10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Single-Cycle Viral Gene Expression, Rather Than Progressive Replication and Oncolysis, Is Required for VSV Therapy of B16 Melanoma

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          A fully intact immune system would be expected to hinder the efficacy of oncolytic virotherapy by inhibiting viral replication. Simultaneously, however, it may also enhance antitumor therapy through initiation of pro-inflammatory, antiviral cytokine responses at the tumor site. The aim of the current study was to investigate the role of a fully intact immune system upon the antitumor efficacy of an oncolytic virus. In this respect, injection of oncolytic Vesicular Stomatitis Virus (VSV) into subcutaneous B16ova melanomas in C57Bl/6 mice leads to tumor regression, but it is not associated with viral replicative burst in the tumor. In contrast, intratumoral delivery of VSV induces an acute proinflammatory reaction, which quickly resolves concomitantly with virus clearance. Consistent with the hypothesis that therapy may not be dependent upon the ability of VSV to undergo progressive rounds of replication, a single-cycle VSV is equally effective as a fully replication-competent VSV, whereas, inactivated viruses do not generate therapy. Even though therapy is dependent upon host CD8+ and NK cells, these effects are not associated with IFN-γ-dependent responses against either the virus or tumor. There is, however, a strong correlation between viral gene expression, induction of proinflammatory reaction in the tumor and in vivo therapy. Overall, our results suggest that acute innate antiviral immune response, which rapidly clears VSV from B16ova tumors, is associated with the therapy observed in this model. Therefore, the antiviral immune response to an oncolytic virus mediates an intricate balance between safety, restriction of oncolysis and, potentially, significant immune-mediated antitumor therapy.

          Related collections

          Most cited references39

          • Record: found
          • Abstract: found
          • Article: not found

          VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents.

          Ideally, an oncolytic virus will replicate preferentially in malignant cells, have the ability to treat disseminated metastases, and ultimately be cleared by the patient. Here we present evidence that the attenuated vesicular stomatitis strains, AV1 and AV2, embody all of these traits. We uncover the mechanism by which these mutants are selectively attenuated in interferon-responsive cells while remaining highly lytic in 80% of human tumor cell lines tested. AV1 and AV2 were tested in a xenograft model of human ovarian cancer and in an immune competent mouse model of metastatic colon cancer. While highly attenuated for growth in normal mice, both AV1 and AV2 effected complete and durable cures in the majority of treated animals when delivered systemically.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Exploiting tumor-specific defects in the interferon pathway with a previously unknown oncolytic virus.

            Interferons are circulating factors that bind to cell surface receptors, activating a signaling cascade, ultimately leading to both an antiviral response and an induction of growth inhibitory and/or apoptotic signals in normal and tumor cells. Attempts to exploit the ability of interferons to limit the growth of tumors in patients has met with limited results because of cancer-specific mutations of gene products in the interferon pathway. Although interferon-non-responsive cancer cells may have acquired a growth/survival advantage over their normal counterparts, they may have simultaneously compromised their antiviral response. To test this, we used vesicular stomatitis virus (VSV), an enveloped, negative-sense RNA virus exquisitely sensitive to treatment with interferon. VSV rapidly replicated in and selectively killed a variety of human tumor cell lines even in the presence of doses of interferon that completely protected normal human primary cell cultures. A single intratumoral injection of VSV was effective in reducing the tumor burden of nude mice bearing subcutaneous human melanoma xenografts. Our results support the use of VSV as a replication-competent oncolytic virus and demonstrate a new strategy for the treatment of interferon non-responsive tumors.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Recombinant vesicular stomatitis viruses from DNA.

              We assembled a DNA clone containing the 11,161-nt sequence of the prototype rhabdovirus, vesicular stomatitis virus (VSV), such that it could be transcribed by the bacteriophage T7 RNA polymerase to yield a full-length positive-strand RNA complementary to the VSV genome. Expression of this RNA in cells also expressing the VSV nucleocapsid protein and the two VSV polymerase subunits resulted in production of VSV with the growth characteristics of wild-type VSV. Recovery of virus from DNA was verified by (i) the presence of two genetic tags generating restriction sites in DNA derived from the genome, (ii) direct sequencing of the genomic RNA of the recovered virus, and (iii) production of a VSV recombinant in which the glycoprotein was derived from a second serotype. The ability to generate VSV from DNA opens numerous possibilities for the genetic analysis of VSV replication. In addition, because VSV can be grown to very high titers and in large quantities with relative ease, it may be possible to genetically engineer recombinant VSVs displaying foreign antigens. Such modified viruses could be useful as vaccines conferring protection against other viruses.
                Bookmark

                Author and article information

                Journal
                9421525
                8603
                Gene Ther
                Gene Ther.
                Gene therapy
                0969-7128
                1476-5462
                28 December 2013
                17 December 2009
                February 2010
                25 February 2014
                : 17
                : 2
                : 158-170
                Affiliations
                [1 ]Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
                [2 ]Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
                [3 ]Department of Microbiology and Immunology and Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Miami, FL 33136, USA
                [4 ]Institute of Molecular Medicine, University of Leeds, Leeds, LS9 7TF, UK
                Author notes
                [* ]Corresponding author's address: Mayo Clinic / Guggenheim 18 / 200 First Street SW / Rochester, MN 55905 / 507-284-3178 / vile.richard@ 123456mayo.edu
                Article
                NIHMS150322
                10.1038/gt.2009.161
                3934361
                20016540
                f0479fa4-eae9-475a-b3fc-4f759086666f
                History
                Funding
                Funded by: National Cancer Institute : NCI
                Award ID: R01 CA130878 || CA
                Funded by: National Cancer Institute : NCI
                Award ID: R01 CA107082 || CA
                Categories
                Article

                Molecular medicine
                oncolytic viruses,experimental melanoma,interferon type i,gene therapy,virus replication,rhabdovirus

                Comments

                Comment on this article