4.7 Article

Novel phosphatidylserine-binding molecule enhances antitumor T-cell responses by targeting immunosuppressive exosomes in human tumor microenvironments

期刊

出版社

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2021-003148

关键词

immunotherapy; melanoma; drug evaluation; preclinical; T-lymphocytes; tumor microenvironment

资金

  1. National Institutes of Health [R43 CA224602, R43 CA257721-01A1, CA034196, R21 CA205794, R01 CA204261, HL-70227, R50CA2111108]
  2. Empire State Development
  3. University at Buffalo Center for Advanced Technology in Big Data and Health Sciences grant
  4. Roswell Park Cancer Center Support grant [P30CA016056]

向作者/读者索取更多资源

The study demonstrated that ExoBlock has high avidity for binding to PS, significantly blocking the immunosuppressive activity of human ovarian tumor and melanoma-associated exosomes in vitro. Additionally, ExoBlock enhanced T cell-mediated tumor suppression in both the X-mouse and OTX models, leading to reduction in tumor burden, metastasis, and circulating PS+ exosomes in tumor-bearing mice. These results suggest that targeting exosomal PS in TMEs using ExoBlock is a promising strategy to enhance antitumor T-cell responses.
Background The human tumor microenvironment (TME) is a complex and dynamic milieu of diverse acellular and cellular components, creating an immunosuppressive environment, which contributes to tumor progression. We have previously shown that phosphatidylserine (PS) expressed on the surface of exosomes isolated from human TMEs is causally linked to T-cell immunosuppression, representing a potential immunotherapeutic target. In this study, we investigated the effect of ExoBlock, a novel PS-binding molecule, on T-cell responses in the TME. Methods We designed and synthesized a new compound, (ZnDPA)(6)-DP-15K, a multivalent PS binder named ExoBlock. The PS-binding avidity of ExoBlock was tested using an in vitro competition assay. The ability of this molecule to reverse exosome-mediated immunosuppression in vitro was tested using human T-cell activation assays. The in vivo therapeutic efficacy of ExoBlock was then tested in two different human tumor xenograft models, the melanoma-based xenomimetic (X-)mouse model, and the ovarian tumor-based omental tumor xenograft (OTX) model. Results ExoBlock was able to bind PS with high avidity and was found to consistently and significantly block the immunosuppressive activity of human ovarian tumor and melanoma-associated exosomes in vitro. ExoBlock was also able to significantly enhance T cell-mediated tumor suppression in vivo in both the X-mouse and the OTX model. In the X-mouse model, ExoBlock suppressed tumor recurrence in a T cell-dependent manner. In the OTX model, ExoBlock treatment resulted in an increase in the number as well as function of CD4 and CD8 T cells in the TME, which was associated with a reduction in tumor burden and metastasis, as well as in the number of circulating PS+ exosomes in tumor-bearing mice. Conclusion Our results establish that targeting exosomal PS in TMEs with ExoBlock represents a promising strategy to enhance antitumor T-cell responses.

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

4.7
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