4.6 Article

M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance

期刊

ONCOIMMUNOLOGY
卷 12, 期 1, 页码 -

出版社

TAYLOR & FRANCIS INC
DOI: 10.1080/2162402X.2023.2210959

关键词

apolipoprotein E; exosomes; immune checkpoint blockade therapy; immunogenicity; macrophages

向作者/读者索取更多资源

M2-like tumor-associated macrophages undermine immune checkpoint blockade therapy by secreting exosomes that decrease tumor immunogenicity through downregulating MHC-I expression. Sensitizing therapy efficacy can be achieved by enhancing ATPase activity of BiP using ApoE ligand EZ-482 to boost tumor immunogenicity. Therefore, ApoE may serve as a predictive factor and potential therapeutic target for ICB resistance in M2-enriched cancer patients.
T-cell-based immune checkpoint blockade therapy (ICB) can be undermined by local immunosuppressive M2-like tumor-associated macrophages (TAMs). However, modulating macrophages has proved difficult as the molecular and functional features of M2-TAMs on tumor growth are still uncertain. Here we reported that immunosuppressive M2 macrophages render cancer cells resistant to CD8(+) T-cell-dependent tumor-killing refractory ICB efficacy by secreting exosomes. Proteomics and functional studies revealed that M2 macrophage-derived exosome (M2-exo) transmitted apolipoprotein E (ApoE) to cancer cells conferring ICB resistance by downregulated MHC-I expression curbing tumor intrinsic immunogenicity. Mechanistically, M2 exosomal ApoE diminished the tumor-intrinsic ATPase activity of binding immunoglobulin protein (BiP) to decrease tumor MHC-I expression. Sensitizing ICB efficacy can be achieved by the administration of ApoE ligand, EZ-482, enhancing ATPase activity of BiP to boost tumor-intrinsic immunogenicity. Therefore, ApoE may serve as a predictor and a potential therapeutic target for ICB resistance in M2-TAMs-enriched cancer patients. Collectively, our findings signify that the exosome-mediated transfer of functional ApoE from M2 macrophages to the tumor cells confers ICB resistance. Our findings also provide a preclinical rationale for treating M2-enriched tumors with ApoE ligand, EZ-482, to restore sensitivity to ICB immunotherapy.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据