Abstract

As recent advancements in the chimeric antigen receptor-T cells have revolutionized the way blood cancers are handled, potential benefits from producing off-the-shelf, standardized immune cells entail the need for development of allogeneic immune cell therapy. However, host rejection driven by HLA disparity in adoptively transferred allogeneic T cells remains a key obstacle to the universal donor T cell therapy. To evade donor HLA-mediated immune rejection, we attempted to eliminate T cell’s HLA through the CRISPR/Cas9 gene editing system. First, we screened 60 gRNAs targeting B2M and multiple sets of gRNA each targeting α chains of HLA-II (DPA, DQA and DRA, respectively) using web-based design tools, and identified specific gRNA sequences highly efficient for target deletion without carrying off-target effects. Multiplex genome editing of primary human T cells achieved by the newly discovered gRNAs yielded HLA-I- or HLA-I/II-deficient T cells that were phenotypically unaltered and functionally intact. The overnight mixed lymphocyte reactions demonstrated the HLA-I-negative cells induced decreased production of IFN-γ and TNF-α in alloreactive T cells, and deficiency of HLA-I/II in T cells further dampened the inflammatory responses. Taken together, our approach will provide an efficacious pathway toward the universal donor cell generation by manipulating HLA expression in therapeutic T cells.

Highlights

  • The emergence of chimeric antigen receptor (CAR)-T cell therapy has changed the paradigm of cancer immunotherapy by virtue of its durable remission with manageable toxicity profile

  • In vitro mixed lymphocyte reactions revealed that alloresponses in responder cells were dampened against HLA-I/II-negative T cells compared to HLA-I-negative cells, solidifying a conceptual framework that narrates the role of HLA-II plays in infused, therapeutic allogeneic cells during host-mediated rejection

  • Off-the-shelf CAR-T cells produced from healthy allogeneic donor T cells with the “fittest” phenotypes can increase the likelihood of therapeutic response and overcome the barriers of the autologous CAR-T cell therapy

Read more

Summary

Introduction

The emergence of chimeric antigen receptor (CAR)-T cell therapy has changed the paradigm of cancer immunotherapy by virtue of its durable remission with manageable toxicity profile. A recent report in a trial of autologous CAR-T cell therapy documented the unintentional transduction of a single leukemic B cell with anti-CD19 CAR during manufacturing and its product masking the target antigen, thereby escaping CAR-T recognition and resisting the ­therapy[8]. These limitations could be overcome by employing allogeneic T cells obtained from “fitted” healthy donors as a source of universal CAR-T cell production. In vitro mixed lymphocyte reactions revealed that alloresponses in responder cells were dampened against HLA-I/II-negative T cells compared to HLA-I-negative cells, solidifying a conceptual framework that narrates the role of HLA-II plays in infused, therapeutic allogeneic cells during host-mediated rejection

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call