Intratumoral expression of interleukin 23 variants using oncolytic vaccinia virus elicit potent antitumor effects on multiple tumor models via tumor microenvironment modulation

Theranostics. 2021 May 3;11(14):6668-6681. doi: 10.7150/thno.56494. eCollection 2021.

Abstract

Background: Newly emerging cancer immunotherapy has led to significant progress in cancer treatment; however, its efficacy is limited in solid tumors since the majority of them are "cold" tumors. Oncolytic viruses, especially when properly armed, can directly target tumor cells and indirectly modulate the tumor microenvironment (TME), resulting in "hot" tumors. These viruses can be applied as a cancer immunotherapy approach either alone or in combination with other cancer immunotherapies. Cytokines are good candidates to arm oncolytic viruses. IL-23, an IL-12 cytokine family member, plays many roles in cancer immunity. Here, we used oncolytic vaccinia viruses to deliver IL-23 variants into the tumor bed and explored their activity in cancer treatment on multiple tumor models. Methods: Oncolytic vaccinia viruses expressing IL-23 variants were generated by homologue recombination. The characteristics of these viruses were in vitro evaluated by RT-qPCR, ELISA, flow cytometry and cytotoxicity assay. The antitumor effects of these viruses were evaluated on multiple tumor models in vivo and the mechanisms were investigated by RT-qPCR and flow cytometry. Results: IL-23 prolonged viral persistence, probably mediated by up-regulated IL-10. The sustainable IL-23 expression and viral oncolysis elevated the expression of Th1 chemokines and antitumor factors such as IFN-γ, TNF-α, Perforin, IL-2, Granzyme B and activated T cells in the TME, transforming the TME to be more conducive to antitumor immunity. This leads to a systemic antitumor effect which is dependent on CD8+ and CD4+ T cells and IFN-γ. Oncolytic vaccinia viruses could not deliver stable IL-23A to the tumor, attributed to the elevated tristetraprolin which can destabilize the IL-23A mRNA after the viral treatment; whereas vaccinia viruses could deliver membrane-bound IL-23 to elicit a potent antitumor effect which might avoid the possible toxicity normally associated with systemic cytokine exposure. Conclusion: Either secreted or membrane-bound IL-23-armed vaccinia virus can induce potent antitumor effects and IL-23 is a candidate cytokine to arm oncolytic viruses for cancer immunotherapy.

Keywords: IL-23; cancer immunotherapy; efficacy.; oncolytic virus; tumor microenvironment.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adenocarcinoma / immunology
  • Adenocarcinoma / therapy*
  • Adenocarcinoma / virology
  • Animals
  • CD4-Positive T-Lymphocytes / immunology
  • CD8-Positive T-Lymphocytes / immunology
  • Cell Line, Tumor
  • Chemokines / metabolism
  • Colonic Neoplasms / immunology
  • Colonic Neoplasms / therapy*
  • Colonic Neoplasms / virology
  • Disease Models, Animal
  • Female
  • Genetic Vectors
  • Granzymes / metabolism
  • Humans
  • Immunotherapy / methods*
  • Interferon-gamma / metabolism
  • Interleukin-12 / metabolism
  • Interleukin-2 / metabolism
  • Interleukin-23 / genetics
  • Interleukin-23 / metabolism
  • Interleukin-23 / pharmacology*
  • Mice
  • Mice, Inbred BALB C
  • Mice, Inbred C57BL
  • Oncolytic Viruses / genetics*
  • Oncolytic Viruses / metabolism
  • Perforin / metabolism
  • Tumor Microenvironment / genetics
  • Tumor Microenvironment / immunology*
  • Tumor Necrosis Factor-alpha / metabolism
  • Vaccinia virus / genetics*
  • Vaccinia virus / metabolism

Substances

  • Chemokines
  • Interleukin-2
  • Interleukin-23
  • Tumor Necrosis Factor-alpha
  • Perforin
  • Interleukin-12
  • Interferon-gamma
  • Granzymes