K. S. Schluns and L. Lefrançois, Cytokine control of memory T-cell development and survival, Nat. Rev. Immunol, vol.3, pp.269-279, 2003.

A. C. Boesteanu and P. D. Katsikis, Memory T cells need CD28 costimulation to remember, Semin. Immunol, vol.21, pp.69-77, 2009.

T. M. K?-undig, A. Shahinian, K. Kawai, H. W. Mittr?-ucker, E. Sebzda et al., Duration of TCR stimulation determines costimulatory requirement of T cells, Immunity, vol.5, pp.41-52, 1996.

A. Viola and A. Lanzavecchia, T cell activation determined by T cell receptor number and tunable thresholds, Science, vol.273, pp.104-106, 1996.

M. Suresh, J. K. Whitmire, L. E. Harrington, C. P. Larsen, T. C. Pearson et al., Role of CD28-B7 interactions in generation and maintenance of CD8 T cell memory, J. Immunol, vol.167, pp.5565-5573, 2001.

S. K. Kim, K. S. Schluns, and L. Lefrançois, Induction and visualization of mucosal memory CD8 T cells following systemic virus infection, J. Immunol, vol.163, pp.4125-4132, 1999.

T. A. Weaver, A. H. Charafeddine, A. Agarwal, A. P. Turner, M. Russell et al., Alefacept promotes co-stimulation blockade based allograft survival in nonhuman primates, Nat. Med, vol.15, pp.746-749, 2009.

O. Nadazdin, S. Boskovic, T. Murakami, G. Tocco, R. Smith et al., Host alloreactive memory T cells influence tolerance to kidney allografts in nonhuman primates, Sci. Transl. Med, vol.3, pp.86-51, 2011.

D. Eastwood, L. Findlay, S. Poole, C. Bird, M. Wadhwa et al., Monoclonal antibody TGN1412 trial failure explained by species differences in CD28 expression on CD4 + effector memory T-cells, Br. J. Pharmacol, vol.161, pp.512-526, 2010.

N. Poirier, C. Mary, S. L. Bas-bernardet, V. Daguin, L. Belarif et al., Advantages of Papio anubis for preclinical testing of immunotoxicity of candidate therapeutic antagonist antibodies targeting CD28, MAbs, vol.6, pp.697-707, 2014.

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

N. Poirier, G. Blancho, and B. Vanhove, Alternatives to calcineurin inhibition in renal transplantation: belatacept, the first co-stimulation blocker, Immunotherapy, vol.2, pp.625-636, 2010.

S. M. Krummey, J. A. Cheeseman, J. A. Conger, P. S. Jang, A. K. Mehta et al., High CTLA-4 expression on Th17 cells results in increased sensitivity to CTLA-4 coinhibition and resistance to belatacept, Am. J. Transplant, vol.14, pp.607-614, 2014.

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

N. Poirier, A. M. Azimzadeh, T. Zhang, N. Dilek, C. Mary et al., Inducing CTLA-4-dependent immune regulation by selective CD28 blockade promotes regulatory T cells in organ transplantation, Sci. Transl. Med, vol.2, pp.17-27, 2010.

A. Dugast, T. Haudebourg, F. Coulon, M. Heslan, F. Haspot et al., Myeloidderived suppressor cells accumulate in kidney allograft tolerance and specifically suppress effector T cell expansion, J. Immunol, vol.180, pp.7898-7906, 2008.

N. Dilek, N. Poirier, C. Usal, B. Martinet, G. Blancho et al., Control of transplant tolerance and intragraft regulatory T cell localization by myeloid-derived suppressor cells and CCL5, J. Immunol, vol.188, pp.4209-4216, 2012.

T. Zhang, S. Fresnay, E. Welty, N. Sangrampurkar, E. Rybak et al., Selective CD28 blockade attenuates acute and chronic rejection of murine cardiac allografts in a CTLA-4-dependent manner, Am. J. Transplant, vol.11, pp.1599-1609, 2011.

N. Dilek, N. Poirier, P. Hulin, F. Coulon, C. Mary et al., Targeting CD28, CTLA-4 and PD-L1 costimulation differentially controls immune synapses and function of human regulatory and conventional T-cells, PLoS One, vol.8, p.83139, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02165637

N. Poirier, C. Mary, N. Dilek, J. Hervouet, D. Minault et al., Preclinical efficacy and immunological safety of FR104, an antagonist anti-CD28 monovalent Fab9 antibody, Am. J. Transplant, vol.12, pp.2630-2640, 2012.

C. Mary, F. Coulon, N. Poirier, N. Dilek, B. Martinet et al., Antagonist properties of monoclonal antibodies targeting human CD28: role of valency and the heavy-chain constant domain, MAbs, vol.5, pp.47-55, 2013.

N. Poirier, T. Haudebourg, C. Brignone, N. Dilek, J. Hervouet et al., Antibody-mediated depletion of lymphocyte-activation gene-3 (LAG-3 + )-activated T lymphocytes prevents delayed-type hypersensitivity in non-human primates, Clin. Exp. Immunol, vol.164, pp.265-274, 2011.

N. Poirier, N. Dilek, C. Mary, S. Ville, F. Coulon et al., FR104, an antagonist anti-CD28 monovalent Fab9 antibody, prevents alloimmunization and allows calcineurin inhibitor minimization in nonhuman primate renal allograft, Am. J. Transplant, vol.15, pp.88-100, 2015.

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

K. M. Rogers, J. W. Ritchey, M. Payton, D. H. Black, and R. Eberle, Neuropathogenesis of herpesvirus papio 2 in mice parallels infection with Cercopithecine herpesvirus 1 (B virus) in humans, J. Gen. Virol, vol.87, pp.267-276, 2006.

U. Galili, M. R. Clark, S. B. Shohet, J. Buehler, and B. A. Macher, Evolutionary relationship between the natural anti-Gal antibody and the Gal alpha 1-3Gal epitope in primates, Proc. Natl. Acad. Sci. USA, vol.84, pp.1369-1373, 1987.

Z. Wu, S. J. Bensinger, J. Zhang, C. Chen, X. Yuan et al., Homeostatic proliferation is a barrier to transplantation tolerance, Nat. Med, vol.10, pp.87-92, 2004.

A. B. Adams, T. C. Pearson, and C. P. Larsen, Heterologous immunity: an overlooked barrier to tolerance, Immunol. Rev, vol.196, pp.147-160, 2003.

I. R. Badell, M. C. Russell, P. W. Thompson, A. P. Turner, T. A. Weaver et al., LFA-1-specific therapy prolongs allograft survival in rhesus macaques, J. Clin. Invest, vol.120, pp.4520-4531, 2010.

H. Xu, S. D. Perez, J. Cheeseman, A. K. Mehta, and A. D. Kirk, The alloand viral-specific immunosuppressive effect of belatacept, but not tacrolimus, attenuates with progressive T cell maturation, Am. J. Transplant, vol.14, pp.319-332, 2014.

S. M. Krummey, T. L. Floyd, D. Liu, M. E. Wagener, M. Song et al., Candida-elicited murine Th17 cells express high Ctla-4 compared with Th1 cells and are resistant to costimulation blockade, J. Immunol, vol.192, pp.2495-2504, 2014.

J. H. Rowe, T. M. Johanns, J. M. Ertelt, J. C. Lai, and S. S. Way, Cytotoxic T-lymphocyte antigen 4 blockade augments the T-cell response primed by attenuated Listeria monocytogenes resulting in more rapid clearance of virulent bacterial challenge, Immunology, vol.128, issue.1, pp.471-478, 2009.

V. Mirenda, S. J. Jarmin, R. David, J. Dyson, D. Scott et al., Physiologic and aberrant regulation of memory T-cell trafficking by the costimulatory molecule CD28, Blood, vol.109, pp.2968-2977, 2007.

S. P. Raychaudhuri, S. Kundu-raychaudhuri, K. Tamura, T. Masunaga, K. Kubo et al., FR255734, a humanized, Fc-silent, anti-CD28 antibody, improves psoriasis in the SCID mouse-psoriasis xenograft model, J. Invest. Dermatol, vol.128, pp.1969-1976, 2008.

S. L. Shiao, J. M. Mcniff, T. Masunaga, K. Tamura, K. Kubo et al., Immunomodulatory properties of FK734, a humanized anti-CD28 monoclonal antibody with agonistic and antagonistic activities, Transplantation, vol.83, pp.304-313, 2007.

J. B. Wing, W. Ise, T. Kurosaki, and S. Sakaguchi, Regulatory T cells control antigen-specific expansion of Tfh cell number and humoral immune responses via the coreceptor CTLA-4, Immunity, vol.41, pp.1013-1025, 2014.

P. T. Sage, A. M. Paterson, S. B. Lovitch, and A. H. Sharpe, The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells, Immunity, vol.41, pp.1026-1039, 2014.

C. J. Wang, F. Heuts, V. Ovcinnikovs, L. Wardzinski, C. Bowers et al., CTLA-4 controls follicular helper T-cell differentiation by regulating the strength of CD28 engagement, Proc. Natl. Acad. Sci. USA, vol.112, pp.524-529, 2015.

P. T. Sage, D. Alvarez, J. Godec, U. H. Von-andrian, and A. H. Sharpe, Circulating T follicular regulatory and helper cells have memory-like properties, J. Clin. Invest, vol.124, pp.5191-5204, 2014.

L. M. Tsai and D. Yu, Follicular helper T-cell memory: establishing new frontiers during antibody response, Immunol. Cell Biol, vol.92, pp.57-63, 2014.

J. P. Weber, F. Fuhrmann, and A. Hutloff, T-follicular helper cells survive as long-term memory cells, Eur. J. Immunol, vol.42, pp.1981-1988, 2012.

L. Bas-bernardet, S. , X. Tillou, J. Branchereau, N. Dilek et al., Bortezomib, C1-inhibitor and plasma exchange do not prolong the survival of multi-transgenic GalT-KO pig kidney xenografts in baboons, Am. J. Transplant, vol.15, pp.358-370, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02148475

L. Bas-bernardet, S. , X. Tillou, N. Poirier, N. Dilek et al., Xenotransplantation of galactosyl-transferase knockout, CD55, CD59, CD39, and fucosyl-transferase transgenic pig kidneys into baboons, Transplant. Proc, vol.43, pp.3426-3430, 2011.

N. Poirier and G. Blancho, Recombinant human C1-inhibitor inhibits cytotoxicity induced by allo-and xenoantibodies, Transplant. Proc, vol.40, pp.581-583, 2008.

C. P. Larsen, T. C. Pearson, A. B. Adams, P. Tso, N. Shirasugi et al., Rational development of LEA29Y (belatacept), a high-affinity variant of CTLA4-Ig with potent immunosuppressive properties, Am. J. Transplant, vol.5, pp.443-453, 2005.

C. L. Mccarthy, O. Tuohy, D. A. Compston, D. S. Kumararatne, A. J. Coles et al., Immune competence after alemtuzumab treatment of multiple sclerosis, Neurology, vol.81, pp.872-876, 2013.

Y. Xing, H. M. Song, M. Wei, Y. Liu, Y. H. Zhang et al., Clinical significance of variations in levels of Epstein-Barr Virus (EBV) antigen and adaptive immune response during chronic active EBV infection in children, J. Immunotoxicol, vol.10, pp.387-392, 2013.

A. Shahinian, K. Pfeffer, K. P. Lee, T. M. K?-undig, K. Kishihara et al., Differential T cell costimulatory requirements in CD28-deficient mice, Science, vol.261, pp.609-612, 1993.

S. T. Martin, J. T. Powell, M. Patel, and D. Tsapepas, Risk of posttransplant lymphoproliferative disorder associated with use of belatacept, Am. J. Health Syst. Pharm, vol.70, pp.1977-1983, 2013.