. .. Sommaire,

I. A. Généralités,

I. C. Épidémiologie,

I. D. Étiologie and .. .. ,

. Ii and . Traitement,

, 3. La réaction du greffon contre l'hôte

, 1. Etape 1 : Activation des cellules de l'hôte, vol.2

. Iv, La réaction chronique du greffon contre l'hôte

V. .. La-barrière-intestinale,

V. B. ,

V. ,

V. B. ,

V. .. Toxicité,

. .. Vii.-atteinte-de-la-barrière-intestinale-dans-la-gvh-aiguë,

. Viii and . .. Renforcement-de-la-barrière-intestinale,

. B. Viii and .. .. Renforcement, Consequences of induction chemotherapy on intestinal barrier and microbiota in acute myeloid leukemia: a translational study Running Title: Intestinal barrier, microbiota and chemotherapy

T. Hueso, Marie Joncquel Chevallier-Curt, vol.2, issue.2

A. Inra, U. Paris-saclay-;-lee, J. Kim, H. Joo, and Y. , Prospective Randomized Comparison of Idarubicin and High-Dose Daunorubicin in Induction Chemotherapy for Newly Diagnosed Acute Myeloid Leukemia, Hôpital Cochin -HUPC, vol.1, issue.24, pp.2754-2763, 2017.

A. Kolonen, M. Sinisalo, and R. Huttunen, Bloodstream infections in acute myeloid leukemia patients treated according to the Finnish Leukemia Group AML-2003 protocol -a prospective nationwide study, Infect Dis (Auckl), vol.49, pp.799-808, 2017.

J. R. Conn, E. M. Catchpoole, N. Runnegar, S. J. Mapp, and K. A. Markey, Low rates of antibiotic resistance and infectious mortality in a cohort of high-risk hematology patients: A single center, retrospective analysis of blood stream infection, PLoS One, vol.12, issue.5, pp.1-13, 2017.

C. Ubeda, Y. Taur, and R. R. Jenq, Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans, J Clin Invest, vol.120, issue.12, pp.4332-4341, 2010.

L. Wrzosek, S. Miquel, and M. L. Noordine, Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent
URL : https://hal.archives-ouvertes.fr/hal-01001072

, BMC Biol, vol.11, p.61, 2013.

B. O. Schroeder, Fight them or feed them: How the intestinal mucus layer manages the gut microbiota, Gastroenterol Rep, vol.7, issue.1, pp.3-12, 2019.

T. Allain, C. B. Amat, J. P. Motta, A. Manko, and A. G. Buret, Interactions of Giardia sp. with the intestinal barrier: Epithelium, mucus, and microbiota. Tissue Barriers, vol.5, pp.1-16, 2017.

V. Gouyer, F. Gottrand, and J. L. Desseyn, The extraordinarily complex but highly structured organization of intestinal mucus-gel unveiled in multicolor images, PLoS One, vol.6, issue.4, pp.13-16, 2011.

M. Johansson, H. Sjövall, and G. C. Hansson, The gastrointestinal mucus system in health and disease, Nat Rev Gastroenterol Hepatol, vol.10, issue.6, pp.352-361, 2013.

V. Gouyer, L. Dubuquoy, and C. Robbe-masselot, Delivery of a mucin domain enriched in cysteine residues strengthens the intestinal mucous barrier, Pediatr Blood Cancer, vol.5, pp.1188-1194, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02380839

T. Hueso, J. Gauthier, J. Chevalier-curt, and M. , Association Between Low Plasma Level of Citrulline Before Allogeneic Hematopoietic Cell Transplantation and Severe Gastrointestinal Graft vs Host Disease, Clin Gastroenterol Hepatol, vol.16, issue.6, pp.908-917, 2018.

T. Suzuki, S. Yoshida, and H. Hara, Physiological concentrations of short-chain fatty acids immediately suppress colonic epithelial permeability, Br J Nutr, vol.100, issue.2, pp.297-305, 2008.

A. Lan, A. Bruneau, and M. Bensaada, Increased induction of apoptosis by Propionibacterium freudenreichii TL133 in colonic mucosal crypts of human microbiota-associated rats treated with 1,2-dimethylhydrazine, Br J Nutr, vol.100, issue.6, pp.1251-1259, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01454096

J. Zuber, I. Radtke, and T. S. Pardee, Mouse models of human AML accurately predict chemotherapy response, Genes Dev, vol.23, issue.7, pp.877-889, 2009.

M. Wunderlich, B. Mizukawa, and F. Chou, Correcting for Microbial Blooms in Fecal Samples during Room-Temperature Shipping, mSystems, vol.2, issue.2, pp.199-215, 2017.

C. Camacho, G. Coulouris, and V. Avagyan, BLAST + : architecture and applications

, BMC Bioinformatics, vol.9, issue.12, pp.1-9, 2009.

C. Quast, E. Pruesse, and P. Yilmaz, The SILVA ribosomal RNA gene database project: Improved data processing and web-based tools, Nucleic Acids Res, vol.41, pp.590-596, 2013.

K. Katoh and D. M. Standley, MAFFT Multiple Sequence Alignment Software Version 7 : Improvements in Performance and Usability, Mol Biol Evol, vol.30, issue.4, pp.772-780, 2013.

M. N. Price, P. S. Dehal, and A. P. Arkin, FastTree 2 -Approximately Maximum-Likelihood Trees for Large Alignments, PLoS One, vol.5, issue.3, p.9490, 2010.

C. Lozupone and R. Knight, UniFrac : a New Phylogenetic Method for Comparing Microbial Communities, Appl Environ Microbiol, vol.71, issue.12, pp.8228-8235, 2005.

C. A. Lozupone, M. Hamady, S. T. Kelley, and R. Knight, Quantitative and Qualitative Diversity Measures Lead to Different Insights into Factors That Structure Microbial Communities, Appl Environ Microbiol, vol.73, issue.5, pp.1576-1585, 2007.

Y. Vasquez-baeza, A. Gonzalez, L. Smarr, D. Mcdonald, and J. T. Morton, Forum Bringing the Dynamic Microbiome to Life with Animations, Cell Host Microbe, vol.21, issue.1, pp.7-10, 2017.

Y. Shono, M. D. Docampo, and J. U. Peled, Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice, Sci Transl Med, vol.8, issue.339, pp.339-71, 2016.

M. Arumugam, J. Raes, and E. Pelletier, Enterotypes of the human gut microbiome, 1195.
URL : https://hal.archives-ouvertes.fr/cea-00903625

D. Seguy, A. Duhamel, M. Rejeb, and . Ben, Better outcome of patients undergoing enteral tube feeding after myeloablative conditioning for allogeneic stem cell transplantation, Transplantation, vol.94, issue.3, pp.287-294, 2012.

F. Gonzales, B. Bruno, A. Fuentes, and M. , Better early outcome with enteral rather than parenteral nutrition in children undergoing MAC allo-SCT, Clin Nutr, vol.37, issue.6, pp.2113-2121, 2018.

T. Hueso, V. Coiteux, C. Curt, and M. , Citrulline and Monocyte-Derived Macrophage Reactivity before Conditioning Predict Acute Graft-versus-Host Disease, Biol Blood Marrow Transplant, vol.23, issue.6, 2017.

A. Rashidi, R. Shanley, and S. G. Holtan, Pretransplant Serum Citrulline Predicts Acute Graft-versus-Host Disease, Biol Blood Marrow Transplant, vol.24, issue.11, pp.2190-2196, 2018.

J. Desseyn, V. Gouyer, and F. Gottrand, Biological modeling of mucus to modulate mucus barriers, Am J Physiol Gastrointest Liver Physiol, issue.30, pp.225-227, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02176893

D. A. Arber, A. Orazi, and R. Hasserjian, The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia, Blood, vol.127, issue.20, pp.2391-2405, 2016.

M. C. Bene, G. Castoldi, and W. Knapp, Proposals for the immunological classification of acute leukemias, Leukemia, vol.9, issue.10, pp.1783-1786, 1995.

J. M. Bennett, D. Catovsky, and M. Daniel, Proposals for the Classification of the Acute Leukaemias French-American-British (FAB) Co-operative Group, Br J Haematol, vol.33, issue.4, pp.451-458, 1976.

H. Döhner, D. J. Weisdorf, and C. D. Bloomfield, Acute Myeloid Leukemia, N Engl J Med, vol.373, issue.12, pp.1136-1152, 2015.

H. Döhner, E. Estey, and D. Grimwade, Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel, Blood, vol.129, issue.4, pp.424-448, 2017.

N. Mi, K. Tanaka, T. Shintani, T. Takahashi, and N. Kamada, Chromosomal Instability in Acute Myelocytic Leukemia and Myelodysplastic Syndrome Patients among Atomic Bomb Survivors, J Radiat Res, vol.40, issue.2, pp.159-167, 1999.

M. S. Linet, S. Yin, and E. S. Gilbert, A retrospective cohort study of cause-specific mortality and incidence of hematopoietic malignancies in Chinese benzene-exposed workers, Int J Cancer, vol.137, issue.9, pp.2184-2197, 2015.

L. M. Morton, G. M. Dores, and M. A. Tucker, Evolving risk of therapy-related acute myeloid leukemia following cancer chemotherapy among adults in the United States, Blood, vol.121, issue.15, pp.2996-3004, 1975.

B. Deschler and M. Lübbert, Acute myeloid leukemia: Epidemiology and etiology, Cancer, vol.107, issue.9, pp.2099-2107, 2006.

S. Farber, L. K. Diamond, R. D. Mercer, R. F. Sylvester, and J. A. Wolff, Temporary Remission in Acute Leukemia in Children Produced by Folic Acid Antagonist, 4-Aminopteroyl-Glutamic Acid (Aminopterin), N Engl J Med, vol.238, issue.23, pp.787-793, 1948.

M. Boiron, C. Jacquillat, and M. Weil, Daunorubicin in the treatment of acute myelocytic leukaemia, Lancet, vol.1, issue.7590, pp.330-333, 1969.

E. S. Henderson, Treatment of Acute Leukemia, Ann Intern Med, vol.69, issue.3, pp.628-632, 1968.

D. Crowther, C. Bateman, and C. P. Vartan, Combination chemotherapy using lasparaginase, daunorubicin, and cytosine arabinoside in adults with acute myelogenous leukaemia, Br Med J, vol.4, issue.5734, pp.513-517, 1970.

J. W. Yates, H. J. Wallace, R. R. Ellison, and J. F. Holland, Cytosine arabinoside (NSC 63878) and daunorubicin (NSC 83142) therapy in acute non lymphocytic leukemia, Cancer Chemother Rep, vol.57, issue.4, pp.485-488, 1973.

T. Murphy and K. Yee, Cytarabine and daunorubicin for the treatment of acute myeloid leukemia, Expert Opin Pharmacother, vol.18, issue.16, pp.1765-1780, 2017.

J. Lee, H. Kim, and Y. Joo, Prospective Randomized Comparison of Idarubicin and High-Dose Daunorubicin in Induction Chemotherapy for Newly Diagnosed Acute Myeloid Leukemia, J Clin Oncol, vol.35, issue.24, pp.2754-2763, 2017.

R. K. Hills, S. Castaigne, and F. R. Appelbaum, Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: A metaanalysis of individual patient data from randomised controlled trials, Lancet Oncol, vol.15, issue.9, pp.986-996, 2014.

C. Röllig, H. Serve, and A. Hüttmann, Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): A multicentre, phase 2, randomised controlled trial, Lancet Oncol, vol.16, issue.16, pp.1691-1699, 2015.

R. M. Stone, S. J. Mandrekar, and B. L. Sanford, Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 Mutation, N Engl J Med, vol.377, issue.5, pp.454-464, 2017.

J. E. Lancet, G. L. Uy, and J. E. Cortes, Cpx-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients with Newly Diagnosed Secondary Acute Myeloid Leukemia, J Clin Oncol, vol.36, issue.26, pp.2684-2692, 2018.

F. W. Gunz, B. I. Bach, P. E. Crossen, J. E. Mellor, S. Singh et al., Relevance of the cytogenetic status in acute leukemia in adults, J Natl Cancer Inst, vol.50, issue.1, pp.55-61, 1973.

P. C. Vincent, R. Sutherland, M. Bradley, D. Lind, and F. W. Gunz, Marrow Culture Studies in Adult Acute Leukemia at Présentation and During Remission, Blood, vol.49, issue.6, pp.903-912, 1977.

A. M. Mauer, New Direction in The Treatment of Acute Lymphoblastic Leukemia in Children, N Engl J Med, vol.315, issue.5, pp.316-317, 1986.

R. P. Gale, Advances in The Treamtent of Acute Myelogenous Leukemia, N Engl J Med, vol.300, issue.21, pp.1189-1199, 2009.

P. A. Cassileth, C. B. Begg, B. Mark, K. S. Benham, C. Weiler et al., A Randomized Study of the Efficacy of Consolidation Therapy in Adult Acute Nonlymphocytic Leukemia, Blood, vol.63, issue.4, pp.843-847, 1984.

G. Tricot, M. A. Boogaerts, R. Vlietinck, M. P. Emonds, and R. L. Verwilghen, The role of intensive remission induction and consolidation therapy in patients with acute myeloid leukaemia, Br J Haematol, vol.66, issue.1, pp.37-44, 1987.

S. N. Wolff, R. H. Herzig, and J. W. Fay, High-dose cytarabine and daunorubicin as consolidation therapy for acute myeloid leukemia in first remission: Long-term followup and results, J Clin Oncol, vol.7, issue.9, pp.1260-1267, 1989.

B. Löwenberg, Sense and nonsense of high-dose cytarabine for acute myeloid leukemia, Blood, vol.121, issue.1, pp.26-28, 2013.

E. D. Thomas, H. L. Lochte, J. H. Cannon, O. D. Sahler, and J. W. Ferrebee, Supralethal whole body irradiation and isologous marrow transplantation in man, J Clin Invest, vol.38, issue.10, pp.1709-1716, 1959.

E. D. Thomas, J. A. Collins, E. C. Herman, and J. W. Ferrebee, Marrow transplants in lethally irradiated dogs given methotrexate, Blood, vol.19, pp.217-228, 1962.

G. Mathé, J. L. Amiel, and J. Niemetz, Greffes de moelle osseuse apres irradiation totale chez des souris leucemiques suivies de l' administration d 'un produit antimitotique, Bull World Health Organ, vol.26, issue.5, pp.692-695, 1962.

J. R. Passweg, H. Baldomero, and P. Bader, Hematopoietic stem cell transplantation in Europe 2014: More than 40 000 transplants annually, Bone Marrow Transplant, vol.51, issue.6, pp.786-792, 2016.

W. D. Shlomchik, Graft versus host diseases, Nat Rev Immunol, vol.7, pp.340-352, 2007.

J. Klein and A. Sato, The HLA System. First of Two Parts, N Engl J Med, vol.343, issue.10, pp.702-709, 2000.

I. Yakoub-agha, F. Mesnil, and M. Kuentz, Allogeneic marrow stem-cell transplantation from human leukocyte antigen-identical siblings versus human leukocyte antigen-allelic-matched unrelated donors (10/10) in patients with standardrisk hematologic malignancy: A prospective study from the French, J Clin Oncol, vol.24, issue.36, pp.5695-5702, 2006.

W. Saber, S. Opie, J. D. Rizzo, M. J. Zhang, M. M. Horowitz et al., Outcomes after matched unrelated donor versus identical sibling hematopoietic cell transplantation in adults with acute myelogenous leukemia, Blood, vol.119, issue.17, pp.3908-3916, 2012.

F. R. Appelbaum, Alternative donor transplantation for adults with acute leukemia, Best Pract Res Clin Haematol, vol.27, issue.3-4, pp.272-277, 2014.

L. Luznik, P. V. O'donnell, and H. J. Symons, HLA-Haploidentical Bone Marrow Transplantation for Hematologic Malignancies Using Nonmyeloablative Conditioning and High-Dose, Posttransplantation Cyclophosphamide, Biol Blood Marrow Transplant, vol.14, issue.6, pp.641-650, 2008.

S. O. Ciurea, M. J. Zhang, and A. A. Bacigalupo, Haploidentical transplant with posttransplant cyclophosphamide vs matched unrelated donor transplant for acute myeloid leukemia, Blood, vol.126, issue.8, pp.1033-1040, 2015.

A. Ruggeri, M. Labopin, and G. Sanz, Comparison of outcomes after unrelated cord blood and unmanipulated haploidentical stem cell transplantation in adults with acute leukemia, Leukemia, vol.29, issue.9, pp.1891-1900, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02007180

C. Anasetti, B. R. Logan, and S. J. Lee, Peripheral-blood stem cells versus bone marrow from unrelated donors, N Engl J Med, vol.367, issue.16, pp.1487-1496, 2012.

A. Bacigalupo, K. Ballen, and D. Rizzo, Defining the Intensity of Conditioning Regimens: Working Definitions, Biol Blood Marrow Transplant, vol.15, issue.12, pp.1628-1633, 2009.

M. L. Sorror, M. B. Maris, and R. Storb, Hematopoietic cell transplantation ( HCT )-specific comorbidity index : a new tool for risk assessment before allogeneic HCT, Blood, vol.106, issue.8, pp.2912-2919, 2005.

V. T. Ho, C. Revta, and P. G. Richardson, Hepatic veno-occlusive disease after hematopoietic stem cell transplantation: Update on defibrotide and other current investigational therapies, Bone Marrow Transplant, vol.41, issue.3, pp.229-237, 2008.

M. Tomblyn, T. Chiller, and H. Einsele, Guidelines for Preventing Infectious Complications among Hematopoietic Cell Transplantation Recipients: A Global Perspective, Biol Blood Marrow Transplant, vol.15, issue.10, pp.1143-1238, 2009.

G. Cannas, C. Pautas, and E. Raffoux, Infectious complications in adult acute myeloid leukemia: analysis of the Acute Leukemia French Association-9802 prospective multicenter clinical trial, Leuk Lymphoma, vol.53, issue.6, pp.1068-1076, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01016571

J. R. Conn, E. M. Catchpoole, N. Runnegar, S. J. Mapp, and K. A. Markey, Low rates of antibiotic resistance and infectious mortality in a cohort of high-risk hematology patients: A single center, retrospective analysis of blood stream infection, PLoS One, vol.12, issue.5, pp.1-13, 2017.

N. Rorije, M. M. Shea, and G. Satyanarayana, BK virus disease after allogeneic stem cell transplantation: A cohort analysis, Biol Blood Marrow Transplant, vol.20, issue.4, pp.564-570, 2014.

M. Nucci and E. Anaissie, How we treat invasive fungal diseases in patients with acute leukemia, Blood, vol.124, issue.26, pp.3858-3870, 2014.

A. H. Filipovich, D. Weisdorf, and S. Pavletic, National Institutes of Health Consensus Development Project on criteria for clinical trials in chronic graft-versus-host disease: I. diagnosis and staging working group report, Biol Blood Marrow Transplant, vol.11, issue.12, pp.945-956, 2005.

B. D. Przepiorka, T. L. Smith, and J. Folloder, Risk Factors for Acute Graft-Versus-Host Disease After Allogeneic Blood Stem Cell Transplantation, pp.1465-1470, 1999.

M. Flowers, Y. Inamoto, and P. A. Carpenter, Comparative Analysis of Risk Factors for Acute GvHD and for Chronic GvHD disease according to NIH consensus Criteria, Blood, vol.117, issue.11, pp.3214-3219, 2011.

H. Glucksberg, R. Storb, and A. Fefer, Clinical manifestations of graft-versus-host disease in human recipients of marrow from HLA-matched sibling donors, Transplantation, vol.18, issue.4, pp.295-304, 1974.

D. Przepiorka, D. Weisdorf, and P. Martin, Consensus Conference on Acute GVHD Grading, vol.15, pp.825-828, 1994.

I. Yakoub-agha, V. Maunoury, and A. Wacrenier, Impact of Small Bowel Exploration Using Video-Capsule Endoscopy in the Management of Acute Gastrointestinal Graftversus-Host Disease, Transplantation, vol.78, issue.11, pp.1697-1701, 2004.

R. Billingham, The biology of graft-versus-host reactions, Harvey Lect, vol.62, pp.21-78, 1967.

J. H. Antin and J. L. Ferrara, Cytokine dysregulation and acute graft-versus-host disease, Blood, vol.80, issue.12, pp.2964-2968, 1992.

J. L. Ferrara, R. Levy, and N. J. Chao, Pathophysiologic mechanisms of acute graft-vs.-host disease, Biol Blood Marrow Transplant, vol.5, issue.6, pp.347-356, 1999.

G. R. Hill and J. L. Ferrara, The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: rationale for the use of cytokine shields in allogeneic bone marrow transplantation, Blood, vol.95, issue.9, pp.2754-2759, 2000.

J. L. Ferrara, J. E. Levine, P. Reddy, and E. Holler, Graft-versus-host disease, Lancet, vol.373, issue.9674, pp.1550-1561, 2009.

G. R. Hill, J. M. Crawford, K. R. Cooke, Y. S. Brinson, L. Pan et al., Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines, Blood, vol.90, issue.8, pp.3204-3213, 1997.

F. P. Nestel, K. S. Price, T. A. Seemayer, and W. S. Lapp, Macrophage priming and lipopolysaccharide-triggered release of tumor necrosis factor alpha during graftversus-host disease, J Exp Med, vol.175, issue.2, pp.405-413, 1992.

S. Heidegger, M. Van-den-brink, T. Haas, and H. Poeck, The role of pattern-recognition receptors in graft-versus-host disease and graft-versus-leukemia after allogeneic stem cell transplantation, Front Immunol, vol.5, pp.1-7, 2014.

W. D. Shlomchik, M. S. Couzens, and C. Tang, Prevention of Graft Versus Host Disease by Inactivation of Host, Science, vol.285, pp.412-415, 1999.

C. C. Matte, J. Liu, and J. Cormier, Donor APCs are required for maximal GVHD but not for GVL, Nat Med, vol.10, issue.9, pp.987-992, 2004.

C. A. Wysocki, A. Panoskaltsis-mortari, B. R. Blazar, and J. S. Serody, Leukocyte migration and graft-versus-host disease, Blood, vol.105, issue.11, pp.4191-4199, 2005.

D. Kägi, F. Vignaux, and B. Ledermann, Fas and perforin pathways as major mechanisms of T cell-mediated cytotoxicity, Science, vol.265, issue.5171, pp.528-530, 1994.

B. Lowin, M. Hahne, C. Mattmann, and J. Tschopp, Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways, Nature, vol.370, issue.6491, pp.650-652, 1994.

K. R. Cooke, G. R. Hill, and J. M. Crawford, Tumor necrosis factor-alpha production to lipopolysaccharide stimulation by donor cells predicts the severity of experimental acute graft-versus-host disease, J Clin Invest, vol.102, issue.10, pp.1882-1891, 1998.

R. Storb, H. J. Deeg, and J. Whitehead, Methotrexate and Cyclosporine Compared with Cyclosporine Alone for Prophylaxis of Acute Graft versus Host Disease after Marrow Transplantation for Leukemia, N Engl J Med, vol.314, issue.12, pp.729-735, 1986.

V. Ratanatharathorn, R. A. Nash, and D. Przepiorka, Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation, Blood, vol.92, issue.7, pp.2303-2314, 1998.

B. Bolwell, R. Sobecks, and B. Pohlman, A prospective randomized trial comparing cyclosporine and short course methotrexate with cyclosporine and mycophenolate mofetil for GVHD prophylaxis in myeloablative allogeneic bone marrow transplantation, Bone Marrow Transplant, vol.34, issue.7, pp.621-625, 2004.

C. Cutler, S. Li, and V. T. Ho, Extended follow-up of methotrexate-free immunosuppression using sirolimus and tacrolimus in related and unrelated donor peripheral blood stem cell transplantation, Blood, vol.109, issue.7, pp.3108-3114, 2007.

J. Finke, W. A. Bethge, and C. Schmoor, Standard graft-versus-host disease prophylaxis with or without anti-T-cell globulin in haematopoietic cell transplantation from matched unrelated donors: a randomised, open-label, multicentre phase 3 trial, Lancet Oncol, vol.10, issue.9, pp.855-864, 2009.

M. Mohty, M. Labopin, and M. L. Balère, Antithymocyte globulins and chronic graft-vshost disease after myeloablative allogeneic stem cell transplantation from HLAmatched unrelated donors: A report from the Sociéte Française de Greffe de Moelle et de Thérapie Cellulaire, Leukemia, vol.24, issue.11, pp.1867-1874, 2010.

C. J. Lee, B. N. Savani, and M. Mohty, Haploidentical hematopoietic cell transplantation for adult acute myeloid leukemia: A position statement from the acute leukemia working party of the European society for blood and marrow transplantation, Haematologica, vol.102, issue.11, pp.1810-1822, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01633849

S. Z. Pavletic and D. H. Fowler, Are we making progress in GVHD prophylaxis and treatment?, Hematology Am Soc Hematol Educ Program, vol.2012, pp.251-264, 2012.

M. L. Macmillan, D. J. Weisdorf, and J. E. Wagner, Response of 443 patients to steroids as primary therapy for acute graft-versus-host disease: Comparison of grading systems, Biol Blood Marrow Transplant, vol.8, issue.7, pp.387-394, 2002.

J. Bolaños-meade, B. R. Logan, and A. M. Alousi, Phase 3 clinical trial of steroids/mycophenolate mofetil vs steroids/placebo as therapy for acute GVHD: BMT CTN 0802, Blood, vol.124, issue.22, pp.3221-3227, 2014.

A. M. Alousi, D. J. Weisdorf, and B. R. Logan, Etanercept, mycophenolate, denileukin, or pentostatin plus corticosteroids for acute graft-versus-host disease: A randomized phase 2 trial from the Blood and Marrow Transplant Clinical Trials Network, Blood, vol.114, issue.3, pp.511-517, 2009.

R. Zeiser, A. Burchert, and C. Lengerke, Ruxolitinib in corticosteroid-refractory graftversus-host disease after allogeneic stem cell transplantation: A multicenter survey, Leukemia, vol.29, issue.10, pp.2062-2068, 2015.

N. Von-bubnoff, G. Ihorst, and O. Grishina, Ruxolitinib in GvHD (RIG) study: a multicenter, randomized phase 2 trial to determine the response rate of Ruxolitinib and best available treatment (BAT) versus BAT in steroid-refractory acute graftversus-host disease (aGvHD) (NCT02396628), BMC Cancer, vol.18, issue.1, p.1132, 2018.

M. H. Jagasia, H. T. Greinix, and M. Arora, National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group Report, Biol Blood Marrow Transplant, vol.21, issue.3, pp.389-401, 2015.

P. A. Carpenter, C. L. Kitko, and S. Elad, National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: V. The 2014 Ancillary Therapy and Supportive Care Working Group Report, Biol Blood Marrow Transplant, vol.21, issue.7, pp.1167-1187, 2015.

D. Wolff, M. Schleuning, V. Harsdorf, and S. , Consensus conference on clinical practice in chronic GVHD: Second-line treatment of chronic graft-versus-host disease, Biol Blood Marrow Transplant, vol.17, issue.1, pp.1-17, 2011.

L. Magro, J. Gauthier, and M. Richet, Scleral lenses for severe chronic GvHD-related keratoconjunctivitis sicca: A retrospective study by the SFGM-TC, Bone Marrow Transplant, vol.52, issue.6, pp.878-882, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01777907

L. Magro, E. Forcade, and C. Giraud, Management of the chronic graft versus host disease: Guidelines from the Francophone society of bone marrow transplantation and cellular therapies (SFGM-TC), Bull Cancer, vol.104, issue.12, pp.145-168, 2017.

B. N. Savani, M. L. Griffith, S. Jagasia, and S. J. Lee, How I treat late effects in adults after allogeneic stem cell transplantation, Blood, vol.117, issue.11, pp.3002-3009, 2011.

S. C. Nalle and J. R. Turner, Intestinal barrier loss as a critical pathogenic link between inflammatory bowel disease and graft-versus-host disease, Mucosal Immunol, vol.8, pp.1-11, 2015.

J. R. Turner, Intestinal mucosal barrier function in health and disease, Nat Rev Immunol, vol.9, issue.11, pp.799-809, 2009.

R. Williamson, Intestinal Adaptation, N Engl J Med, vol.298, issue.25, pp.1393-1402, 1978.

G. Den-besten, K. Van-eunen, A. K. Groen, K. Venema, D. J. Reijngoud et al., The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism, J Lipid Res, vol.54, issue.9, pp.2325-2340, 2013.

F. Rivera-chávez, L. F. Zhang, and F. Faber, Depletion of Butyrate-Producing Clostridia from the Gut Microbiota Drives an Aerobic Luminal Expansion of Salmonella, Cell Host Microbe, vol.19, issue.4, pp.443-454, 2016.

M. S. Inan, R. J. Rasoulpour, L. Yin, A. K. Hubbard, D. W. Rosenberg et al., The luminal short-chain fatty acid butyrate modulates NF-?B activity in a human colonic epithelial cell line, Gastroenterology, vol.118, issue.4, pp.724-734, 2000.

M. Johansson, J. M. Holmén-larsson, and G. C. Hansson, The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of hostmicrobial interactions, Proc Natl Acad Sci, vol.108, pp.4659-4665, 2011.

N. Asker, M. Axelsson, S. O. Olofsson, and G. C. Hansson, Dimerization of the human MUC2 mucin in the endoplasmic reticulum is followed by a N-glycosylationdependent transfer of the mono-and dimers to the Golgi apparatus, J Biol Chem, vol.273, issue.30, pp.18857-18863, 1998.

M. Johansson, J. K. Gustafsson, and J. Holmen-larsson, Bacteria penetrate the normally impenetrable inner colon mucus layer in both murine colitis models and patients with ulcerative colitis, Gut, vol.63, issue.2, pp.281-291, 2014.

M. Zarepour, K. Bhullar, and M. Montero, The mucin muc2 limits pathogen burdens and epithelial barrier dysfunction during salmonella enterica serovar typhimurium colitis, Infect Immun, vol.81, issue.10, pp.3672-3683, 2013.

M. Johansson, H. Sjövall, and G. C. Hansson, The gastrointestinal mucus system in health and disease, Nat Rev Gastroenterol Hepatol, vol.10, issue.6, pp.352-361, 2013.

P. B. Eckburg, E. M. Bik, and C. N. Bernstein, Microbiology: Diversity of the human intestinal microbial flora, Science, vol.308, issue.5728, pp.1635-1638, 2005.

C. A. Lozupone, J. I. Stombaugh, J. I. Gordon, J. K. Jansson, and R. Knight, Diversity, stability and resilience of the human gut microbiota, Nature, vol.489, issue.7415, pp.220-230, 2012.

X. C. Morgan, N. Segata, and C. Huttenhower, Biodiversity and functional genomics in the human microbiome, Trends Genet, vol.29, issue.1, pp.51-58, 2013.

R. E. Ley, M. Hamady, and C. Lozupone, Evolution of mammals and their gut microbes, Science, vol.320, issue.5883, pp.1647-1651, 2008.

M. Arumugam, J. Raes, and E. Pelletier, Enterotypes of the human gut microbiome, Nature, vol.473, issue.7346, pp.174-180, 2011.
URL : https://hal.archives-ouvertes.fr/cea-00903625

R. B. Sartor, Microbial Influences in Inflammatory Bowel Diseases, Gastroenterology, vol.134, issue.2, pp.577-594, 2008.

J. Walter and R. Ley, The Human Gut Microbiome: Ecology and Recent Evolutionary Changes, Annu Rev Microbiol, vol.65, issue.1, pp.411-429, 2011.

N. Cerf-bensussan and V. Gaboriau-routhiau, The immune system and the gut microbiota: Friends or foes?, Nat Rev Immunol, vol.10, issue.10, pp.735-744, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01204303

S. Westcarr, P. Farshori, J. Wyche, and A. Wa, Apoptosis and differentiation in the crypt-villus unit of the rat small intestine, J Submicrosc Cytol Pathol, vol.31, issue.1, pp.15-30, 1999.

S. Garattini, A. Guaitani, N. E. Palma, and V. , Studies on the Selectivity of Antitumor Agents, Cancer Res, vol.27, pp.1309-1437, 1967.

L. Lubitz and H. Ekert, Reversible Changes in Duodenal Mucosa Associated With Intensive Chemotherapy Followed By Autologous Marrow Rescue, Lancet, vol.314, issue.8141, pp.532-533, 1979.

K. F. Bradstock, J. P. Matthews, and R. M. Lowenthal, A randomized trial of high-versus conventional-dose cytarabine in consolidation chemotherapy for adult de novo acute myeloid leukemia in first remission after induction therapy containing high-dose cytarabine, Blood, vol.105, issue.2, pp.481-488, 2005.

K. F. Bradstock, E. Link, J. Iulio, and . Di, Idarubicin dose escalation during consolidation therapy for adult acute myeloid leukemia, J Clin Oncol, vol.35, issue.15, pp.1678-1685, 2017.

D. M. Keefe, J. Brealey, G. J. Goland, and A. G. Cummins, Chemotherapy for cancer causes apoptosis that precedes hypoplasia in crypts of the small intestine in humans, Gut, vol.47, issue.5, pp.632-637, 2000.

J. M. Bowen, R. J. Gibson, D. M. Keefe, and A. G. Cummins, Cytotoxic chemotherapy upregulates pro-apoptotic Bax and Bak in the small intestine of rats and humans, Pathology, vol.37, issue.1, pp.56-62, 2005.

J. M. Bowen, R. J. Gibson, A. G. Cummins, and D. Keefe, Intestinal mucositis: The role of the Bcl-2 family, p53 and caspases in chemotherapy-induced damage, Support Care Cancer, vol.14, issue.7, pp.713-731, 2006.

S. T. Sonis, Pathobiology of mucositis, Nat Rev Cancer, vol.4, issue.4, pp.277-284, 2004.

L. S. Elting, C. Cooksley, M. Chambers, S. B. Cantor, E. Manzullo et al., The burdens of cancer therapy: Clinical and economic outcomes of chemotherapyinduced mucositis, Cancer, vol.98, issue.7, pp.1531-1539, 2003.

D. Keefe, A. G. Cummins, B. M. Dale, D. Kotasek, T. A. Robb et al., Effect of highdose chemotherapy on intestinal permeability in humans, Clin Sci, vol.92, issue.4, pp.385-389, 1997.

K. D. Fine, S. Ana, C. A. Porter, J. L. Fordtran, and J. S. , Effect of changing intestinal flow rate on a measurement of intestinal permeability, Gastroenterology, vol.108, issue.4, pp.983-989, 1995.

A. L. Buchman, M. E. Ament, D. J. Jenden, and C. Ahn, Choline deficiency is associated with increased risk for venous catheter thrombosis, J Parenter Enter Nutr, vol.30, issue.4, pp.317-320, 2006.

M. Coëffier, S. Claeyssens, and S. Lecleire, Combined enteral infusion of glutamine, carbohydrates, and antioxidants modulates gut protein metabolism in humans, Am J Clin Nutr, vol.88, issue.5, pp.1284-1290, 2008.

M. Crowther, A. Avenell, and D. J. Culligan, Systematic review and meta-analyses of studies of glutamine supplementation in haematopoietic stem cell transplantation, Bone Marrow Transplant, vol.44, issue.7, pp.413-425, 2009.

J. E. Johansson and T. Ekman, Gastro-intestinal toxicity related to bone marrow transplantation: disruption of the intestinal barrier precedes clinical findings, Bone Marrow Transplant, vol.19, issue.9, pp.921-925, 1997.

M. Vancamelbeke and S. Vermeire, The intestinal barrier: a fundamental role in health and disease, Expert Rev Gastroenterol Hepatol, vol.11, issue.9, pp.821-834, 2017.

N. Blijlevens, J. P. Donnelly, D. Pauw, and B. E. , Mucosal barrier injury: Biology, pathology, clinical counterparts and consequences of intensive treatment for haematological malignancy: An overview, Bone Marrow Transplant, vol.25, issue.12, pp.1269-1278, 2000.

E. J. Bow and J. B. Meddings, Intestinal mucosal dysfunction and infection during remissioninduction therapy for acute myeloid leukaemia, Leukemia, vol.20, issue.12, pp.2087-2092, 2006.

P. Crenn, C. Lucas, C. Thuiller, F. Cynober, L. Messing et al., Postabsorptive Plasma Citrulline Concentrations Is a Marker of Absorptive Enterocyte Mass and Intestinal Failure in Humans, Gastroenterology, vol.119, pp.1446-1505, 2000.

L. Lutgens, N. Blijlevens, N. Deutz, J. P. Donnelly, P. Lambin et al., Monitoring myeloablative therapy-induced small bowel toxicity by serum citrulline concentration: A comparison with sugar permeability tests, Cancer, vol.103, issue.1, pp.191-199, 2005.

N. Blijlevens, L. Lutgens, A. Schattenberg, and J. P. Donnelly, Citrulline: a potentially simple quantitative marker of intestinal epithelial damage following myeloablative therapy, Bone Marrow Transplant, vol.34, issue.3, pp.193-196, 2004.

A. H. Herbers, T. Feuth, J. P. Donnelly, and N. M. Blijlevens, Citrulline-based assessment score: First choice for measuring and monitoring intestinal failure after high-dose chemotherapy, Ann Oncol, vol.21, issue.8, pp.1706-1711, 2010.

P. Crenn, N. Neveux, and S. Chevret, Plasma l-citrulline concentrations and its relationship with inflammation at the onset of septic shock: A pilot study, J Crit Care, vol.29, issue.2, pp.315-316, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-00924312

A. Herbers, N. Blijlevens, J. P. Donnelly, and T. De-witte, Bacteraemia coincides with low citrulline concentrations after high-dose melphalan in autologous HSCT recipients, Bone Marrow Transplant, vol.42, issue.5, pp.345-349, 2008.

P. Crenn, B. Messing, and L. Cynober, Citrulline as a biomarker of intestinal failure due to enterocyte mass reduction, Clin Nutr, vol.27, issue.3, pp.328-339, 2008.

S. Takashima, M. Kadowaki, and K. Aoyama, The Wnt agonist R-spondin1 regulates systemic graft-versus-host disease by protecting intestinal stem cells, J Exp Med, vol.208, issue.2, pp.285-294, 2011.

E. Hayase, D. Hashimoto, and K. Nakamura, R-Spondin1 expands Paneth cells and prevents dysbiosis induced by graft-versus-host disease, J Exp Med, vol.214, issue.12, pp.3507-3518, 2017.

A. Joly, A. Deepti, and A. Seignez, The HSP90 inhibitor, 17AAG, protects the intestinal stem cell niche and inhibits graft versus host disease development, Oncogene, vol.35, pp.1-10, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01434737

Y. Eriguchi, S. Takashima, and H. Oka, Graft-versus-host disease disrupts intestinal microbial ecology by inhibiting Paneth cell production of ?-defensins, Blood, vol.120, issue.1, pp.223-231, 2012.

A. M. Hanash, J. A. Dudakov, and G. Hua, Interleukin-22 Protects Intestinal Stem Cells from Immune-Mediated Tissue Damage and Regulates Sensitivity to Graft versus Host Disease, Immunity, vol.37, issue.2, pp.339-350, 2012.

M. Couturier, B. Lamarthée, and J. Arbez, IL-22 deficiency in donor T cells attenuates murine acute graft-versus-host disease mortality while sparing the graft-versusleukemia effect, Leukemia, vol.27, issue.7, pp.1527-1537, 2013.

R. Noth, J. Lange-grumfeld, and E. Stüber, Increased intestinal permeability and tight junction disruption by altered expression and localization of occludin in a murine graft versus host disease model, BMC Gastroenterol, vol.11, issue.1, p.109, 2011.

A. M. Marchiando, L. Shen, and . Graham-w-vallen, The epithelial barrier is maintained by in vivo tight junction expansion during pathologic intestinal epithelial shedding, Gastroenterology, vol.140, issue.4, pp.1208-1218, 2011.

D. W. Van-bekkum, J. Roodenburg, P. J. Heidt, and D. Van-der-waaij, Mitigation of Secondary Disease of Allogeneic Mouse Radiation Chimeras by Modification of the Intestinal Microflora, J Natl Cancer Inst, vol.52, issue.2, pp.401-404, 1974.

C. D. Buckner, R. A. Clift, and J. E. Sanders, Protective environment for marrow transplant recipients. A prospective study, Ann Intern Med, vol.89, issue.6, pp.893-901, 1978.

J. M. Vossen, P. J. Heidt, H. Van-den-berg, E. Gerritsen, J. Hermans et al., Prevention of infection and graft-versus-host disease by suppression of intestinal microflora in children treated with allogeneic bone marrow transplantation, Eur J Clin Microbiol Infect Dis, vol.9, issue.1, pp.14-23, 1990.

D. W. Beelen, E. Haralambie, and H. Brandt, Evidence that sustained growth suppression of intestinal anaerobic bacteria reduces the risk of acute graft-versushost disease after sibling marrow transplantation, Blood, vol.80, issue.10, pp.2668-2676, 1992.

D. W. Beelen, A. Elmaagacli, and K. D. Muller, Influence of Intestinal Bacterial Decontamination Using Metronidazole And Ciprofloxacin Or Ciprofloxacin Alone On The Development Of Acute Graft-Versus-Host Disease After Marrow Transplantation In Patients With Hematologic Malignancies: Final Results And Long-term Follow-up Of An Open-Label Prospective Randomized Trial, Blood, vol.93, issue.10, pp.3267-3275, 1999.

Y. Cong, S. L. Brandwein, and R. P. Mccabe, CD4+ T cells reactive to enteric bacterial antigens in spontaneously colitic C3H/HeJBir mice: Increased T helper cell type 1 response and ability to transfer disease, J Exp Med, vol.187, issue.6, pp.855-864, 1998.

V. Gaboriau-routhiau, S. Rakotobe, and E. Lécuyer, The Key Role of Segmented Filamentous Bacteria in the Coordinated Maturation of Gut Helper T Cell Responses, Immunity, vol.31, issue.4, pp.677-689, 2009.

I. I. Ivanov, K. Atarashi, and N. Manel, Induction of intestinal Th17 cells by segmented filamentous bacteria, Cell, vol.139, issue.3, pp.485-498, 2009.

Y. Taur, R. R. Jenq, and M. Perales, The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation, Blood, vol.124, issue.7, pp.1174-1182, 2014.

R. R. Jenq, Y. Taur, and S. M. Devlin, Intestinal Blautia Is Associated with Reduced Death from Graft-versus-Host Disease, Biol Blood Marrow Transplant, vol.21, issue.8, pp.1373-1383, 2015.

J. U. Peled, S. M. Devlin, and A. Staffas, Intestinal Microbiota and Relapse After Hematopoietic-Cell Transplantation, J Clin Oncol, vol.35, issue.15, pp.1650-1659, 2017.

R. R. Jenq, C. Ubeda, and Y. Taur, Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation, J Exp Med, vol.209, issue.5, pp.903-911, 2012.

Y. Shono, M. D. Docampo, and J. U. Peled, Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice, Sci Transl Med, vol.8, issue.339, pp.339-71, 2016.

D. Weber, P. J. Oefner, and K. Dettmer, Rifaximin preserves intestinal microbiota balance in patients undergoing allogeneic stem cell transplantation, Bone Marrow Transplant, pp.1-6, 2015.

T. Teshima, P. Reddy, and R. Zeiser, Acute Graft-versus-Host Disease: Novel Biological Insights, Biol Blood Marrow Transplant, vol.22, issue.1, pp.11-16, 2016.

C. L. Farrell, J. V. Bready, and K. L. Rex, Keratinocyte growth factor protects mice from chemotherapy and radiation-induced gastrointestinal injury and mortality, Cancer Res, vol.58, issue.5, pp.933-939, 1998.

O. I. Krijanovski, G. R. Hill, and K. R. Cooke, Keratinocyte growth factor separates graftversus-leukemia effects from graft-versus-host disease, Blood, vol.94, issue.2, pp.825-831, 1999.

A. Panoskaltsis-mortari, P. A. Taylor, and J. S. Rubin, Keratinocyte growth factor facilitates alloengraftment and ameliorates graft-versus-host disease in mice by a mechanism independent of repair of conditioning-induced tissue injury, Blood, vol.96, issue.13, pp.4350-4356, 2000.

A. Vanclée, L. Lutgens, and E. Oving, Keratinocyte growth factor ameliorates acute graft-versus-host disease in a novel nonmyeloablative haploidentical transplantation model, Bone Marrow Transplant, vol.36, issue.10, pp.907-915, 2005.

B. R. Blazar, D. J. Weisdorf, and T. Defor, Phase 1/2 randomized, placebo-control trial of palifermin to prevent graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT), Blood, vol.108, issue.9, pp.3216-3222, 2006.

J. E. Levine, B. R. Blazar, T. Defor, J. Ferrara, and D. J. Weisdorf, Long-term follow-up of a phase I/II randomized, placebo-controlled trial of palifermin to prevent graft-versushost disease (GVHD) after related donor allogeneic hematopoietic cell transplantation (HCT), Biol Blood Marrow Transplant, vol.14, issue.9, pp.1017-1021, 2008.

R. Spielberger, P. Stiff, and W. Bensinger, Palifermin for Oral Mucositis after Intensive Therapy for Hematologic Cancers, N Engl J Med, vol.351, issue.25, pp.2590-2598, 2004.

K. F. Bradstock, E. Link, and M. Collins, A randomized trial of prophylactic palifermin on gastrointestinal toxicity after intensive induction therapy for acute myeloid leukaemia, Br J Haematol, vol.167, issue.5, pp.618-625, 2014.

D. J. Drucker, P. Erlich, S. L. Asa, and P. L. Brubaker, Induction of intestinal epithelial proliferation by glucagon-like peptide 2, Proc Natl Acad Sci, vol.93, issue.15, pp.7911-7916, 1996.

C. X. Dong, W. Zhao, and C. Solomon, The intestinal epithelial insulin-like growth factor-1 receptor links glucagon-like peptide-2 action to gut barrier function, Endocrinology, vol.155, issue.2, pp.370-379, 2014.

H. Kissow, B. Hartmann, J. J. Holst, and S. S. Poulsen, Glucagon-like peptide-1 as a treatment for chemotherapy-induced mucositis, Gut, vol.62, issue.12, pp.1724-1733, 2013.

L. B. Bindels, A. M. Neyrinck, and A. Loumaye, Increased gut permeability in cancer cachexia: Mechanisms and clinical relevance, Oncotarget, vol.9, issue.26, pp.18224-18238, 2018.

A. Orhan, I. Gögenur, and H. Kissow, The intestinotrophic effects of glucagon-like peptide-2 in relation to intestinal neoplasia, J Clin Endocrinol Metab, vol.103, issue.8, pp.2827-2837, 2018.

D. Seguy, C. Berthon, and J. Micol, Enteral feeding and early outcomes of patients undergoing allogeneic stem cell transplantation following myeloablative conditioning, Transplantation, vol.82, issue.6, pp.835-839, 2006.

S. Azarnoush, B. Bruno, and L. Beghin, Enteral nutrition: a first option for nutritional support of children following allo-SCT?, Bone Marrow Transplant, vol.47, issue.9, pp.1191-1195, 2012.

D. Seguy, A. Duhamel, M. Rejeb, and . Ben, Better outcome of patients undergoing enteral tube feeding after myeloablative conditioning for allogeneic stem cell transplantation, Transplantation, vol.94, issue.3, pp.287-294, 2012.

F. Gonzales, B. Bruno, A. Fuentes, and M. , Better early outcome with enteral rather than parenteral nutrition in children undergoing MAC allo-SCT, Clin Nutr, vol.37, issue.6, pp.2113-2121, 2018.

D. M. Lilly and R. H. Stillwell, Probiotics: Growth-Promoting Factors Produced by Microorganisms, Science, vol.147, issue.3659, pp.747-748, 1964.

A. Geirnaert, M. Calatayud, and C. Grootaert, Butyrate-producing bacteria supplemented in vitro to Crohn's disease patient microbiota increased butyrate production and enhanced intestinal epithelial barrier integrity, Sci Rep, vol.7, issue.1, pp.1-14, 2017.

C. Y. Yeung, W. T. Chan, C. Jiang, and . Bin, Amelioration of chemotherapy-induced intestinal mucositis by orally administered probiotics in a mouse model, PLoS One, vol.10, issue.9, pp.1-16, 2015.

A. Gerbitz, Probiotic effects on experimental graft-versus-host disease: let them eat yogurt, Blood, vol.103, issue.11, pp.4365-4367, 2004.

E. Gorshein, C. Wei, and S. Ambrosy, Lactobacillus rhamnosus GG probiotic enteric regimen does not appreciably alter the gut microbiome or provide protection against GVHD after allogeneic hematopoietic stem cell transplantation, Clin Transplant, vol.31, p.12947, 2017.

S. A. Cohen, M. C. Woodfield, N. Boyle, Z. Stednick, M. Boeckh et al., Incidence and outcomes of bloodstream infections among hematopoietic cell transplant recipients from species commonly reported to be in over-the-counter probiotic formulations, Transpl Infect Dis, vol.18, issue.5, pp.699-705, 2016.

A. J. Riquelme, M. A. Calvo, and A. M. Guzma, Saccharomyces cerevisiae Fungemia After Saccharomyces boulardii Treatment in Immunocompromised Patients, J Clin Gastroenterol, vol.36, issue.1, pp.41-43, 2003.

J. S. Bakken, T. Borody, and L. J. Brandt, Treating clostridium difficile infection with fecal microbiota transplantation, Clin Gastroenterol Hepatol, vol.9, issue.12, pp.1044-1049, 2011.

K. Kakihana, Y. Fujioka, and W. Suda, Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut, Blood, vol.128, issue.16, pp.2083-2088, 2016.

Z. Defilipp, J. U. Peled, and S. Li, Third-party fecal microbiota transplantation following allo-HCT reconstitutes microbiome diversity, Blood Adv, vol.2, issue.7, pp.745-753, 2018.

L. Bastard, Q. Ward, T. Sidiropoulos, and D. , Fecal microbiota transplantation reverses antibiotic and chemotherapy-induced gut dysbiosis in mice, Sci Rep, vol.8, issue.1, pp.1-11, 2018.

U. A. Wenzel, M. K. Magnusson, and A. Rydström, Spontaneous colitis in Muc2-deficient mice reflects clinical and cellular features of active ulcerative colitis, PLoS One, vol.9, issue.6, pp.1-12, 2014.

A. Velcich, W. C. Yang, and J. Heyer, Colorectal cancer in mice genetically deficient in the mucin Muc2, Science, vol.295, issue.5560, pp.1726-1729, 2002.

C. A. Hickey, K. A. Kuhn, and D. L. Donermeyer, Colitogenic Bacteroides thetaiotaomicron antigens access host immune cells in a sulfatase-dependent manner via outer membrane vesicles, Cell Host Microbe, vol.17, issue.5, pp.672-680, 2015.

B. Demouveaux, V. Gouyer, F. Gottrand, T. Narita, and J. L. Desseyn, Gel-forming mucin interactome drives mucus viscoelasticity, Adv Colloid Interface Sci, vol.252, pp.69-82, 2018.

V. Gouyer, L. Dubuquoy, and C. Robbe-masselot, Delivery of a mucin domain enriched in cysteine residues strengthens the intestinal mucous barrier, Sci Rep, vol.5, p.9577, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02380839

M. J. Van-vliet, W. Tissing, and C. Dun, Chemotherapy Treatment in Pediatric Patients with Acute Myeloid Leukemia Receiving Antimicrobial Prophylaxis Leads to a Relative Increase of Colonization with Potentially Pathogenic Bacteria in the Gut, Clin Infect Dis, vol.49, issue.2, pp.262-270, 2009.

M. J. Van-vliet, W. Tissing, and E. Rings, Citrulline as a Marker for Chemotherapy Induced Mucosal Barrier Injury in Pedatric Patients, Pediatr Blood Cancer, vol.53, pp.1188-1194, 2009.

J. Zuber, I. Radtke, and T. S. Pardee, Mouse models of human AML accurately predict chemotherapy response, Genes Dev, vol.23, issue.7, pp.877-889, 2009.

M. Wunderlich, B. Mizukawa, and F. Chou, AML cells are differentially sensitive to chemotherapy treatment in a human xenograft model, Blood, vol.121, issue.12, pp.90-97, 2013.

J. R. Galloway-peña, D. P. Smith, and P. Sahasrabhojane, The role of the gastrointestinal microbiome in infectious complications during induction chemotherapy for acute myeloid leukemia, Cancer, vol.122, issue.14, pp.2186-2196, 2016.

C. Ubeda, Y. Taur, and R. R. Jenq, Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans, J Clin Invest, vol.120, issue.12, pp.4332-4341, 2010.

A. C. Harris, J. Ferrara, and T. M. Braun, Plasma biomarkers of lower gastrointestinal and liver acute GVHD, Blood, vol.119, issue.12, pp.2960-2963, 2012.

G. A. Kennedy, A. Varelias, and S. Vuckovic, Addition of interleukin-6 inhibition with tocilizumab to standard graft-versus-host disease prophylaxis after allogeneic stemcell transplantation: A phase 1/2 trial, Lancet Oncol, vol.15, issue.13, pp.1451-1459, 2014.

M. T. Vander-lugt, T. M. Braun, and S. Hanash, ST2 as a Marker for Risk of Therapy-Resistant Graft-versus-Host Disease and Death, N Engl J Med, vol.369, issue.6, pp.529-539, 2013.

G. B. Mcdonald, L. Tabellini, B. E. Storer, R. L. Lawler, P. J. Martin et al., Plasma biomarkers of acute GVHD and nonrelapse mortality: Predictive value of measurements before GVHD onset and treatment, Blood, vol.126, issue.1, pp.113-120, 2015.

M. J. Hartwell, U. Özbek, and E. Holler, An early-biomarker algorithm predicts lethal graft-versus-host disease and survival, JCI Insight, vol.2, issue.3, p.89798, 2017.

, Résumés La chimiothérapie d'induction et de consolidation suivie d'allogreffe de cellules souches hématopoïétiques (allo-CSH) immunologiques, telles que la réaction aiguë du greffon contre l'hôte (GvH)

, une étude translationnelle, l'impact et les conséquences de cette chimiothérapie d'induction sur la barrière intestinale. Pour cela, nous avons évalué sur une série de 15 patients atteints de LAM, les paramètres cliniques, biologiques (citrulline plasmatique et acide gras à chaine courte dans les selles) et microbiologiques (qPCR et séquençages des fèces) avant la chimiothérapie d'induction (T0)

, Cette chimiothérapie d'induction a ensuite été adaptée pour pouvoir être administrée, sans ajout d'aucun antibiotique, à des souris sauvages (WT) et à des souris transgéniques dont le mucus intestinal est renforcé (Tg222) afin d'évaluer : i. les dommages intestinaux (citrulline plasmatique et analyse des coupes histologiques d'iléon avec marquage de l'apoptose (TUNEL)