Intravenous injection of oncolytic picornavirus SVV-001 prolongs animal survival in a panel of primary tumor–based orthotopic xenograft mouse models of pediatric …

Z Liu, X Zhao, H Mao, PA Baxter, Y Huang, L Yu… - Neuro …, 2013 - academic.oup.com
Z Liu, X Zhao, H Mao, PA Baxter, Y Huang, L Yu, L Wadhwa, JM Su, A Adesina, L Perlaky…
Neuro-oncology, 2013academic.oup.com
Abstract Background Seneca Valley virus (SVV-001) is a nonpathogenic oncolytic virus that
can be systemically administered and can pass through the blood–brain barrier. We
examined its therapeutic efficacy and the mechanism of tumor cell infection in pediatric
malignant gliomas. Methods In vitro antitumor activities were examined in primary cultures,
preformed neurospheres, and self-renewing glioma cells derived from 6 patient tumor
orthotopic xenograft mouse models (1 anaplastic astrocytoma and 5 GBM). In vivo …
Background
Seneca Valley virus (SVV-001) is a nonpathogenic oncolytic virus that can be systemically administered and can pass through the blood–brain barrier. We examined its therapeutic efficacy and the mechanism of tumor cell infection in pediatric malignant gliomas.
Methods
In vitro antitumor activities were examined in primary cultures, preformed neurospheres, and self-renewing glioma cells derived from 6 patient tumor orthotopic xenograft mouse models (1 anaplastic astrocytoma and 5 GBM). In vivo therapeutic efficacy was examined by systemic treatment of preformed xenografts in 3 permissive and 2 resistant models. The functional role of sialic acid in mediating SVV-001 infection was investigated using neuraminidase and lectins that cleave or competitively bind to linkage-specific sialic acids.
Results
SVV-001 at a multiplicity of infection of 0.5 to 25 replicated in and effectively killed primary cultures, preformed neurospheres, and self-renewing stemlike single glioma cells derived from 4 of the 6 glioma models in vitro. A single i.v. injection of SVV-001 (5 × 1012 viral particles/kg) led to the infection of orthotopic xenografts without harming normal mouse brain cells, resulting in significantly prolonged survival in all 3 permissive and 1 resistant mouse models (P < .05). Treatment with neuraminidase and competitive binding using lectins specific for α2,3-linked and/or α2,6-linked sialic acid significantly suppressed SVV-001 infectivity (P < .01).
Conclusion
SVV-001 possesses strong antitumor activity against pediatric malignant gliomas and utilizes α2,3-linked and α2,6-linked sialic acids as mediators of tumor cell infection. Our findings support the consideration of SVV-001 for clinical trials in children with malignant glioma.
Oxford University Press