Faculty Opinions recommendation of A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity.

Author(s):  
Michael Farrar
2018 ◽  
Vol 90 ◽  
pp. 116-131 ◽  
Author(s):  
Stefanie Koristka ◽  
Alexandra Kegler ◽  
Ralf Bergmann ◽  
Claudia Arndt ◽  
Anja Feldmann ◽  
...  

2021 ◽  
Author(s):  
Yannick D. Muller ◽  
Leonardo M.R. Ferreira ◽  
Emilie Ronin ◽  
Patrick Ho ◽  
Vinh Nguyen ◽  
...  

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+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.


2018 ◽  
Vol 102 ◽  
pp. S742
Author(s):  
Katharina Zimmermann ◽  
Fatih Noyan ◽  
Michael Hust ◽  
Reinhard Schwinzer ◽  
Joachim Hundrieser ◽  
...  

2019 ◽  
Vol 103 ◽  
pp. 102289 ◽  
Author(s):  
Michel Tenspolde ◽  
Katharina Zimmermann ◽  
Leonie C. Weber ◽  
Martin Hapke ◽  
Maren Lieber ◽  
...  

Immunity ◽  
2011 ◽  
Vol 35 (1) ◽  
pp. 109-122 ◽  
Author(s):  
Dipica Haribhai ◽  
Jason B. Williams ◽  
Shuang Jia ◽  
Derek Nickerson ◽  
Erica G. Schmitt ◽  
...  

2016 ◽  
Vol 17 (3) ◽  
pp. 286-296 ◽  
Author(s):  
Yoon Park ◽  
Hyung-seung Jin ◽  
Justine Lopez ◽  
Jeeho Lee ◽  
Lujian Liao ◽  
...  

2016 ◽  
Vol 126 (4) ◽  
pp. 1413-1424 ◽  
Author(s):  
Katherine G. MacDonald ◽  
Romy E. Hoeppli ◽  
Qing Huang ◽  
Jana Gillies ◽  
Dan S. Luciani ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document