Cell Mol Immunol | 沉默SIRPα可增强CAR-M在实体瘤中的抗肿瘤疗效(第四军医大学)

文摘   2024-10-09 23:55   上海  

Paper Reading

01

Silencing of SIRPα enhances the antitumor efficacy of CAR-M in solid tumors 


Zhang H, et al.

Cell Mol Immunol. 2024

    

    The potential of macrophage-mediated phagocytosis as a cancer treatment is promising. Blocking the CD47-SIRPα interaction with a CD47-specific antibody significantly enhances macrophage phagocytosis. However, concerns regarding their toxicity to nontumor cells remain substantial. Here, the authors engineered chimeric antigen receptor macrophages (CAR-Ms) by fusing a humanized single-chain variable fragment with FcγRIIa and integrating short hairpin RNA to silence SIRPα, thereby disrupting the CD47-SIRPα signaling pathway. These modified CAR-shSIRPα-M cells exhibited an M1-like phenotype, superior phagocytic function, substantial cytotoxic effects on HER2-positive tumor cells, and the ability to eliminate patient-derived organoids. In vivo, CAR-M cells significantly inhibited tumor growth and prolonged survival in tumor-bearing mice. Notably, CAR-shSIRPα-M cells enhanced cytotoxic T-cell infiltration into tumors, thereby enhancing the antitumor response in both the humanized immune system mouse model and immunocompetent mice. Mechanistically, SIRPα inhibition activated inflammatory pathways and the cGAS-STING signaling cascade in CAR-M cells, leading to increased production of proinflammatory cytokines, reactive oxygen species, and nitric oxide, thereby enhancing their antitumor effects. These findings underscore the potential of SIRPα inhibition as a novel strategy to increase the antitumor efficacy of CAR-M cells in cancer immunotherapy, particularly against solid tumors.

DOI: 10.1038/s41423-024-01220-3

02

GLUT1 overexpression in CAR-T cells induces metabolic reprogramming and enhances potency 


Guerrero J. A, et al.
Nat Commun. 2024


    The intensive nutrient requirements needed to sustain T cell activation and proliferation, combined with competition for nutrients within the tumor microenvironment, raise the prospect that glucose availability may limit CAR-T cell function. Here, the authors seek to test the hypothesis that stable overexpression (OE) of the glucose transporter GLUT1 in primary human CAR-T cells would improve their function and antitumor potency. It was observed that GLUT1OE in CAR-T cells increases glucose consumption, glycolysis, glycolytic reserve, and oxidative phosphorylation, and these effects are associated with decreased T cell exhaustion and increased Th17 differentiation. GLUT1OE also induces broad metabolic reprogramming associated with increased glutathione-mediated resistance to reactive oxygen species, and increased inosine accumulation. When challenged with tumors, GLUT1OE CAR-T cells secrete more proinflammatory cytokines and show enhanced cytotoxicity in vitro, and demonstrate superior tumor control and persistence in mouse models. Collectively, these findings support a paradigm wherein glucose availability is rate limiting for effector CAR-T cell function and demonstrate that enhancing glucose availability via GLUT1OE could augment antitumor immune function.

DOI: 10.1038/s41467-024-52666-y




相关阅读:
1. Cell Metab | 应激通过亚精胺介导的I型干扰素下调触发肠易激综合征伴腹泻(中山大学附属第三医院)

2. Sci Immunol | NF-κB维持T细胞活化后 X 染色体失活 (美国国立卫生研究院)


END


关注我们获取更多免疫学文献资讯

Editor & Reviewer: Yanwen Zhu


李斌课题组
文献阅读共享
 最新文章