Oncolytic adenovirus expressing IL-23 and p35 elicits IFN-γ- and TNF-α-co-producing T cell-mediated antitumor immunity

PLoS One. 2013 Jul 3;8(7):e67512. doi: 10.1371/journal.pone.0067512. Print 2013.

Abstract

Cytokine immunogene therapy is a promising strategy for cancer treatment. Interleukin (IL)-12 boosts potent antitumor immunity by inducing T helper 1 cell differentiation and stimulating cytotoxic T lymphocyte and natural killer cell cytotoxicity. IL-23 has been proposed to have similar but not overlapping functions with IL-12 in inducing Th1 cell differentiation and antitumor immunity. However, the therapeutic effects of intratumoral co-expression of IL-12 and IL-23 in a cancer model have yet to be investigated. Therefore, we investigated for the first time an effective cancer immunogene therapy of syngeneic tumors via intratumoral inoculation of oncolytic adenovirus co-expressing IL-23 and p35, RdB/IL23/p35. Intratumoral administration of RdB/IL23/p35 elicited strong antitumor effects and increased survival in a murine B16-F10 syngeneic tumor model. The levels of IL-12, IL-23, interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α) were elevated in RdB/IL23/p35-treated tumors. Moreover, the proportion of regulatory T cells was markedly decreased in mice treated with RdB/IL23/p35. Consistent with these data, mice injected with RdB/IL23/p35 showed massive infiltration of CD4(+) and CD8(+) T cells into the tumor as well as enhanced induction of tumor-specific immunity. Importantly, therapeutic mechanism of antitumor immunity mediated by RdB/IL23/p35 is associated with the generation and recruitment of IFN-γ- and TNF-α-co-producing T cells in tumor microenvironment. These results provide a new insight into therapeutic mechanisms of IL-12 plus IL-23 and provide a potential clinical cancer immunotherapeutic agent for improved antitumor immunity.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adenoviridae / genetics*
  • Animals
  • Cytokines / genetics*
  • Cytokines / immunology
  • Gene Dosage
  • Gene Expression
  • Gene Order
  • Genetic Therapy
  • Genetic Vectors / administration & dosage
  • Genetic Vectors / genetics*
  • Interferon-gamma / genetics
  • Interferon-gamma / immunology
  • Interleukin-12 Subunit p35 / genetics
  • Interleukin-12 Subunit p35 / immunology
  • Interleukin-12 Subunit p40 / genetics
  • Interleukin-12 Subunit p40 / immunology
  • Interleukin-23 / genetics
  • Interleukin-23 / immunology
  • Male
  • Mice
  • Neoplasms / immunology*
  • Neoplasms / mortality
  • Neoplasms / therapy*
  • Oncolytic Virotherapy
  • Oncolytic Viruses / genetics
  • T-Lymphocyte Subsets / immunology*
  • T-Lymphocyte Subsets / metabolism*
  • Tumor Necrosis Factor-alpha / genetics
  • Tumor Necrosis Factor-alpha / immunology

Substances

  • Cytokines
  • Interleukin-12 Subunit p35
  • Interleukin-12 Subunit p40
  • Interleukin-23
  • Tumor Necrosis Factor-alpha
  • Interferon-gamma

Grants and funding

This work was supported by grants from the Ministry of Knowledge Economy (10030051, Dr. CO Yun), the Korea Science and Engineering Foundation (R15-2004-024-02001-0, 2009K001644, 2010-0029220, Dr. CO Yun), and the Korea Food and Drug Administration (KFDA-13172-306 to Dr. CO Yun). Yan Li is a graduate student sponsored by KOSEF through National Core Research Center for Nanomedical Technology, Seoul, Korea. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.