1. Academic Validation
  2. Targeting sphingosine 1-phosphate receptor 3 inhibits T-cell exhaustion and regulates recruitment of proinflammatory macrophages to improve antitumor efficacy of CAR-T cells against solid tumor

Targeting sphingosine 1-phosphate receptor 3 inhibits T-cell exhaustion and regulates recruitment of proinflammatory macrophages to improve antitumor efficacy of CAR-T cells against solid tumor

  • J Immunother Cancer. 2023 Aug;11(8):e006343. doi: 10.1136/jitc-2022-006343.
Ge Gao # 1 2 Weiting Liao # 3 Pei Shu # 1 2 Qizhi Ma 2 Xia He 1 2 4 Benxia Zhang 1 2 Diyuan Qin 1 2 Yongsheng Wang 5
Affiliations

Affiliations

  • 1 Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China.
  • 2 Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Sichuan University West China Hospital, Chengdu, Sichuan, China.
  • 3 Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China.
  • 4 Department of Clinical Research Management, Sichuan University West China Hospital, Chengdu,Sichuan, China.
  • 5 Division of Thoracic Tumor Multimodality Treatment, Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China wangys@scu.edu.cn.
  • # Contributed equally.
Abstract

Backgrounds: Chimeric antigen receptor (CAR)-modified T cells (CAR-T) are limited in solid tumors due to the hostile tumor microenvironment (TME). Combination therapy could be a promising approach to overcome this obstacle. Recent studies have shown that sphingosine 1-phosphate receptor (S1PR)3 has tremendous potential in regulating the immune environment. However, the functional significance of S1PR3 in T-cell-based immunotherapies and the molecular mechanisms have not been fully understood.

Methods: Here, we studied the combination of EpCAM-specific CAR T-cell therapy with pharmacological blockade of S1PR3 against solid tumor. We have applied RNA sequencing, flow cytometry, ELISA, cellular/molecular immunological technology, and mouse models of solid cancers.

Results: Our study provided evidence that S1PR3 high expression is positively associated with resistance to programmed cell death protein-1 (PD-1)-based immunotherapy and increased T-cell exhaustion. In addition, pharmacological inhibition of S1PR3 improves the efficacy of anti-PD-1 therapy. Next, we explored the possible combination of S1PR3 Antagonist with murine EpCAM-targeted CAR-T cells in immunocompetent mouse models of breast Cancer and colon Cancer. The results indicated that the S1PR3 Antagonist could significantly enhance the efficacy of murine EpCAM CAR-T cells in vitro and in vivo. Mechanistically, the S1PR3 Antagonist improved CAR-T cell activation, regulated the central memory phenotype, and reduced CAR-T cell exhaustion in vitro. Targeting S1PR3 was shown to remodel the TME through the recruitment of proinflammatory macrophages by promoting macrophage activation and proinflammatory phenotype polarization, resulting in improved CAR-T cell infiltration and amplified recruitment of CD8+T cells.

Conclusions: This work demonstrated targeting S1PR3 could increase the antitumor activities of CAR-T cell therapy at least partially by inhibiting T-cell exhaustion and remodeling the TME through the recruitment of proinflammatory macrophages. These findings provided additional rationale for combining S1PR3 inhibitor with CAR-T cells for the treatment of solid tumor.

Keywords

immunotherapy; lymphocytes, tumor-infiltrating; macrophages; receptors, chimeric antigen; tumor microenvironment.

Figures
Products