Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor activity

B Prinzing, CC Zebley, CT Petersen, Y Fan… - Science translational …, 2021 - science.org
B Prinzing, CC Zebley, CT Petersen, Y Fan, AA Anido, Z Yi, P Nguyen, H Houke, M Bell…
Science translational medicine, 2021science.org
Chimeric antigen receptor (CAR) T cell therapy is revolutionizing cancer immunotherapy for
patients with B cell malignancies and is now being developed for solid tumors and chronic
viral infections. Although clinical trials have demonstrated the curative potential of CAR T
cell therapy, a substantial and well-established limitation is the heightened contraction and
transient persistence of CAR T cells during prolonged antigen exposure. The underlying
mechanism (s) for this dysfunctional state, often termed CAR T cell exhaustion, remains …
Chimeric antigen receptor (CAR) T cell therapy is revolutionizing cancer immunotherapy for patients with B cell malignancies and is now being developed for solid tumors and chronic viral infections. Although clinical trials have demonstrated the curative potential of CAR T cell therapy, a substantial and well-established limitation is the heightened contraction and transient persistence of CAR T cells during prolonged antigen exposure. The underlying mechanism(s) for this dysfunctional state, often termed CAR T cell exhaustion, remains poorly defined. Here, we report that exhaustion of human CAR T cells occurs through an epigenetic repression of the T cell’s multipotent developmental potential. Deletion of the de novo DNA methyltransferase 3 alpha (DNMT3A) in T cells expressing first- or second-generation CARs universally preserved the cells’ ability to proliferate and mount an antitumor response during prolonged tumor exposure. The increased functionality of the exhaustion-resistant DNMT3A knockout CAR T cells was coupled to an up-regulation of interleukin-10, and genome-wide DNA methylation profiling defined an atlas of genes targeted for epigenetic silencing. This atlas provides a molecular definition of CAR T cell exhaustion, which includes many transcriptional regulators that limit the “stemness” of immune cells, including CD28, CCR7, TCF7, and LEF1. Last, we demonstrate that this epigenetically regulated multipotency program is firmly coupled to the clinical outcome of prior CAR T cell therapies. These data document the critical role epigenetic mechanisms play in limiting the fate potential of human T cells and provide a road map for leveraging this information for improving CAR T cell efficacy.
AAAS