Targeting of interferon-beta to produce a specific, multi-mechanistic oncolytic vaccinia virus

PLoS Med. 2007 Dec;4(12):e353. doi: 10.1371/journal.pmed.0040353.

Abstract

Background: Oncolytic viruses hold much promise for clinical treatment of many cancers, but a lack of systemic delivery and insufficient tumor cell killing have limited their usefulness. We have previously demonstrated that vaccinia virus strains are capable of systemic delivery to tumors in mouse models, but infection of normal tissues remains an issue. We hypothesized that interferon-beta (IFN-beta) expression from an oncolytic vaccinia strain incapable of responding to this cytokine would have dual benefits as a cancer therapeutic: increased anticancer effects and enhanced virus inactivation in normal tissues. We report the construction and preclinical testing of this virus.

Methods and findings: In vitro screening of viral strains by cytotoxicity and replication assay was coupled to cellular characterization by phospho-flow cytometry in order to select a novel oncolytic vaccinia virus. This virus was then examined in vivo in mouse models by non-invasive imaging techniques. A vaccinia B18R deletion mutant was selected as the backbone for IFN-beta expression, because the B18R gene product neutralizes secreted type-I IFNs. The oncolytic B18R deletion mutant demonstrated IFN-dependent cancer selectivity and efficacy in vitro, and tumor targeting and efficacy in mouse models in vivo. Both tumor cells and tumor-associated vascular endothelial cells were targeted. Complete tumor responses in preclinical models were accompanied by immune-mediated protection against tumor rechallenge. Cancer selectivity was also demonstrated in primary human tumor explant tissues and adjacent normal tissues. The IFN-beta gene was then cloned into the thymidine kinase (TK) region of this virus to create JX-795 (TK-/B18R-/IFN-beta+). JX-795 had superior tumor selectivity and systemic intravenous efficacy when compared with the TK-/B18R- control or wild-type vaccinia in preclinical models.

Conclusions: By combining IFN-dependent cancer selectivity with IFN-beta expression to optimize both anticancer effects and normal tissue antiviral effects, we were able to achieve, to our knowledge for the first time, tumor-specific replication, IFN-beta gene expression, and efficacy following systemic delivery in preclinical models.

Publication types

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

MeSH terms

  • Animals
  • Cell Survival
  • Endothelium, Vascular / metabolism
  • Endothelium, Vascular / pathology
  • Genes, Reporter
  • HCT116 Cells
  • Haplorhini
  • Humans
  • Interferon-alpha / metabolism
  • Interferon-beta / genetics
  • Interferon-beta / metabolism*
  • Luciferases
  • Mice
  • Mice, Inbred BALB C
  • Mice, Inbred C57BL
  • NIH 3T3 Cells
  • Neoplasms, Experimental / blood supply
  • Neoplasms, Experimental / genetics
  • Neoplasms, Experimental / metabolism
  • Neoplasms, Experimental / pathology
  • Neoplasms, Experimental / therapy*
  • Oncolytic Virotherapy*
  • Oncolytic Viruses / genetics
  • Oncolytic Viruses / metabolism*
  • Sequence Deletion
  • Thymidine Kinase / genetics
  • Thymidine Kinase / metabolism
  • Time Factors
  • Tissue Distribution
  • Vaccinia virus / enzymology
  • Vaccinia virus / genetics
  • Vaccinia virus / metabolism*
  • Viral Proteins / genetics
  • Viral Proteins / metabolism

Substances

  • Interferon-alpha
  • Viral Proteins
  • B18R protein, Vaccinia virus
  • Interferon-beta
  • Luciferases
  • Thymidine Kinase