1. Academic Validation
  2. Self-assembly nanovaccine containing TLR7/8 agonist and STAT3 inhibitor enhances tumor immunotherapy by augmenting tumor-specific immune response

Self-assembly nanovaccine containing TLR7/8 agonist and STAT3 inhibitor enhances tumor immunotherapy by augmenting tumor-specific immune response

  • J Immunother Cancer. 2021 Aug;9(8):e003132. doi: 10.1136/jitc-2021-003132.
Lele Zhang 1 2 Jiacheng Huang 1 2 Xiaona Chen 1 2 Caixu Pan 1 2 Yong He 1 Rong Su 1 2 Danjing Guo 1 2 Shengyong Yin 1 2 Shuai Wang 1 2 Lin Zhou 1 2 Jianxiang Chen 3 4 Shusen Zheng 5 2 Yiting Qiao 5 2
Affiliations

Affiliations

  • 1 Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
  • 2 NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, People's Republic of China.
  • 3 College of Pharmacy, Hangzhou Normal University Hangzhou School of Medicine, Hangzhou, Zhejiang, China [email protected] [email protected] [email protected].
  • 4 Department of Hepatology, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China.
  • 5 Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China [email protected] [email protected] [email protected].
Abstract

Background: Cancer vaccines are a promising strategy for Cancer Immunotherapy. Cancer vaccines elicits a specific cytotoxic immune response to tumor antigens. However, the efficacy of traditional peptide-based Cancer vaccines is limited due to the inefficient delivery of antigens and adjuvants to dendritic cells (DCs). Therefore, it is necessary to develop a novel rationally designed Cancer vaccine to maximize its desired effects.

Methods: A Self-assembling Vehicle-free Multi-component Antitumor nanoVaccine (SVMAV) was constructed by using an unsaturated fatty acid docosahexaenoic acid (DHA)-conjugated antigen and R848 (a Toll-like Receptor 7/8 agonist) to encapsulate stattic (a signal transducer and activator of transcription 3 inhibitor). The characteristics of SVMAV were investigated. The ability of SVMAV to promote DC functions was examined by in vitro analysis. The antitumor effects of SVMAV and its combination with antiprogrammed cell death protein 1 antibody (aPD-1) were also investigated in vivo. The potential application of SVMAV for neoantigen-targeted, personalized Cancer vaccines was examined in an orthotopic hepatocellular carcinoma model.

Results: The obtained SVMAV efficiently migrated into lymph nodes and primed CD8+ T cells for exert neoantigen-specific killing by promoting the antigen uptake by DCs, stimulating DC maturation, and enhancing antigen cross-presentation, due to the simultaneous delivery of the antigen, R848 and stattic. SVMAV could not only yield a robust antitumor effect for primary melanoma allografts, but also exert a protective effect for lung metastases. Moreover, combination treatment of SVMAV and aPD-1 exerted synergistic antitumor activity and extended the survival duration of melanoma-bearing mice. Notably, a cell line-specific neoantigen-based SVMAV was designed according to predicted neoantigens for Hepa1-6 cells to examine the potential application of SVMAV for personalized Cancer vaccine. Encouragingly, neoantigen-specific SVMAV achieved stronger antitumor activity than aPD-1 in an orthotopic hepatocellular Cancer model established with Hepa1-6 cells.

Conclusions: In summary, this study offers an efficient codelivery platform for neoantigens and immunoregulatory compounds to enhance immune responses during Cancer immune therapy.

Keywords

antigens; dendritic cells; immunogenicity; immunotherapy; neoplasm; vaccine.

Figures
Products