Pathology, organ distribution, and immune response after single and repeated intravenous injection of rats with clinical-grade parvovirus H1.

Parvovirus H1 (H1PV) is an autonomous parvovirus that is transmitted in rodent populations. Its natural host is rats. H1PV infection is nonpathogenic except in rat and hamster fetuses and newborns. H1PV infection of human cancer cells caused strong oncolytic effects in preclinical models. For a clinical trial of H1PV in patients with brain tumors, clinical-grade H1PV was produced according to Good Manufacturing Practices. This report focuses on results obtained after a single high-dose intravenous injection of highly purified H1PV in 30 rats and multiple (n = 17) intravenous injections at 3 dose levels in 223 rats. In both studies, no virus-related mortality or macroscopic organ changes related to H1PV occurred. Histopathology after multiple virus injections revealed minimal diffuse bile duct hyperplasia in livers of animals of the highest dose group and germinal center development in spleens of animals from the high-dose group. Liver changes were reversible within a 2-wk recovery period after the last injection. Hematology, blood chemistry, and coagulation analyses did not reveal significant toxicologic changes due to H1PV. Virus injection stimulated the production of IgG antibodies but did not alter mononuclear cell function or induce cytokine release. PCR analysis showed dose-dependent levels of viral genomes in all organs tested. The virus was excreted primarily through feces. These data provide important information regarding H1PV infection in its natural host. Due to the confirmation of the favorable safety profile of H1PV in a permissive animal model, a phase I/IIa clinical trial of H1PV in brain tumor patients could be initiated.

[1]  M. von Knebel Doeberitz,et al.  Phase I/IIa study of intratumoral/intracerebral or intravenous/intracerebral administration of Parvovirus H-1 (ParvOryx) in patients with progressive primary or recurrent glioblastoma multiforme: ParvOryx01 protocol , 2012, BMC Cancer.

[2]  O. Witt,et al.  Parvovirus H1 selectively induces cytotoxic effects on human neuroblastoma cells , 2010, International journal of cancer.

[3]  C. Sommer,et al.  Regression of advanced rat and human gliomas by local or systemic treatment with oncolytic parvovirus H-1 in rat models. , 2010, Neuro-oncology.

[4]  J. Rommelaere,et al.  Oncolytic parvoviruses as cancer therapeutics. , 2010, Cytokine & growth factor reviews.

[5]  A. Ho,et al.  Oncolytic rat parvovirus H-1PV, a candidate for the treatment of human lymphoma: In vitro and in vivo studies. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[6]  N. Giese,et al.  Improvement of Gemcitabine-Based Therapy of Pancreatic Carcinoma by Means of Oncolytic Parvovirus H-1PV , 2009, Clinical Cancer Research.

[7]  P. Champéroux,et al.  An optimised methodology for the neurobehavioural assessment in rodents. , 2007, Journal of pharmacological and toxicological methods.

[8]  M. Aprahamian,et al.  Combined oncolytic and vaccination activities of parvovirus H-1 in a metastatic tumor model. , 2007, Oncology reports.

[9]  A. Smith,et al.  Persistent rat parvovirus infection in individually housed rats , 2005, Archives of Virology.

[10]  K. Soike,et al.  Infection of newborn and fetal hamsters induced by inoculation of Lu III parvovirus , 1976, Archives of Virology.

[11]  D. Nettelbeck,et al.  Genetic replacement of the adenovirus shaft fiber reduces liver tropism in ovarian cancer gene therapy. , 2004, Human gene therapy.

[12]  C. Herold-Mende,et al.  Parvovirus H‐1 infection of human glioma cells leads to complete viral replication and efficient cell killing , 2004, International journal of cancer.

[13]  S. Irwin,et al.  Comprehensive observational assessment: Ia. A systematic, quantitative procedure for assessing the behavioral and physiologic state of the mouse , 1968, Psychopharmacologia.

[14]  M. Rentrop,et al.  Parvoviruses are inefficient in inducing interferon-β, tumor necrosis factor-α, or interleukin-6 in mammalian cells , 2004, Medical Microbiology and Immunology.

[15]  J. Jauniaux,et al.  Chimeric and Pseudotyped Parvoviruses Minimize the Contamination of Recombinant Stocks with Replication-Competent Viruses and Identify a DNA Sequence That Restricts Parvovirus H-1 in Mouse Cells , 2003, Journal of Virology.

[16]  D. Besselsen,et al.  Detection of H-1 parvovirus and Kilham rat virus by PCR , 1995, Journal of clinical microbiology.

[17]  J. Gierthy,et al.  Inhibition of 7,12-dimethylbenz(a)anthracene-induced tumors in Syrian hamsters by prior infection with H-1 parvovirus. , 1982, Cancer research.

[18]  L. Kilham,et al.  Enhanced proliferation of H-1 virus in livers of rats infected with Cysticercus fasciolaris. , 1970, The Journal of infectious diseases.

[19]  L. Kilham,et al.  Transplacental infection of rats and hamsters induced by oral and parenteral inoculations of H-l and rat viruses (RV). , 1969, Teratology.

[20]  S. Baron,et al.  Production of Interferon during Rat Virus Infection∗ , 1968, Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine.

[21]  N. Ledinko,et al.  Inhibition by H-1 virus of the incidence of tumors produced by adenovirus 12 in hamsters. , 1968, Virology.

[22]  L. Kilham,et al.  Rat Virus (RV) Infections of Pregnant, Fetal and Newborn Rats.∗ , 1964, Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine.

[23]  L. Kilham,et al.  Congenital Anomalies Induced in Hamster Embryos with H-1 Virus , 1964, Science.

[24]  L. Kilham,et al.  Rat Virus (RV) Infection in Fetal and Pregnant Hamsters.∗ , 1963, Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine.