Frontiers in Immunology (Sep 2021)

Precision Engineering of an Anti-HLA-A2 Chimeric Antigen Receptor in Regulatory T Cells for Transplant Immune Tolerance

  • Yannick D. Muller,
  • Yannick D. Muller,
  • Leonardo M. R. Ferreira,
  • Leonardo M. R. Ferreira,
  • Leonardo M. R. Ferreira,
  • Emilie Ronin,
  • Emilie Ronin,
  • Patrick Ho,
  • Patrick Ho,
  • Patrick Ho,
  • Vinh Nguyen,
  • Vinh Nguyen,
  • Gaetano Faleo,
  • Gaetano Faleo,
  • Yu Zhou,
  • Karim Lee,
  • Kevin K. Leung,
  • Nikolaos Skartsis,
  • Nikolaos Skartsis,
  • Anupurna M. Kaul,
  • Arend Mulder,
  • Frans H. J. Claas,
  • James A. Wells,
  • Jeffrey A. Bluestone,
  • Jeffrey A. Bluestone,
  • Qizhi Tang,
  • Qizhi Tang

DOI
https://doi.org/10.3389/fimmu.2021.686439
Journal volume & issue
Vol. 12

Abstract

Read online

Infusion of regulatory T cells (Tregs) engineered with a chimeric antigen receptor (CAR) targeting donor-derived human leukocyte antigen (HLA) is a promising strategy to promote transplant tolerance. Here, we describe an anti-HLA-A2 CAR (A2-CAR) generated by grafting the complementarity-determining regions (CDRs) of a human monoclonal anti-HLA-A2 antibody into the framework regions of the Herceptin 4D5 single-chain variable fragment and fusing it with a CD28-ζ signaling domain. The CDR-grafted A2-CAR maintained the specificity of the original antibody. We then generated HLA-A2 mono-specific human CAR Tregs either by deleting the endogenous T-cell receptor (TCR) via CRISPR/Cas9 and introducing the A2-CAR using lentiviral transduction or by directly integrating the CAR construct into the TCR alpha constant locus using homology-directed repair. These A2-CAR+TCRdeficient human Tregs maintained both Treg phenotype and function in vitro. Moreover, they selectively accumulated in HLA-A2-expressing islets transplanted from either HLA-A2 transgenic mice or deceased human donors. A2-CAR+TCRdeficient Tregs did not impair the function of these HLA-A2+ islets, whereas similarly engineered A2-CAR+TCRdeficientCD4+ conventional T cells rejected the islets in less than 2 weeks. A2-CAR+TCRdeficient Tregs delayed graft-versus-host disease only in the presence of HLA-A2, expressed either by co-transferred peripheral blood mononuclear cells or by the recipient mice. Altogether, we demonstrate that genome-engineered mono-antigen-specific A2-CAR Tregs localize to HLA-A2-expressing grafts and exhibit antigen-dependent in vivo suppression, independent of TCR expression. These approaches may be applied towards developing precision Treg cell therapies for transplant tolerance.

Keywords