Elsevier

Current Opinion in Virology

Volume 13, August 2015, Pages 17-24
Current Opinion in Virology

Double-faceted mechanism of parvoviral oncosuppression

https://doi.org/10.1016/j.coviro.2015.03.008Get rights and content

Highlights

  • H-1PV propagation in human glioma cells can be improved through forced selection.

  • Arming of H-1PV vectors with immunostimulants enhances tumor vaccine effects.

  • H-1PV cooperates (in)directly with immune cells in achieving tumor suppression.

  • Post-translational modifications of the NS-1 protein regulate H-1PV oncolytic activity.

  • Gliomas from H-1PV-treated patients contain viral NS1 proteins and immune infiltrates.

The H-1 parvovirus (H-1PV) exerts oncosuppressive action that has two components: oncotoxicty and immunostimulation. While many human tumor cells, including conventional drug-resistant ones, can be killed by H-1PV, some fail to support progeny virus production, necessary for infection propagation in neoplastic tissues. This limitation can be overcome through forced selection of H-1PV variants capable of enhanced multiplication and spreading in human tumor cells. In the context of further developing H-1PV for use in cancer therapy, arming it with immunostimulatory CpG motifs under conditions preserving replication and oncolysis enhances its action as an anticancer vaccine adjuvant. A first clinical study of H-1PV treatment in glioma patients has yielded evidence of intratumoral synthesis of the viral oncotoxic protein NS1 and immune cell infiltration.

Introduction

Oncolytic parvoviruses belong to the species rodent protoparvovirus 1 and consist of an icosahedral protein capsid about 25 nm in diameter, containing a linear single-stranded DNA genome of approximately 5000 nucleotides. This genome comprises two transcription units, encoding nonstructural and capsid proteins, whose integrity is important for viral infectiousness [1]. The present review focuses on the oncolytic parvovirus H-1PV, whose natural host is the rat but which can replicate in and kill a number of tumor-derived human cells, while sparing their normal counterparts. As H-1PV also exerts tumor-suppressive action in various animal models (see [2, 3] for recent reviews), it is being assessed and developed as a potential tool for cancer therapy and/or prevention. Its antitumor action has two components: oncotoxicity and anticancer immunostimulation. Its adjuvant effect depends, at least in part, on the immunogenicity of viral oncolysates, and is instrumental in mediating virus-induced anticancer vaccination in animal models (reviewed in [4, 5, 6]). This review aims to illustrate the duality of H-1PV antitumor action and to discuss prospects for its optimization through virus adaptation, engineering, and/or combination with other agents.

Section snippets

Oncotoxic activity of rodent protoparvoviruses

Oncolytic parvoviruses induce molecular disturbances that jeopardize the survival of infected tumor cells. Besides exhausting cell metabolites as a result of their multiplication, they encode proteins that may participate in shutting down cell DNA replication and gene expression, by binding and withholding cellular factors controlling these processes [2]. Some parvoviral products, such as the 83-kDa multi-function nonstructural protein NS1, appear to alter target cell integrity by directly or

Immune mediation of H-1 parvovirus oncosuppression

There is growing evidence that the immune reactions triggered by non-pathogenic H-1PV depend largely on whether tumor tissues are present or not. Several in vitro studies have consistently shown that infection of human peripheral blood mononuclear cells (PBMCs) by H-1PV is not productive, although the first steps of viral replication do occur. More detailed analysis has identified B-cells, macrophages, and natural killer (NK) cells as the main immune cell subpopulations infected by H-1PV. In

Conclusion: clinical evidence of parvovirus oncolytic and immunomodulatory effects

In an immunocompetent rat model of advanced malignant glioma, stereotactic intratumoral injection of H-1PV caused progressive reduction of tumor size and changes in tumor morphology, resulting in complete remission in the majority of treated animals. Immunohistochemical analyses of tumor remnants at a regression stage corresponding to 30–70% of the initial tumor volume revealed the presence of the cytotoxic parvoviral nonstructural protein NS1 in the residual tumor masses, but not in the

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgments

We are indebted to members of the laboratory for contributing to animal immunodepletion and monitoring (Dr. Svitlana Grekova, Dr. Ute Koch and Manuel Fischer) and virus adaptation (Nadja Thomas). We wish to thank Mrs. Dagmar Anders for help with graphical artwork and Dr. Kathleen Broman for critical reading of the manuscript. The authors’ experimental work under review was supported by grants from Oryx GmbH and the German Research Council (DFG grant RA 1891/2-1).

References (50)

  • S.P. Grekova et al.

    Parvoviruses  tools to fine-tune anticancer immune responses

    Oncoimmunology

    (2012)
  • Z. Raykov et al.

    Parvoviruses: the friendly anticancer immuno-modulator

  • M. Moehler et al.

    Oncolytic virotherapy as emerging immunotherapeutic modality: potential of parvovirus H-1

    Front Oncol

    (2014)
  • J.P. Nuesch et al.

    A viral adaptor protein modulating casein kinase II activity induces cytopathic effects in permissive cells

    Proc Natl Acad Sci USA

    (2007)
  • M. Di Piazza et al.

    Cytosolic activation of cathepsins mediates parvovirus H-1-induced killing of cisplatin and TRAIL-resistant glioma cells

    J Virol

    (2007)
  • S. Bar et al.

    Vesicular transport of progeny parvovirus particles through ER and Golgi regulates maturation and cytolysis

    PLoS Pathog

    (2013)
  • J.K. Krady et al.

    Transcriptional activation by the parvoviral nonstructural protein NS-1 is mediated via a direct interaction with Sp1

    Mol Cell Biol

    (1995)
  • C. Lorson et al.

    An Sp1-binding site and TATA element are sufficient to support full transactivation by proximally bound NS1 protein of minute virus of mice

    Virology

    (1998)
  • T. Bashir et al.

    In vivo accumulation of cyclin A and cellular replication factors in autonomous parvovirus minute virus of mice-associated replication bodies

    J Virol

    (2001)
  • R.O. Adeyemi et al.

    Parvovirus minute virus of mice induces a DNA damage response that facilitates viral replication

    PLoS Pathog

    (2010)
  • Z. Ruiz et al.

    Recruitment of DNA replication and damage response proteins to viral replication centers during infection with NS2 mutants of Minute Virus of Mice (MVM)

    Virology

    (2011)
  • B. Rayet et al.

    Induction of programmed cell death by parvovirus H-1 in U937 cells: connection with the tumor necrosis factor alpha signalling pathway

    J Virol

    (1998)
  • Z. Ran et al.

    Parvovirus H-1-induced cell death: influence of intracellular NAD consumption on the regulation of necrosis and apoptosis

    Virus Res

    (1999)
  • S. Mousset et al.

    The cytotoxicity of the autonomous parvovirus minute virus of mice nonstructural proteins in FR3T3 rat cells depends on oncogene expression

    J Virol

    (1994)
  • L. Daeffler et al.

    Modulation of minute virus of mice cytotoxic activities through site-directed mutagenesis within the NS coding region

    J Virol

    (2003)
  • Cited by (25)

    • Oncolytic H-1 Parvovirus Shows Safety and Signs of Immunogenic Activity in a First Phase I/IIa Glioblastoma Trial

      2017, Molecular Therapy
      Citation Excerpt :

      Two previous applications of H-1PV in humans revealed no virus-related pathogenic effects.18,19 The oncosuppressive activity of H-1PV was demonstrated in numerous preclinical studies in glioblastoma and other tumor models.20,21 In rats, H-1PV can cross the blood-brain barrier, causing intracranial tumor regression after intravenous injection.22

    • Cancer Microbiology

      2022, Journal of the National Cancer Institute
    View all citing articles on Scopus
    3

    Present address: Experimental Neurosurgery, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany.

    View full text