Your browser doesn't support javascript.
loading
Pilot Study of Delayed ICOS/ICOS-L Blockade With αCD40 to Modulate Pathogenic Alloimmunity in a Primate Cardiac Allograft Model.
O'Neill, Natalie A; Zhang, Tianshu; Braileanu, Gheorghe; Cheng, Xiangfei; Hershfeld, Alena; Sun, Wenji; Reimann, Keith A; Dahi, Sia; Kubicki, Natalia; Hassanein, Wessam; Laird, Christopher; Cimeno, Arielle; Azimzadeh, Agnes M; Pierson, Richard N.
Afiliação
  • O'Neill NA; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Zhang T; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Braileanu G; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Cheng X; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Hershfeld A; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Sun W; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Reimann KA; MassBiologics, University of Massachusetts Medical School, Boston, MA.
  • Dahi S; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Kubicki N; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Hassanein W; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Laird C; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Cimeno A; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
  • Azimzadeh AM; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.
Transplant Direct ; 4(2): e344, 2018 Feb.
Article em En | MEDLINE | ID: mdl-29464205
ABSTRACT

BACKGROUND:

Inducible costimulator (ICOS) is rapidly upregulated with T-cell stimulation and may represent an escape pathway for T-cell costimulation in the setting of CD40/CD154 costimulation blockade. Induction treatment exhibited no efficacy in a primate renal allograft model, but rodent transplant models suggest that the addition of delayed ICOS/ICOS-L blockade may prolong allograft survival and prevent chronic rejection. Here, we ask whether ICOS-Ig treatment, timed to anticipate ICOS upregulation, prolongs NHP cardiac allograft survival or attenuates pathogenic alloimmunity.

METHODS:

Cynomolgus monkey heterotopic cardiac allograft recipients were treated with αCD40 (2C10R4, d0-90) either alone or with the addition of delayed ICOS-Ig (d63-110).

RESULTS:

Median allograft survival was similar between ICOS-Ig + αCD40 (120 days, 120-125 days) and αCD40 (124 days, 89-178 days) treated animals, and delayed ICOS-Ig treatment did not prevent allograft rejection in animals with complete CD40 receptor coverage. Although CD4+ TEM cells were decreased in peripheral blood (115 ± 24) and mLNs (49 ± 1.9%) during ICOS-Ig treatment compared with monotherapy (214 ± 27%, P = 0.01; 72 ± 9.9%, P = 0.01, respectively), acute and chronic rejection scores and kinetics of alloAb elaboration were similar between groups.

CONCLUSIONS:

Delayed ICOS-Ig treatment with the reagent tested is probably ineffective in modulating pathogenic primate alloimmunity in this model.

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Revista: Transplant Direct Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Moldávia

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Revista: Transplant Direct Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Moldávia