Unknown

Dataset Information

0

Deletion of Cbl-b inhibits CD8+ T-cell exhaustion and promotes CAR T-cell function.


ABSTRACT:

Background

Chimeric antigen receptor (CAR) T-cell therapy is an emerging option for cancer treatment, but its efficacy is limited, especially in solid tumors. This is partly because the CAR T cells become dysfunctional and exhausted in the tumor microenvironment. However, the key pathways responsible for impaired function of exhausted cells remain unclear, which is essential to overcome CAR T-cell exhaustion.

Methods

Analysis of RNA-sequencing data from CD8+ tumor-infiltrating lymphocytes (TILs) led to identification of Cbl-b as a potential target. The sequencing data were validated using a syngeneic MC38 colon cancer model. To analyze the in vivo role of Cbl-b in T-cell exhaustion, tumor growth, % PD1+Tim3+ cells, and expression of effector cytokines were analyzed in cbl-b+/+ and cbl-b-/- mice. To evaluate the therapeutic potential of Cbl-b depletion, we generated a new CAR construct, hCEAscFv-CD28-CD3?.GFP, that recognizes human carcinoembryonic antigen (CEA). cbl-b+/+ and cbl-b-/- CEA-CAR T cells were generated by retroviral transduction. Rag -/- mice bearing MC38-CEA cells were injected with cbl-b+/+ and cbl-b-/- ; CEA-CAR T cells, tumor growth, % PD1+Tim3+ cells and expression of effector cytokines were analyzed.

Results

Our results show that the E3 ubiquitin ligase Cbl-b is upregulated in exhausted (PD1+Tim3+) CD8+ TILs. CRISPR-Cas9-mediated inhibition of Cbl-b restores the effector function of exhausted CD8+ TILs. Importantly, the reduced growth of syngeneic MC38 tumors in cbl-b-/- mice was associated with a marked reduction of PD1+Tim3+ CD8+ TILs. Depletion of Cbl-b inhibited CAR T-cell exhaustion, resulting in reduced MC38-CEA tumor growth, reduced PD1+Tim3+ cells and increased expression of interferon gamma, tumor necrosis factor alpha, and increased tumor cell killing.

Conclusion

Our studies demonstrate that deficiency of Cbl-b overcomes endogenous CD8+ T-cell exhaustion, and deletion of Cbl-b in CAR T cells renders them resistant to exhaustion. Our results could facilitate the development of efficient CAR T-cell therapy for solid tumors by targeting Cbl-b.

SUBMITTER: Kumar J 

PROVIDER: S-EPMC7813298 | biostudies-literature | 2021 Jan

REPOSITORIES: biostudies-literature

altmetric image

Publications

Deletion of Cbl-b inhibits CD8<sup>+</sup> T-cell exhaustion and promotes CAR T-cell function.

Kumar Jitendra J   Kumar Ritesh R   Kumar Singh Amir A   Tsakem Elviche L EL   Kathania Mahesh M   Riese Matthew J MJ   Theiss Arianne L AL   Davila Marco L ML   Venuprasad K K  

Journal for immunotherapy of cancer 20210101 1


<h4>Background</h4>Chimeric antigen receptor (CAR) T-cell therapy is an emerging option for cancer treatment, but its efficacy is limited, especially in solid tumors. This is partly because the CAR T cells become dysfunctional and exhausted in the tumor microenvironment. However, the key pathways responsible for impaired function of exhausted cells remain unclear, which is essential to overcome CAR T-cell exhaustion.<h4>Methods</h4>Analysis of RNA-sequencing data from CD8<sup>+</sup> tumor-infil  ...[more]

Similar Datasets

| S-EPMC7292629 | biostudies-literature
| S-EPMC7061417 | biostudies-literature
| S-EPMC7710757 | biostudies-literature
| S-EPMC6706391 | biostudies-literature
| S-EPMC6921985 | biostudies-literature
| S-EPMC8369465 | biostudies-literature
| S-EPMC8774026 | biostudies-literature
| S-EPMC5380094 | biostudies-literature
| S-EPMC8600539 | biostudies-literature
| S-EPMC5512309 | biostudies-literature