4.7 Article

Intratumoral administration of STING-activating nanovaccine enhances T cell immunotherapy

期刊

出版社

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2021-003960

关键词

vaccination; lymphocytes; tumor-infiltrating; translational medical research; immunotherapy

资金

  1. National Natural Science Foundation of China [81873922, 81873438]
  2. Shanghai Yang Fan Program [19YF1403900]
  3. National Institutes of Health [R01 CA192221, U01CA218422]
  4. Mendelson-Young Endowment in Cancer Therapeutics

向作者/读者索取更多资源

Our study found that intratumoral administration of PC7A nanovaccine achieved stronger antitumor immunity and efficacy compared to subcutaneous injection, primarily dependent on CD8(+) cytotoxic T cells. Mechanistic investigation revealed that intratumoral delivery significantly increased the infiltration of antigen-specific T cells in tumors, despite lower systemic T cell levels compared to subcutaneous injection.
Background Cancer vaccines are able to achieve tumor-specific immune editing in early-phase clinical trials. However, the infiltration of cytotoxic T cells into immune-deserted tumors is still a major limiting factor. An optimized vaccine approach to induce antigen-specific T cells that can perform robust tumor infiltration is important to accelerate their clinical translation. We previously developed a STING-activating PC7A nanovaccine that produces a strong anti-tumor T cell response on subcutaneous injection. This study systematically investigated the impact of administration methods on the performance of nanovaccines. Methods Tumor growth inhibition by intratumoral delivery and subcutaneous delivery of nanovaccine was investigated in TC-1 human papillomavirus-induced cancer model and B16-OVA melanoma model. Nanovaccine distribution in vivo was detected by clinical camera imaging, systemic T cell activation and tumor infiltration were tested by in vivo cytotoxicity killing assay and flow cytometry. For mechanism analysis, T cell recruitment was investigated by in vivo migration blocking assay, multiplex chemokine array, flow cytometry, RT-qPCR, chemotaxis assay and gene knockout mice. Results Nanovaccine administration was found to alter T cell production and infiltration in tumors. Intratumoral delivery of nanovaccines displayed superior antitumor effects in multiple tumor models compared with subcutaneous delivery. Mechanistic investigation revealed that intratumoral administration of the nanovaccine significantly increased the infiltration of antigen-specific T cells in TC-1 tumors, despite the lower systemic levels of T cells compared with subcutaneous injection. The inhibition of tumor growth by nanovaccines is primarily dependent on CD8(+) cytotoxic T cells. Nanovaccine accumulation in tumors upregulates CXCL9 expression in myeloid cells in a STING dependent manner, leading to increased recruitment of IFN gamma-expressing CD8(+) T cells from the periphery, and IFN gamma reciprocally stimulates CXCL9 expression in myeloid cells, resulting in positive feedback between myeloid-CXCL9 and T cell-IFN gamma to promote T cell recruitment. However, the STING agonist alone could not sustain this effect in the presence of a systemic deficiency in antigen-specific T cells. Conclusions Our results demonstrate that intratumoral administration of PC7A nanovaccine achieved stronger antitumor immunity and efficacy over subcutaneous injection. These data suggest intratumoral administration should be included in the therapeutic design in the clinical use of nanovaccine.

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