4.8 Article

RASA2 ablation in T cells boosts antigen sensitivity and long-term function

Journal

NATURE
Volume 609, Issue 7925, Pages 174-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41586-022-05126-w

Keywords

-

Funding

  1. NIH/NCI K08 [1K08CA252605-01]
  2. Burroughs Wellcome Fund Career Award for Medical Scientists
  3. Lydia Preisler Shorenstein Donor Advised Fund
  4. Care-for-Rare Foundation
  5. German Research Foundation (DFG)
  6. NIH [R35NS105068]
  7. Parker Institute for Cancer Immunotherapy
  8. Grand Multiple Myeloma Translational Initiative
  9. NIH/NCI [R01NS106379-02, R01CA173750, R01NS121249, K99CA256262]
  10. Assisi foundation
  11. Damon Runyon Cancer Research Foundation Physician-Scientist Training Award
  12. American Lebanese Syrian Associated Charities (ALSAC)
  13. St Jude Comprehensive Cancer Center (SJCCC) Neurobiology and Brain Tumor Program (NBTP) [P30CA021765]
  14. NIH Shared Instrumentation Grant [1S10OD010786-01]
  15. National Cancer Institute of the National Institutes of Health [P30CA082103]
  16. Parker Institute for Cancer Immunotherapy, PICI
  17. Simons Foundation
  18. Cancer Research Institute (CRI) Lloyd J. Old STAR grant
  19. Innovative Genomics Institute

Ask authors/readers for more resources

This study identifies RASA2 as a potential target gene that can enhance the efficacy of T cell therapies for cancer treatment. Ablation of RASA2 increases the activation and cytolytic activity of T cells, leading to improved cancer cell killing. Furthermore, RASA2-deficient CAR T cells have a competitive advantage in the bone marrow and extended the survival of mice in preclinical models. These findings highlight the importance of targeting RASA2 in enhancing T cell therapies for cancer treatment.
The efficacy of adoptive T cell therapies for cancer treatment can be limited by suppressive signals from both extrinsic factors and intrinsic inhibitory checkpoints(1,2). Targeted gene editing has the potential to overcome these limitations and enhance T cell therapeutic function(3-10). Here we performed multiple genome-wide CRISPR knock-out screens under different immunosuppressive conditions to identify genes that can be targeted to prevent T cell dysfunction. These screens converged on RASA2, a RAS GTPase-activating protein (RasGAP) that we identify as a signalling checkpoint in human T cells, which is downregulated upon acute T cell receptor stimulation and can increase gradually with chronic antigen exposure. RASA2 ablation enhanced MAPK signalling and chimeric antigen receptor (CAR) T cell cytolytic activity in response to target antigen. Repeated tumour antigen stimulations in vitro revealed that RASA2-deficient T cells show increased activation, cytokine production and metabolic activity compared with control cells, and show a marked advantage in persistent cancer cell killing. RASA2-knockout CAR T cells had a competitive fitness advantage over control cells in the bone marrow in a mouse model of leukaemia. Ablation of RASA2 in multiple preclinical models of T cell receptor and CAR T cell therapies prolonged survival in mice xenografted with either liquid or solid tumours. Together, our findings highlight RASA2 as a promising target to enhance both persistence and effector function in T cell therapies for cancer treatment.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available