I. Berber, C. Aydin, and B. Yigit, Blood pressure and graft outcome in renal transplantation, Transplant Proc, vol.37, pp.997-998, 2005.

B. J. Nankivell, R. J. Borrows, C. Fung, O. Connell, P. J. Allen et al., The natural history of chronic allograft nephropathy, N Engl J Med, vol.349, pp.2326-2333, 2003.

E. Porrini, P. Delgado, and C. Bigo, Impact of metabolic syndrome on graft function and survival after cadaveric renal transplantation, Am J Kidney Dis, vol.48, pp.134-142, 2006.

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.

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

F. Vincenti, C. Larsen, and A. Durrbach, Costimulation blockade with belatacept in renal transplantation, N Engl J Med, vol.353, pp.770-781, 2005.

F. Vincenti, B. Charpentier, and Y. Vanrenterghem, A phase III study of belatacept-based immunosuppression regimens versus cyclosporine in renal transplant recipients (BENEFIT study), Am J Transplant, vol.10, pp.535-546, 2010.

F. Vincenti, C. P. Larsen, and J. Alberu, Three-year outcomes from BENEFIT, a randomized, active-controlled, parallel-group study in adult kidney transplant recipients, Am J Transplant, vol.12, pp.210-217, 2012.

A. Durrbach, J. M. Pestana, and T. Pearson, A phase III study of belatacept versus cyclosporine in kidney transplants from extended criteria donors (BENEFIT-EXT study), Am J Transplant, vol.10, pp.547-557, 2010.

Y. Vanrenterghem, B. Bresnahan, and J. Campistol, Belataceptbased regimens are associated with improved cardiovascular and metabolic risk factors compared with cyclosporine in kidney transplant recipients (BENEFIT and BENEFIT-EXT studies), Transplantation, vol.91, pp.976-983, 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.

N. Poirier, A. M. Azimzadeh, and T. Zhang, 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.

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

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.

T. Zhang, S. Fresnay, and E. Welty, 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.

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

F. Haspot, F. Villemain, and G. Laflamme, Differential effect of CD28 versus B7 blockade on direct pathway of allorecognition and self-restricted responses, Blood, vol.99, pp.2228-2234, 2002.

J. Li, K. Semple, and W. Suh, Roles of CD28, CTLA4, and inducible costimulator in acute graft-versus-host disease in mice, Biol Blood Marrow Transplant, vol.17, pp.962-969, 2011.

M. J. Butte, M. E. Keir, T. B. Phamduy, A. H. Sharpe, and G. J. Freeman, Programmed death-1 ligand 1 interacts specifically with the B 7-1 costimulatory molecule to inhibit T cell responses, Immunity, vol.27, pp.111-122, 2007.

M. J. Butte, V. Peña-cruz, M. Kim, G. J. Freeman, and A. H. Sharpe, Interaction of human PD-L1 and B7-1, Mol Immunol, vol.45, pp.3567-3572, 2008.

S. Yao, Y. Zhu, and G. Zhu, B7-h2 is a costimulatory ligand for CD28 in human, Immunity, vol.34, pp.729-740, 2011.

P. J. Perrin, C. H. June, J. H. Maldonado, R. B. Ratts, and M. K. Racke, Blockade of CD28 during in vitro activation of encephalitogenic T cells or after disease onset ameliorates experimental autoimmune encephalomyelitis, J Immunol, vol.163, pp.1704-1710, 1999.

S. P. Raychaudhuri, S. Kundu-raychaudhuri, and K. Tamura, 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.

X. 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, pp.1624-1630, 2004.

X. Z. Yu, S. J. Bidwell, P. J. Martin, and C. Anasetti, CD28-specific antibody prevents graft-versus-host disease in mice, J Immunol, vol.164, pp.4564-4568, 2000.

I. A. Laskowski, J. Pratschke, and M. J. Wilhelm, Anti-CD28 monoclonal antibody therapy prevents chronic rejection of renal allografts in rats, J Am Soc Nephrol, vol.13, pp.519-527, 2002.

V. M. Dong, X. Yuan, A. J. Coito, A. M. Waaga, M. H. Sayegh et al., Mechanisms of targeting CD28 by a signaling monoclonal antibody in acute and chronic allograft rejection, Transplantation, vol.73, pp.1310-1317, 2002.

C. Guillonneau, C. Sé-veno, and A. Dugast, Anti-CD28 antibodies modify regulatory mechanisms and reinforce tolerance in CD40Ig-treated heart allograft recipients, J Immunol, vol.179, pp.8164-8171, 2007.

H. Urakami, D. V. Ostanin, T. Hü-nig, and M. B. Grisham, Combination of donor-specific blood transfusion with anti-CD28 antibody synergizes to prolong graft survival in rat liver transplantation, Transplant Proc, vol.38, pp.3244-3246, 2006.

T. J. Dengler, G. Szabo, and B. Sido, Prolonged allograft survival but no tolerance induction by modulating CD28 antibody JJ319 after high-responder rat heart transplantation, Transplantation, vol.67, pp.392-398, 1999.

F. Haspot, C. Sé-veno, and A. Dugast, Anti-CD28 antibody-induced kidney allograft tolerance related to tryptophan degradation and TCR class II B7 regulatory cells, Am J Transplant, vol.5, pp.2339-2348, 2005.

A. Dugast, T. Haudebourg, and F. Coulon, Myeloid-derived 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.

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

X. Tillou, N. Poirier, L. Bas-bernardet, and S. , Recombinant human C1-inhibitor prevents acute antibody-mediated rejection in alloimmunized baboons, Kidney Int, vol.78, pp.152-159, 2010.

G. Wieczorek, A. Asemissen, and F. Model, Quantitative DNA methylation analysis of FOXP3 as a new method for counting regulatory T cells in peripheral blood and solid tissue, Cancer Res, vol.69, pp.599-608, 2009.

N. Poirier, F. Maillet, and M. Barussaud, Acute humoral rejection of renal transplants in alloimmunized pigs, J Surg Res, vol.139, pp.261-268, 2007.

N. Poirier and G. Blancho, Recombinant human c1-inhibitor inhibits cytotoxicity induced by allo-and xenoantibodies, Transplant Proc, vol.40, pp.581-583, 2008.

M. Miyara, Y. Yoshioka, and A. Kitoh, Functional delineation and differentiation dynamics of human CD4þ T cells expressing the FoxP3 transcription factor, Immunity, vol.30, pp.899-911, 2009.

C. De-lafaille, M. A. Lafaille, and J. J. , Natural and adaptive foxp3þ regulatory T cells: More of the same or a division of labor?, Immunity, vol.30, pp.626-635, 2009.

S. K. Chauhan, D. R. Saban, H. K. Lee, and R. Dana, Levels of Foxp3 in regulatory T cells reflect their functional status in transplantation, J Immunol, vol.182, pp.148-153, 2009.

M. E. Himmel, K. G. Macdonald, R. V. Garcia, T. S. Steiner, and M. K. Levings, Heliosþ and HeliosÀ cells coexist within the natural FOXP3þ T regulatory cell subset in humans, J Immunol, vol.190, pp.2001-2008, 2013.

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

G. Suntharalingam, M. R. Perry, and S. Ward, Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412, N Engl J Med, vol.355, pp.1018-1028, 2006.

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

H. Ekberg, C. Bernasconi, and H. Tedesco-silva, Calcineurin inhibitor minimization in the symphony study: Observational results 3 years after transplantation, Am J Transplant, vol.9, pp.1876-1885, 2009.

D. Herná-ndez, M. R. Porrini, and E. , Randomized controlled study comparing reduced calcineurin inhibitors exposure versus standard cyclosporine-based immunosuppression, Transplantation, vol.84, pp.706-714, 2007.

R. S. Gaston, J. M. Cecka, and B. L. Kasiske, Evidence for antibodymediated injury as a major determinant of late kidney allograft failure, Transplantation, vol.90, pp.68-74, 2010.

J. Gloor, F. Cosio, D. J. Lager, and M. D. Stegall, The spectrum of antibodymediated renal allograft injury: Implications for treatment, Am J Transplant, vol.8, pp.1367-1373, 2008.

M. A. Linterman and C. G. Vinuesa, Signals that influence T follicular helper cell differentiation and function, Semin Immunopathol, vol.32, pp.183-196, 2010.

E. J. Kim, J. Kwun, and A. C. Gibby, Costimulation blockade alters germinal center responses and prevents antibody-mediated rejection, Am J Transplant, vol.14, pp.59-69, 2014.

L. Fong, S. S. Kwek, and S. O'brien, Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF, Cancer Res, vol.69, pp.609-615, 2009.

J. Yuan, S. Gnjatic, and H. Li, CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit, Proc Natl Acad Sci, vol.105, pp.20410-20415, 2008.

K. Wing, Y. Onishi, and P. Prieto-martin, CTLA-4 control over Foxp3þ regulatory T cell function, Science, vol.322, pp.271-275, 2008.

A. M. Thornton, P. E. Korty, and D. Q. Tran, Expression of Helios, an Ikaros transcription factor family member, differentiates thymicderived from peripherally induced Foxp3þ T regulatory cells, J Immunol, vol.184, pp.3433-3441, 2010.

E. M. Shevach and A. M. Thornton, tTregs, pTregs, and iTregs: Similarities and differences, Immunol Rev, vol.259, pp.88-102, 2014.

B. Salomon, D. J. Lenschow, and L. Rhee, B7/CD28 costimulation is essential for the homeostasis of the CD4þ CD25þ immunoregulatory T cells that control autoimmune diabetes, Immunity, vol.12, pp.431-440, 2000.

Y. Chen, S. Shen, B. K. Gorentla, J. Gao, and X. Zhong, Murine regulatory T cells contain hyperproliferative and death-prone subsets with differential ICOS expression, J Immunol, vol.188, pp.1698-1707, 2012.

K. Kretschmer, I. Apostolou, D. Hawiger, K. Khazaie, M. C. Nussenzweig et al., Inducing and expanding regulatory T cell populations by foreign antigen, Nat Immunol, vol.6, pp.1219-1227, 2005.

M. Belghith, J. A. Bluestone, S. Barriot, J. Mé, J. Bach et al., TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes, Nat Med, vol.9, pp.1202-1208, 2003.

S. Fu, N. Zhang, and A. C. Yopp, TGF-beta induces Foxp3þ Tregulatory cells from CD4þ CD25À precursors, Am J Transplant, vol.4, pp.1614-1627, 2004.

M. J. Benson, K. Pino-lagos, M. Rosemblatt, and R. J. Noelle, All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation, J Exp Med, vol.204, pp.1765-1774, 2007.

R. Li, N. Perez, S. Karumuthil-melethil, B. S. Prabhakar, M. J. Holterman et al., Enhanced engagement of CTLA-4 induces antigenspecific CD4þ CD25þ Foxp3þ and CD4þ CD25À TGF-beta 1þ adaptive regulatory T cells, J Immunol, vol.179, pp.5191-5203, 2007.

H. Chavez, S. Beaudreuil, and K. Abbed, 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, pp.243-248, 2007.

J. A. Bluestone, W. Liu, and J. M. Yabu, The effect of costimulatory and interleukin 2 receptor blockade on regulatory T cells in renal transplantation, Am J Transplant, vol.8, pp.2086-2096, 2008.