Intratumoral administration of STING-activating nanovaccine enhances T cell immunotherapy

Xiaoyi Jiang, Jian Wang, Xichen Zheng, Zhida Liu, Xinyu Zhang, Yuwei Li, Jonathan Wilhelm, Jun Cao, Gang Huang, Jinlan Zhang, Baran Sumer, Jayanthi Lea, Zhigang Lu, Jinming Gao, Min Luo

Research output: Contribution to journalArticlepeer-review

22 Scopus citations

Abstract

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 IFNÎ 3-expressing CD8 + T cells from the periphery, and IFNγreciprocally stimulates CXCL9 expression in myeloid cells, resulting in positive feedback between myeloid-CXCL9 and T cell-IFNγto 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.

Original languageEnglish (US)
Article numbere003960
JournalJournal for ImmunoTherapy of Cancer
Volume10
Issue number5
DOIs
StatePublished - May 27 2022

Keywords

  • immunotherapy
  • lymphocytes, tumor-infiltrating
  • translational medical research
  • vaccination

ASJC Scopus subject areas

  • Immunology and Allergy
  • Immunology
  • Molecular Medicine
  • Oncology
  • Pharmacology
  • Cancer Research

Fingerprint

Dive into the research topics of 'Intratumoral administration of STING-activating nanovaccine enhances T cell immunotherapy'. Together they form a unique fingerprint.

Cite this