M. Jagasia, Risk factors for acute GVHD and survival after hematopoietic cell transplantation, Blood, vol.119, issue.1, pp.296-307, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00849704

S. J. Lee, High-resolution donor-recipient HLA matching contributes to the success of unrelated donor marrow transplantation, Blood, vol.110, issue.13, pp.4576-4583, 2007.

P. J. Shaw, Outcomes of pediatric bone marrow transplantation for leukemia and myelodysplasia using matched sibling, mismatched related, or matched unrelated donors, Blood, vol.116, pp.4007-4015, 2010.

J. E. Wagner, J. S. Thompson, S. L. Carter, and N. A. Kernan, Unrelated Donor Marrow Transplantation Trial. Effect of graft-versus-host disease prophylaxis on 3-year disease-free survival in recipients of unrelated donor bone marrow (T-cell Depletion Trial): a multi-centre, randomised phase II-III trial, Lancet, vol.366, issue.9487, pp.733-741, 2005.

J. D. Rizzo, Recommended screening and preventive practices for long-term survivors after hematopoietic cell transplantation: joint recommendations of the European Group for Blood and Marrow Transplantation, Center for International Blood and Marrow Transplant Research, and the American Society for Blood and Marrow Transplantation (EBMT/CIBMTR/ASBMT)

, Bone Marrow Transplant, vol.37, issue.3, pp.249-261, 2006.

R. Ram and R. Storb, Pharmacologic prophylaxis regimens for acute graft-versus-host disease

, Leuk Lymphoma, vol.54, issue.8, pp.1591-1601, 2013.

P. Blaha, The influence of immunosuppressive drugs on tolerance induction through bone marrow transplantation with costimulation blockade, Blood, vol.101, issue.7, pp.2886-2893, 2003.

A. Furukawa, S. A. Wisel, and Q. Tang, Impact of immune-modulatory drugs on regulatory T cell, Transplantation, vol.100, issue.11, pp.2288-2300, 2016.

J. M. Kremer, Treatment of rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein CTLA4Ig, N Engl J Med, vol.349, issue.20, pp.1907-1915, 2003.

F. Vincenti, A phase III study of belatacept-based immunosuppression regimens versus cyclosporine in renal transplant recipients (BENE-FIT study), Am J Transplant, vol.10, issue.3, pp.535-546, 2010.

D. T. Koura, In vivo T cell costimulation blockade with abatacept for acute graft-versus-host disease prevention: a first-in-disease trial, Biol Blood Marrow Transplant, vol.19, issue.11, pp.1638-1649, 2013.

W. P. Miller, GVHD after haploidentical transplantation: a novel, MHC-defined rhesus macaque model identifies CD28-CD8+ T cells as a reservoir of breakthrough T-cell proliferation during costimulation blockade and sirolimus-based immunosuppression, Blood, vol.116, issue.24, pp.5403-5418, 2010.

J. Pidala, L. Perez, F. Beato, and C. Anasetti, Ustekinumab demonstrates activity in glucocorticoidrefractory acute GVHD, Bone Marrow Transplant, vol.47, issue.5, pp.747-748, 2012.

D. Gardner, L. E. Jeffery, and D. M. Sansom, Understanding the CD28/CTLA-4 (CD152) pathway and its implications for costimulatory blockade, Am J Transplant, vol.14, issue.9, pp.1985-1991, 2014.

S. Chitale and R. Moots, Abatacept: the first T lymphocyte co-stimulation modulator, for the treatment of rheumatoid arthritis, Expert Opin Biol Ther, vol.8, issue.1, pp.115-122, 2008.

D. E. Furst, Updated consensus statement on biological agents for the treatment of rheumatic diseases, Ann Rheum Dis, vol.71, issue.2, pp.2-45, 2011.

M. C. Genovese, Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition, N Engl J Med, vol.353, issue.11, pp.1114-1123, 2005.

C. P. Larsen, Rational development of LEA29Y (belatacept), a high-affinity variant of CTLA4-Ig with potent immunosuppressive properties, Am J Transplant, vol.5, issue.3, pp.443-453, 2005.

F. Vincenti, Costimulation blockade with belatacept in renal transplantation, N Engl J Med, vol.353, issue.8, pp.770-781, 2005.

R. Westhovens, Clinical efficacy and safety of abatacept in methotrexate-naive patients with early rheumatoid arthritis and poor prognostic factors, Ann Rheum Dis, vol.68, issue.12, pp.1870-1877, 2009.

J. M. Kremer, Long-term safety, efficacy and inhibition of radiographic progression with abatacept treatment in patients with rheumatoid arthritis and an inadequate response to methotrexate: 3-year results from the AIM trial, Ann Rheum Dis, vol.70, issue.10, pp.1826-1830, 2011.

M. Schiff, Efficacy and safety of abatacept or infliximab vs. placebo in ATTEST: a phase III, multi-centre, randomised, double-blind, placebo-controlled study in patients with rheumatoid arthritis and an inadequate response to methotrexate, Ann Rheum Dis, vol.67, issue.8, pp.1096-1103, 2008.

R. Westhovens, Safety and efficacy of the selective costimulation modulator abatacept in patients with rheumatoid arthritis receiving background methotrexate: a 5-year extended phase IIB study, J Rheumatol, vol.36, issue.4, pp.736-742, 2009.

H. Chavez, Absence of CD4CD25 regulatory T cell expansion in renal transplanted patients treated in vivo with Belatacept mediated CD28-CD80/86 blockade, Transpl Immunol, vol.17, issue.4, pp.243-248, 2007.

N. Dilek, CTLA-4 and PD-L1 costimulation differentially controls immune synapses and function of human regulatory and conventional T-cells, PLoS ONE, vol.28, issue.12, p.83139, 2013.

S. T. Haile, S. P. Dalal, V. Clements, K. Tamada, and S. Ostrand-rosenberg, Soluble CD80 restores T cell activation and overcomes tumor cell programmed death ligand 1-mediated immune suppression, J Immunol, vol.191, issue.5, pp.2829-2836, 2013.

A. Izawa, A novel alloantigen-specific CD8+PD1+ regulatory T cell induced by ICOS-B7h blockade in vivo, J Immunol, vol.179, issue.2, pp.786-796, 2007.

J. Kurtz, F. Raval, C. Vallot, J. Der, and M. Sykes, CTLA-4 on alloreactive CD4 T cells interacts with recipient CD80/86 to promote tolerance, Blood, vol.113, issue.15, pp.3475-3484, 2009.

J. Levitsky, J. Miller, X. Huang, D. Chandrasekaran, L. Chen et al., Inhibitory effects of belatacept on allospecific regulatory T-cell generation in humans, Transplantation, vol.96, issue.8, pp.689-696, 2013.

L. V. Riella, Deleterious effect of CTLA4-Ig on a Treg-dependent transplant model, Am J Transplant, vol.12, issue.4, pp.846-855, 2012.

B. Salomon and J. A. Bluestone, Complexities of CD28/ B7: CTLA-4 costimulatory pathways in autoimmunity and transplantation, Annu Rev Immunol, vol.19, pp.225-252, 2001.

B. Salomon, B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes, Immunity, vol.12, issue.4, pp.431-440, 2000.

P. Waterhouse, Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4, Science, vol.270, issue.5238, pp.985-988, 1995.

K. Wing, CTLA-4 control over Foxp3+ regulatory T cell function, Science, vol.322, issue.5899, pp.271-275, 2008.

B. H. Koehn, PD-1-dependent mechanisms maintain peripheral tolerance of donor-reactive CD8+ T cells to transplanted tissue, J Immunol, vol.181, issue.8, pp.5313-5322, 2008.

N. Poirier, G. Blancho, and B. Vanhove, A more selective costimulatory blockade of the CD28-B7 pathway, Transpl Int, vol.24, issue.1, pp.2-11, 2011.

N. Poirier, G. Blancho, and B. Vanhove, CD28-specific immunomodulating antibodies: what can be learned from experimental models?, Am J Transplant, vol.12, issue.7, pp.1682-1690, 2012.

X. Z. Yu, M. H. Albert, P. J. Martin, and C. Anasetti, CD28 ligation induces transplantation tolerance by IFN-gamma-dependent depletion of T cells that recognize alloantigens, J Clin Invest, vol.113, issue.11, pp.1624-1630, 2004.

X. Z. Yu, P. J. Martin, and C. Anasetti, Role of CD28 in acute graft-versus-host disease, Blood, vol.92, issue.8, pp.2963-2970, 1998.

X. Z. Yu, P. J. Martin, and C. Anasetti, CD28 signal enhances apoptosis of CD8 T cells after strong TCR ligation, J Immunol, vol.170, issue.6, pp.3002-3006, 2003.

B. Vanhove, Selective blockade of CD28 and not CTLA-4 with a single-chain Fv-alpha1-antitrypsin fusion antibody, Blood, vol.102, issue.2, pp.564-570, 2003.

K. G. Haanstra, Selective blockade of CD28-mediated T cell costimulation protects rhesus monkeys against acute fatal experimental autoimmune encephalomyelitis, J Immunol, vol.194, issue.4, pp.1454-1466, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02148517

K. L. Hippen, Preclinical testing of antihuman CD28 Fab? antibody in a novel nonhuman primate small animal rodent model of xenogenic graft-versus-host disease, Transplantation, vol.100, issue.12, pp.2630-2639, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02150241

D. Liu, 2B4 (CD244) induced by selective CD28 blockade functionally regulates allograft-specific CD8+ T cell responses, J Exp Med, vol.211, issue.2, pp.297-311, 2014.

N. Poirier, FR104, an antagonist anti-CD28 monovalent fab? antibody, prevents alloimmunization and allows calcineurin inhibitor minimization in nonhuman primate renal allograft, Am J Transplant, vol.15, issue.1, pp.88-100, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02148443

N. Poirier, Preclinical efficacy and immunological safety of FR104, an antagonist anti-CD28 monovalent Fab? antibody, Am J Transplant, vol.12, issue.10, pp.2630-2640, 2012.

S. J. Suchard, A monovalent anti-human CD28 domain antibody antagonist: preclinical efficacy and safety, J Immunol, vol.191, issue.9, pp.4599-4610, 2013.

M. P. Vierboom, Clinical efficacy of a new CD28-targeting antagonist of T cell co-stimulation in a non-human primate model of collagen-induced arthritis, Clin Exp Immunol, vol.183, issue.3, pp.405-418, 2016.

S. Ville, Anti-CD28 antibody and belatacept exert differential effects on mechanisms of renal allograft rejection, J Am Soc Nephrol, vol.27, issue.12, pp.3577-3588, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02150329

N. Poirier, Inducing CTLA-4-dependent immune regulation by selective CD28 blockade promotes regulatory T cells in organ transplantation, Sci Transl Med, vol.2, issue.17, pp.17-27, 2010.

S. N. Furlan, Systems analysis uncovers inflammatory Th/Tc17-driven modules during acute GVHD in monkey and human T cells, Blood, vol.128, issue.21, pp.2568-2579, 2016.

S. N. Furlan, Transcriptome analysis of GVHD reveals aurora kinase A as a targetable pathway for disease prevention, Sci Transl Med, vol.7, issue.315, pp.315-191, 2015.

S. Kaliyaperumal, CD8-predominant T-cell CNS infiltration accompanies GVHD in primates and is improved with immunoprophylaxis, Blood, vol.123, issue.12, pp.1967-1969, 2014.

A. Subramanian, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles, Proc Natl Acad Sci U S A, vol.102, issue.43, pp.15545-15550, 2005.

J. Attanasio and E. J. Wherry, Costimulatory and coinhibitory receptor pathways in infectious disease, Immunity, vol.44, issue.5, pp.1052-1068, 2016.

D. W. Huang, B. T. Sherman, and R. A. Lempicki,

, atic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc, vol.4, pp.44-57, 2009.

B. Zhang and S. Horvath, A general framework for weighted gene co-expression network analysis, Stat Appl Genet Mol Biol, vol.4, p.17, 2005.

A. P. Presson, Integrated weighted gene co-expression network analysis with an application to chronic fatigue syndrome, BMC Syst Biol, vol.2, p.95, 2008.

P. Langfelder and S. Horvath, Eigengene networks for studying the relationships between co-expression modules, BMC Syst Biol, vol.1, p.54, 2007.

P. Shannon, Cytoscape: a software environment for integrated models of biomolecular interaction networks, Genome Res, vol.13, issue.11, pp.2498-2504, 2003.

R. Zhang, A. Huynh, G. Whitcher, J. Chang, J. S. Maltzman et al., An obligate cellintrinsic function for CD28 in Tregs, J Clin Invest, vol.123, issue.2, pp.580-593, 2013.

V. Tkachev, Combined OX40L and mTOR blockade controls effector T cell activation while preserving Treg reconstitution after transplant, Sci Transl Med, vol.9, issue.408, p.3085, 2017.

W. P. Arend, M. Malyak, C. J. Guthridge, and C. Gabay, Interleukin-1 receptor antagonist: role in biology, Annu Rev Immunol, vol.16, pp.27-55, 1998.

T. Tanaka and T. Kishimoto, The biology and medical implications of interleukin-6, Cancer Immunol Res, vol.2, issue.4, pp.288-294, 2014.

G. Trinchieri, Interleukin-12 and the regulation of innate resistance and adaptive immunity, Nat Rev Immunol, vol.3, issue.2, pp.133-146, 2003.

J. T. Harty and M. J. Bevan, Specific immunity to Listeria monocytogenes in the absence of IFN gamma, Immunity, vol.3, issue.1, pp.109-117, 1995.

K. R. Kasten, J. Tschöp, S. G. Adediran, D. A. Hildeman, and C. C. Caldwell, T cells are potent early mediators of the host response to sepsis, Shock, vol.34, issue.4, pp.327-336, 2010.

K. A. Messingham, V. P. Badovinac, A. Jabbari, and J. T. Harty, A role for IFN-gamma from antigen-specific CD8+ T cells in protective immunity to Listeria monocytogenes, J Immunol, vol.179, issue.4, pp.2457-2466, 2007.

F. Guo, C. Iclozan, W. K. Suh, C. Anasetti, and X. Z. Yu, CD28 controls differentiation of regulatory T cells from naive CD4 T cells, J Immunol, vol.181, issue.4, pp.2285-2291, 2008.

K. Semple, A. Nguyen, Y. Yes, H. Wang, C. Anasetti et al., Strong CD28 costimulation suppresses induction of regulatory T cells from naive precursors through Lck signaling, Blood, vol.117, issue.11, pp.3096-3103, 2011.

Q. Tang, Cutting edge: CD28 controls peripheral homeostasis of CD4+CD25+ regulatory T cells, J Immunol, vol.171, issue.7, pp.3348-3352, 2003.

A. Huynh, R. Zhang, and L. A. Turka, Signals and pathways controlling regulatory T cells, Immunol Rev, vol.258, issue.1, pp.117-131, 2014.

H. Bour-jordan, B. L. Salomon, H. L. Thompson, G. L. Szot, M. R. Bernhard et al., Costimulation controls diabetes by altering the balance of pathogenic and regulatory T cells, J Clin Invest, vol.114, issue.7, pp.979-987, 2004.

S. O. Ciurea, Improved early outcomes using a T cell replete graft compared with T cell depleted haploidentical hematopoietic stem cell transplantation, Biol Blood Marrow Transplant, vol.18, issue.12, pp.1835-1844, 2012.

A. M. Marmont, T-cell depletion of HLA-identical transplants in leukemia, Blood, vol.78, issue.8, pp.2120-2130, 1991.

M. F. Martelli, Designed" grafts for HLA-haploidentical stem cell transplantation, Blood, vol.123, issue.7, pp.967-973, 2014.

P. J. Martin, Donor CD8 cells prevent allogeneic marrow graft rejection in mice: potential implications for marrow transplantation in humans, J Exp Med, vol.178, issue.2, pp.703-712, 1993.

L. Bry, M. Brigl, and M. B. Brenner, CD4 + -T-cell effector functions and costimulatory requirements essential for surviving mucosal infection with Citrobacter rodentium, Infect Immun, vol.74, issue.1, pp.673-681, 2006.

M. Fröhlich, T. Gogishvili, D. Langenhorst, F. Lühder, and T. Hünig,

, T-cell expansion and effector functions during secondary response in mice, Eur J Immunol, vol.46, issue.7, pp.1644-1655, 2016.

M. A. Linterman, CD28 expression is required after T cell priming for helper T cell responses and protective immunity to infection, Elife, vol.3, p.3180, 2014.

N. Poirier, First-in-human study in healthy subjects with FR104, a pegylated monoclonal antibody fragment antagonist of CD28, J Immunol, vol.197, issue.12, pp.4593-4602, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02150993

R. A. Irizarry, Exploration, normalization, and summaries of high density oligonucleotide array probe level data, Biostatistics, vol.4, issue.2, pp.249-264, 2003.

B. R. Blazar, Engraftment of severe combined immune deficient mice receiving allogeneic bone marrow via In utero or postnatal transfer, Blood, vol.92, issue.10, pp.3949-3959, 1998.

H. B. Zheng, The knife's edge of tolerance: inducing stable multilineage mixed chimerism but with a significant risk of CMV reactivation and disease in rhesus macaques, Am J Transplant, vol.17, issue.3, pp.657-670, 2017.