delivery, vol.24, issue.3, pp.477-87, 2005.
DOI : 10.1562/0031-8655(2000)072<0057:AUODIP>2.0.CO;2

A. S. Derycke and P. A. De-witte, Liposomes for photodynamic therapy, Advanced Drug Delivery Reviews, vol.56, issue.1, pp.17-30, 2004.
DOI : 10.1016/j.addr.2003.07.014

A. M. Richter, LIPOSOMAL DELIVERY OF A PHOTOSENSITIZER, BENZOPORPHYRIN DERIVATIVE MONOACID RING A (BPD), TO TUMOR TISSUE IN A MOUSE TUMOR MODEL, Photochemistry and Photobiology, vol.46, issue.s1, pp.1000-1006, 1993.
DOI : 10.1016/0006-2952(92)90313-8

S. Bonneau, C. Phanie, and . Vever-bizet, Tetrapyrrole photosensitisers, determinants of subcellular localisation and mechanisms of photodynamic processes in therapeutic approaches, Expert Opinion on Therapeutic Patents, vol.58, issue.9, pp.1011-1025, 2008.
DOI : 10.1039/b108586g

R. Bonnett, -tetra(hydroxyphenyl)porphyrin series as tumour photosensitizers, Biochemical Journal, vol.261, issue.1, pp.277-80, 1989.
DOI : 10.1042/bj2610277

URL : https://hal.archives-ouvertes.fr/hal-01439834

J. Garrier, Compartmental Targeting for mTHPC-Based Photodynamic Treatment In Vivo: Correlation of Efficiency, Pharmacokinetics, and??Regional Distribution of Apoptosis, International Journal of Radiation Oncology*Biology*Physics, vol.78, issue.2, pp.563-71
DOI : 10.1016/j.ijrobp.2010.04.009

URL : https://hal.archives-ouvertes.fr/hal-00479855

M. O. Senge and J. C. Brandt, (m-hydroxyphenyl)chlorin)--a second-generation photosensitizer, Temoporfin (Foscan(R) Photochem Photobiol, vol.51015, issue.876, pp.20-1240, 2011.

H. Mojzisova, Photosensitizing properties of chlorins in solution and in membrane-mimicking systems, Photochemical & Photobiological Sciences, vol.68, issue.6, pp.778-87, 2009.
DOI : 10.1039/b822269j

URL : https://hal.archives-ouvertes.fr/hal-00394568

M. H. Teiten, Primary Photodamage Sites and Mitochondrial Events after Foscan?? Photosensitization of MCF-7 Human Breast Cancer Cells??, Photochemistry and Photobiology, vol.462, issue.1, pp.9-14, 2003.
DOI : 10.1562/0031-8655(2003)078<0009:PPSAME>2.0.CO;2

M. H. Teiten, Endoplasmic reticulum and Golgi apparatus are the preferential sites of Foscan localisation in cultured tumour cells Comparative characterization of the efficiency and cellular pharmacokinetics of Foscan-and Foslip-based photodynamic treatment in human biliary tract cancer cell lines, Br J Cancer Photochem Photobiol Sci, vol.88, issue.130 66, pp.146-52, 2003.

L. Lilge, M. Portnoy, and B. C. Wilson, Apoptosis induced in vivo by photodynamic therapy in normal brain and intracranial tumour tissue, Br J Cancer, issue.8, pp.83-1110, 2000.

P. Mroz, Photodynamic therapy of tumors can lead to development of systemic antigen-specific immune response Photodynamic therapy and anti-tumour immunity, PLoS One. Nat Rev Cancer, vol.5, issue.12 67, pp.535-580, 2006.

K. Pizova, Photodynamic therapy for enhancing antitumour immunity, Biomedical Papers, vol.156, issue.2, pp.93-102, 2012.
DOI : 10.5507/bp.2012.056

S. Coutier, Foscan?? (mTHPC) photosensitized macrophage activation: enhancement of phagocytosis, nitric oxide release and tumour necrosis factor-??-mediated cytolytic activity, British Journal of Cancer, vol.81, issue.1, pp.37-42, 1999.
DOI : 10.1038/sj.bjc.6690648

S. O. Gollnick, Altered expression of interleukin 6 and interleukin 10 as a result of photodynamic therapy in vivo The Interrelation Between a Pro-inflammatory Milieu and Fluorescence Diagnosis or Photodynamic Therapy of Human Skin Cell Lines Photodynamic treatment as a novel approach in the therapy of arthritic joints, Cancer Res Lasers Surg Med, vol.57, issue.404, pp.3904-3913, 1997.

L. G. Ratkay, Amelioration of antigen-induced arthritis in rabbits by induction of apoptosis of inflammatory cells with local application of transdermal photodynamic therapy, Arthritis Rheum, issue.3, pp.41-525, 1998.

S. Leong, Transcutaneous Photodynamic Therapy Alters the Development of an Adoptively Transferred Form of Murine Experimental Autoimmune Encephalomyelitis, Photochemistry and Photobiology, vol.264, issue.5, pp.751-758, 1996.
DOI : 10.1016/1011-1344(90)80008-L

W. H. Boehncke, Photodynamic therapy in psoriasis: suppression of cytokine production in vitro and recording of fluorescence modification during treatment in vivo, Archives of Dermatological Research, vol.36, issue.6, pp.286-300, 1994.
DOI : 10.1007/BF00402219

J. Garrier, Foslip??-based photodynamic therapy as a means to improve wound healing, Photodiagnosis and Photodynamic Therapy, vol.8, issue.4, pp.321-328, 2011.
DOI : 10.1016/j.pdpdt.2011.06.003

URL : https://hal.archives-ouvertes.fr/hal-00598501

R. S. Jayasree, The Influence of Photodynamic Therapy on the Wound Healing Process in Rats, Journal of Biomaterials Applications, vol.15, issue.3, pp.176-86, 2001.
DOI : 10.1106/9335-Q0NC-5XCQ-KBYK

S. Werner and R. Grose, Regulation of wound healing by growth factors and cytokines, Physiol Rev, vol.83, issue.3, pp.835-70, 2003.

T. Dai, Y. Y. Huang, and M. R. Hamblin, Photodynamic therapy for localized infections???State of the art, Photodiagnosis and Photodynamic Therapy, vol.6, issue.3-4, pp.3-4, 2009.
DOI : 10.1016/j.pdpdt.2009.10.008

G. O. Simkin, IL-10 Contributes to the Inhibition of Contact Hypersensitivity in Mice Treated with Photodynamic Therapy, The Journal of Immunology, vol.164, issue.5, pp.2457-62, 2000.
DOI : 10.4049/jimmunol.164.5.2457

L. G. Ratkay, J. D. Waterfield, and D. W. Hunt, Photodynamic Therapy in Immune (Non-Oncological) Disorders, BioDrugs, vol.3914, issue.2, pp.127-162, 1998.
DOI : 10.2165/00063030-200014020-00006

D. J. Granville, J. G. Levy, and D. W. Hunt, Photodynamic therapy induces caspase-3 activation in HL-60 cells, Cell Death and Differentiation, vol.4, issue.7, pp.623-631, 1997.
DOI : 10.1038/sj.cdd.4400286

D. J. Granville, release, activation of caspases 3, 6, 7 and 8 followed by Bap31 cleavage in HeLa cells treated with photodynamic therapy, FEBS Letters, vol.16, issue.1-2, pp.5-10, 1998.
DOI : 10.1016/S0014-5793(98)01193-4

D. J. Granville, J. G. Levy, and D. W. Hunt, Photodynamic Treatment with Benzoporphyrin Derivative Monoacid Ring A Produces Protein Tyrosine Phosphorylation Events and DNA Fragmentation in Murine P815 Cells, Photochemistry and Photobiology, vol.63, issue.3, pp.358-62, 1998.
DOI : 10.1006/excr.1994.1093

D. J. Granville, Fas ligand and TRAIL augment the effect of photodynamic therapy on the induction of apoptosis in JURKAT cells, International Immunopharmacology, vol.1, issue.9-10, pp.9-10, 2001.
DOI : 10.1016/S1567-5769(01)00107-2

H. Jiang, Selective Action of the Photosensitizer QLT0074 on Activated Human T Lymphocytes??, Photochemistry and Photobiology, vol.76, issue.2, pp.224-255, 1999.
DOI : 10.1562/0031-8655(2002)0760224SAOTPQ2.0.CO2

S. Marchal, L. Bezdetnaya, and F. Guillemin, -Based Photodynamic Treatment in Human Colon Adenocarcinoma Cell Line HT29, Biochemistry (Moscow), vol.69, issue.1, pp.45-54, 2004.
DOI : 10.1023/B:BIRY.0000016350.61894.be

URL : https://hal.archives-ouvertes.fr/hal-00458990

S. Marchal, Necrotic and apoptotic features of cell death in response to Foscan?? photosensitization of HT29 monolayer and multicell spheroids, Biochemical Pharmacology, vol.69, issue.8, pp.69-1167, 2005.
DOI : 10.1016/j.bcp.2005.01.021

S. Chiu, Phthalocyanine 4 photodynamic therapy-induced apoptosis of mouse L5178Y-R cells results from a delayed but extensive release of cytochrome c from mitochondria, Cancer Letters, vol.165, issue.1, pp.51-59, 2001.
DOI : 10.1016/S0304-3835(01)00422-0

P. Agostinis, Regulatory pathways in photodynamic therapy induced apoptosis, Photochemical & Photobiological Sciences, vol.3, issue.8, pp.721-730, 2004.
DOI : 10.1039/b315237e

B. A. Allison, P. H. Pritchard, and J. G. Levy, Evidence for low-density lipoprotein receptor-mediated uptake of benzoporphyrin derivative, British Journal of Cancer, vol.69, issue.5, pp.69-833, 1994.
DOI : 10.1038/bjc.1994.162

D. E. King, Photodynamic Alteration of the Surface Receptor Expression Pattern of Murine Splenic Dendritic Cells, Scandinavian Journal of Immunology, vol.49, issue.2, pp.184-92, 1999.
DOI : 10.1046/j.1365-3083.1999.00498.x

M. O. Obochi, L. G. Ratkay, and J. G. Levy, Prolonged skin allograft survival after photodynamic therapy associated with modification of donor skin antigenicity, Transplantation, issue.6, pp.63-810, 1997.

D. Renzo and M. , Extracorporeal photopheresis affects co-stimulatory molecule expression and interleukin-10 production by dendritic cells in graft-versus-host disease patients, Clinical & Experimental Immunology, vol.100, issue.3, pp.407-420, 2008.
DOI : 10.1111/j.1365-2249.2007.03577.x

A. Castano, D. T. Hamblin, M. Evans, and S. , Mechanisms in photodynamic therapy : part two--cellular signaling, cell metabolism and modes of cell death Photodiagnosis Photodynamic Therapy): p. 23. 165 Effect of photodynamic therapy on tumor necrosis factor production by murine macrophages The Interrelation Between a Pro-inflammatory Milieu and Fluorescence Diagnosis or Photodynamic Therapy of Human Skin Cell Lines Immunological suppression in mice treated with hematoporphyrin derivative photoradiation, J Natl Cancer Inst Photodiagnosis Photodyn Ther. Cancer Res, vol.24, issue.46, pp.1608-1619, 1986.

S. O. Gollnick, IL-10 does not play a role in cutaneous Photofrin photodynamic therapy-induced suppression of the contact hypersensitivity response, Photochem Photobiol, issue.6, pp.74-811, 2001.

T. Kato, Induction of IL-12 p40 messenger RNA expression and IL-12 production of macrophages via CD40-CD40 ligand interaction, J Immunol, issue.10, pp.156-3932, 1996.

M. Cella, Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation, Journal of Experimental Medicine, vol.184, issue.2, pp.747-52, 1996.
DOI : 10.1084/jem.184.2.747

A. D. Beischer, Synovial ablation in a rabbit rheumatoid arthritis model using photodynamic therapy, ANZ Journal of Surgery, vol.64, issue.7, pp.72-517, 2002.
DOI : 10.1046/j.1445-2197.2002.02451.x

B. Funke, Transdermal photodynamic therapy--a treatment option for rheumatic destruction of small joints? Lasers Surg Med, pp.866-74, 2006.

E. Torikai, Photodynamic therapy using talaporfin sodium for synovial membrane from rheumatoid arthritis patients and collagen-induced arthritis rats, Clinical Rheumatology, vol.31, issue.Part 1, pp.27-751, 2008.
DOI : 10.1007/s10067-007-0794-8

G. Kirdaite, Protoporphyrin IX photodynamic therapy for synovitis, Arthritis & Rheumatism, vol.68, issue.5, pp.1371-1379, 2002.
DOI : 10.1002/art.10199

D. Gabriel, Thrombin-sensitive dual fluorescence imaging and therapeutic agent for detection and treatment of synovial inflammation in murine rheumatoid arthritis, Journal of Controlled Release, vol.163, issue.2, pp.178-86
DOI : 10.1016/j.jconrel.2012.08.022

L. G. Ratkay, Photodynamic therapy; a comparison with other immunomodulatory treatments of adjuvant-enhanced arthritis in MRL-lpr mice, Clinical & Experimental Immunology, vol.3, issue.Suppl., pp.95-373, 1994.
DOI : 10.1111/j.1365-2249.1994.tb07006.x

F. Schmitt, Chitosan-based nanogels for selective delivery of photosensitizers to macrophages and improved retention in and therapy of articular joints, Journal of Controlled Release, vol.144, issue.2, pp.242-50
DOI : 10.1016/j.jconrel.2010.02.008

M. Tanaka, Photodynamic therapy can induce a protective innate immune response against murine bacterial arthritis via neutrophil accumulation The use of transcutaneous photodynamic therapy in the prevention of adjuvant-enhanced arthritis in MRL/lpr mice, PLoS One Clin Immunol Immunopathol, vol.7, issue.62, pp.39823-179, 1994.

M. O. Obochi, TARGETING ACTIVATED LYMPHOCYTES WITH PHOTODYNAMIC THERAPY: SUSCEPTIBILITY OF MITOGEN-STIMULATED SPLENIC LYMPHOCYTES TO BENZOPORPHYRIN DERIVATIVE (BPD) PHOTOSENSITIZATION, Photochemistry and Photobiology, vol.47, issue.3, pp.169-75, 1995.
DOI : 10.1006/clin.1994.1139

D. Chabannes, D. P. Besnier, and V. L. Esnault, Photopheresis affects the course of experimental allergic encephalomyelitis in Lewis rat, Photodermatology, Photoimmunology and Photomedicine, vol.98, issue.5, pp.238-281, 2002.
DOI : 10.1002/(SICI)1521-4141(200003)30:03<908::AID-IMMU908>3.0.CO;2-R

D. P. Besnier, Extracorporeal photochemotherapy for secondary chronic progressive multiple sclerosis: a pilot study, Photodermatology, Photoimmunology and Photomedicine, vol.18, issue.1, pp.36-41, 2002.
DOI : 10.1034/j.1600-0781.2002.180106.x

D. J. Robinson, Improved response of plaque psoriasis after multiple treatments with topical 5- aminolaevulinic acid photodynamic therapy A placebo-controlled randomized study on the clinical effectiveness, immunohistochemical changes and protoporphyrin IX accumulation in fractionated 5-aminolaevulinic acid-photodynamic therapy in patients with psoriasis, Acta Derm Venereol Br J Dermatol, vol.79, issue.1842, pp.451-456, 1999.

R. Bissonnette, Systemic Photodynamic Therapy with Aminolevulinic Acid Induces Apoptosis in Lesional T Lymphocytes of Psoriatic Plaques, Journal of Investigative Dermatology, vol.119, issue.1, pp.77-83, 2002.
DOI : 10.1046/j.1523-1747.2002.01827.x

J. Fransson and A. M. Ros, Clinical and immunohistochemical evaluation of psoriatic plaques treated with topical 5-aminolaevulinic acid photodynamic therapy, Photodermatology, Photoimmunology and Photomedicine, vol.233, issue.6, pp.326-358, 2005.
DOI : 10.1111/1523-1747.ep12349946

Y. K. Tandon, M. F. Yang, and E. D. Baron, Role of photodynamic therapy in psoriasis: a brief review, Photodermatology, Photoimmunology & Photomedicine, vol.224, issue.5, pp.222-252, 2008.
DOI : 10.1111/j.1600-0781.2008.00376.x

L. Favre, Low dose endoluminal photodynamic therapy improves murine T cell-mediated colitis, Endoscopy, vol.43, issue.07, pp.604-620, 2011.
DOI : 10.1055/s-0030-1256382

N. Komoike, Photodynamic diagnosis of colitis-associated dysplasia in a mouse model after oral administration of 5-aminolevulinic acid Extracorporeal photochemotherapy for the treatment of refractory Crohn's disease Extracorporeal photochemotherapy in patients with steroid-dependent Crohn's disease: a prospective pilot study, In Vivo. Transfus Apher Sci Aliment Pharmacol Ther, vol.27, issue.3729, pp.747-53, 2001.

M. T. Abreu, Extracorporeal photopheresis for the treatment of refractory Crohn's disease: results of an open-label pilot study, Inflamm Bowel Dis, issue.6, pp.15-829, 2009.

S. A. Eming, Interrelation of immunity and tissue repair or regeneration, Seminars in Cell & Developmental Biology, vol.20, issue.5, pp.517-544, 2009.
DOI : 10.1016/j.semcdb.2009.04.009

T. A. Wilgus, S. Roy, and J. C. Mcdaniel, Neutrophils and Wound Repair: Positive Actions and Negative Reactions, Advances in Wound Care, vol.2, issue.7, pp.379-388
DOI : 10.1089/wound.2012.0383

D. M. Simpson and R. Ross, The neutrophilic leukocyte in wound repair a study with antineutrophil serum, J Clin Invest, issue.8, pp.51-2009, 1972.

J. V. Dovi, L. K. He, and L. A. Dipietro, Accelerated wound closure in neutrophil-depleted mice, Journal of Leukocyte Biology, vol.73, issue.4, pp.448-55, 2003.
DOI : 10.1189/jlb.0802406

S. A. Eming, T. Krieg, and J. M. Davidson, Inflammation in Wound Repair: Molecular and Cellular Mechanisms, Journal of Investigative Dermatology, vol.127, issue.3, pp.514-539, 2007.
DOI : 10.1038/sj.jid.5700701

W. Posten, Low-Level Laser Therapy for Wound Healing, Dermatologic Surgery, vol.31, issue.3, pp.31-334, 2005.
DOI : 10.1097/00042728-200503000-00016

G. Hubner, Strong Induction of Activin Expression after Injury Suggests an Important Role of Activin in Wound Repair, Developmental Biology, vol.173, issue.2, pp.490-498, 1996.
DOI : 10.1006/dbio.1996.0042

K. W. Liechty, Fetal wound repair results in scar formation in interleukin-10???deficient mice in a syngeneic murine model of scarless fetal wound repair, Journal of Pediatric Surgery, vol.35, issue.6, pp.866-72, 2000.
DOI : 10.1053/jpsu.2000.6868

T. Stachon, IL-1??, IL-1??, IL-6, and IL-8 secretion of human keratocytes following photodynamic inactivation (PDI) in vitro, Graefe's Archive for Clinical and Experimental Ophthalmology, vol.41, issue.11, pp.2585-90
DOI : 10.1007/s00417-013-2465-6

T. Stachon, KGF, FGFb, VEGF, HGF and TGF??1 secretion of human keratocytes following photodynamic inactivation (PDI) in vitro, Graefe's Archive for Clinical and Experimental Ophthalmology, vol.82, issue.8, pp.1987-93
DOI : 10.1007/s00417-013-2370-z

M. R. Hamblin, Antimicrobial Photodynamic Therapy and Photodynamic Inactivation, or Killing Bugs with Dyes and Light-A Symposium-in-Print, Photochemistry and Photobiology, vol.7, issue.3, pp.496-504, 2012.
DOI : 10.1111/j.1751-1097.2012.01139.x

V. G. Garcia, Effect of photodynamic therapy on the healing of cutaneous third-degree-burn: histological study in rats, Lasers in Medical Science, vol.21, issue.Suppl, pp.221-229, 2010.
DOI : 10.1007/s10103-009-0694-z

J. C. Silva, Evaluation of the use of low level laser and photosensitizer drugs in healing, Lasers in Surgery and Medicine, vol.7, issue.5, pp.451-458, 2004.
DOI : 10.1002/lsm.20062

Z. Lu, Photodynamic therapy with a cationic functionalized fullerene rescues mice from fatal wound infections, Nanomedicine, vol.5, issue.10, pp.1525-1558, 2010.
DOI : 10.2217/nnm.10.98

M. Wainwright, ???Safe??? photoantimicrobials for skin and soft-tissue infections, International Journal of Antimicrobial Agents, vol.36, issue.1, pp.14-22
DOI : 10.1016/j.ijantimicag.2010.03.002

S. K. Sharma, Photodynamic Therapy for Cancer and for Infections: What Is the Difference?, Israel Journal of Chemistry, vol.20, issue.8-9, pp.691-705, 2012.
DOI : 10.1002/ijch.201100062

G. Jori, Photodynamic therapy in the treatment of microbial infections: Basic principles and perspective applications, Lasers in Surgery and Medicine, vol.48, issue.5, pp.468-81, 2006.
DOI : 10.1002/lsm.20361

R. Yin, Light based anti-infectives: ultraviolet C irradiation, photodynamic therapy, blue light, and beyond Antibacterial photodynamic therapy using water-soluble formulations of hypericin or mTHPC is effective in inactivation of Staphylococcus aureus, Curr Opin Pharmacol Photochem Photobiol Sci, vol.13, issue.5 93, pp.731-62, 2010.

S. Kranz, Photodynamic suppression of Enterococcus faecalis using the photosensitizer mTHPC, Lasers in Surgery and Medicine, vol.49, issue.3, pp.241-249, 2011.
DOI : 10.1002/lsm.21046

M. R. Hamblin, Rapid Control of Wound Infections by Targeted Photodynamic Therapy Monitored by In Vivo Bioluminescence Imaging??, Photochemistry and Photobiology, vol.67, issue.1, pp.51-58, 2002.
DOI : 10.1562/0031-8655(2002)075<0051:RCOWIB>2.0.CO;2

M. R. Hamblin, Optical Monitoring and Treatment of Potentially Lethal Wound Infections In Vivo, The Journal of Infectious Diseases, vol.187, issue.11, pp.1717-1742, 2003.
DOI : 10.1086/375244

P. S. Zolfaghari, In vivo killing of Staphylococcus aureus using a light-activated antimicrobial agent Bactericidal effects of toluidine blue-mediated photodynamic action on Vibrio vulnificus, BMC Microbiol Antimicrob Agents Chemother, issue.93, pp.27-216, 2005.

F. Gad, Targeted photodynamic therapy of established soft-tissue infections in mice, Photochemical & Photobiological Sciences, vol.3, issue.5, pp.451-459, 2004.
DOI : 10.1039/b311901g

C. E. Millson, Ex-vivo treatment of gastric Helicobacter infection by photodynamic therapy, Journal of Photochemistry and Photobiology B: Biology, vol.32, issue.1-2, pp.59-65, 1996.
DOI : 10.1016/1011-1344(95)07190-3

C. M. Cassidy, Development of Novel Oral Formulations Prepared via Hot Melt Extrusion for Targeted Delivery of Photosensitizer to the Colon, Photochemistry and Photobiology, vol.52, issue.4, pp.867-76, 2011.
DOI : 10.1111/j.1751-1097.2011.00915.x

M. C. Teichert, Treatment of oral candidiasis with methylene blue-mediated photodynamic therapy in an immunodeficient murine model, Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology, vol.93, issue.2, pp.155-60, 2002.
DOI : 10.1067/moe.2002.120051

G. B. Kharkwal, Photodynamic therapy for infections: Clinical applications, Lasers in Surgery and Medicine, vol.18, issue.Suppl 3, pp.755-67, 2011.
DOI : 10.1002/lsm.21080

C. H. Wilder-smith, Photoeradication of Helicobacter pylori using 5-aminolevulinic acid: preliminary human studies, Lasers Surg Med, issue.1, pp.31-49, 2002.

R. A. Ganz, Helicobacter pylori in patients can be killed by visible light, Lasers in Surgery and Medicine, vol.49, issue.4, pp.260-265, 2005.
DOI : 10.1002/lsm.20161

J. W. Lee, B. J. Kim, and M. N. Kim, folliculitis, Lasers in Surgery and Medicine, vol.18, issue.2, pp.192-198, 2010.
DOI : 10.1002/lsm.20857

F. H. Sakamoto, L. Torezan, and R. R. Anderson, Photodynamic therapy for acne vulgaris: A critical review from basics to clinical practice, Journal of the American Academy of Dermatology, vol.63, issue.2, pp.195-211
DOI : 10.1016/j.jaad.2009.09.057

P. Braham, Antimicrobial Photodynamic Therapy May Promote Periodontal Healing Through Multiple Mechanisms, Journal of Periodontology, vol.80, issue.11, pp.1790-1798, 2009.
DOI : 10.1902/jop.2009.090214

A. Mantovani, Cancer-related inflammation, Nature, vol.342, issue.7203, pp.454-436, 2008.
DOI : 10.1038/nature07205

V. Baud and M. Karin, Is NF-??B a good target for cancer therapy? Hopes and pitfalls, Nature Reviews Drug Discovery, vol.105, issue.1, pp.33-40, 2009.
DOI : 10.1038/nrd2781

L. Liu, An apple oligogalactan prevents against inflammation and carcinogenesis by targeting LPS/TLR4/NF-kappaB pathway in a mouse model of colitis-associated colon cancer, Carcinogenesis, issue.10, pp.31-1822

X. Liu, J. M. Wang, and F. R. , Iridoid glycosides fraction of Folium syringae leaves modulates NF-kappaB signal pathway and intestinal epithelial cells apoptosis in experimental colitis IKKbeta links inflammation and tumorigenesis in a mouse model of colitisassociated cancer, PLoS One. Cell, vol.6, issue.93, pp.118-285, 2004.

A. Couturier-maillard, NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer, Journal of Clinical Investigation, vol.123, issue.2, pp.700-711, 2013.
DOI : 10.1172/JCI62236DS1

URL : https://hal.archives-ouvertes.fr/hal-00944194

D. N. Seril, Oxidative stress and ulcerative colitis-associated carcinogenesis: studies in humans and animal models, Carcinogenesis, vol.24, issue.3, pp.353-62, 2003.
DOI : 10.1093/carcin/24.3.353

Y. Meissner and A. Lamprecht, Alternative drug delivery approaches for the therapy of inflammatory bowel disease Ulbrich, W. and A. Lamprecht, Targeted drug-delivery approaches by nanoparticulate carriers in the therapy of inflammatory diseases, J Pharm Sci J R Soc Interface, vol.97, issue.236 7 1, pp.2878-91, 2008.

I. B. Renes, Alterations in Muc2 biosynthesis and secretion during dextran sulfate sodium-induced colitis, American Journal of Physiology - Gastrointestinal and Liver Physiology, vol.282, issue.2, pp.382-391, 2002.
DOI : 10.1152/ajpgi.00229.2001

R. Marcon, receptors: the role of tight junctions and intestinal homeostasis, British Journal of Pharmacology, vol.45, issue.2, pp.389-402, 2013.
DOI : 10.1111/j.1476-5381.2012.02136.x

L. S. Poritz, Loss of the Tight Junction Protein ZO-1 in Dextran Sulfate Sodium Induced Colitis, Journal of Surgical Research, vol.140, issue.1, pp.12-21, 2007.
DOI : 10.1016/j.jss.2006.07.050

C. Audebert, M. Sophie, F. Guillemin, M. Chamaillard, L. Peyrin-biroulet et al., Photodynamic therapy relieves colitis and prevents colitis-associated carcinogenesis in mice
URL : https://hal.archives-ouvertes.fr/hal-01096478

H. Lassalle, S. Marchal, F. Guillemin, R. A. Bezdetnaya, and L. , Aptamers as Remarkable Diagnostic and Therapeutic Agents in Cancer Treatment, Current Drug Metabolism, vol.13, issue.8, pp.1130-1174, 2012.
DOI : 10.2174/138920012802850038

URL : https://hal.archives-ouvertes.fr/hal-00697547

A. Reinhard, A. Bressenot, J. Chevaux, A. François, L. Peyrin-biroulet et al., Photodynamic therapy suppresses colitis inflammation in DSS murine model

C. Audebert, M. Chamaillard, L. Peyrin-biroulet, and L. Bezdetnaya, Evaluation de la thérapie photodynamique dans le traitement des maladies inflammatoires chroniques de l'intestin et la prévention des cancers colorectaux associés, 2014.

H. Lassalle, R. A. Marchal, S. Guillemin, F. Bezdetnaya, and L. , Blood flow and tumor hypoxia following Foscan ® PDT treatment in a murine model, Congrès ESP, vol.2013, pp.2-6, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00868528

C. Audebert, M. Sophie, F. Guillemin, M. Chamaillard, L. Peyrin-biroulet et al., Evaluation de la thérapie photodynamique dans le traitement des maladies inflammatoires chroniques de l'intestin et la prévention des cancers colorectaux associés