4.8 Article

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

期刊

FRONTIERS IN IMMUNOLOGY
卷 12, 期 -, 页码 -

出版社

FRONTIERS MEDIA SA
DOI: 10.3389/fimmu.2021.686439

关键词

chimeric antigen receptor; regulatory T cells; genome editing; transplantation; humanized mouse model; immune tolerance; HLA; Treg

资金

  1. NIDDK [UC4 DK116264, P30 DK063720]
  2. Juno Therapeutics
  3. JDRF [SRA-2019-776-S-B]
  4. Sean N. Parker Autoimmune Laboratory
  5. Swiss National Science Foundation [P300PB_174500]
  6. University Hospital of Geneva
  7. Jeffrey G. Klein Family Diabetes fellowship
  8. HIRN Emerging Leader in Type 1 Diabetes grant

向作者/读者索取更多资源

The study demonstrates that genome-engineered mono-antigen-specific A2-CAR Tregs can localize to HLA-A2-expressing grafts and exhibit antigen-dependent in vivo suppression. These Tregs do not impair the function of islets and can delay graft-versus-host disease. This approach may be applied towards developing precision Treg cell therapies for transplant tolerance.
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-zeta 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(+)TCR(deficient) 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(+)TCR(deficient) Tregs did not impair the function of these HLA-A2(+) islets, whereas similarly engineered A2-CAR(+)TCR(deficient)CD4(+) conventional T cells rejected the islets in less than 2 weeks. A2-CAR(+)TCR(deficient) 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.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据