Abstract

Oncolytic virotherapy is a promising new tool for cancer treatment, but direct lytic destruction of tumor cells is not sufficient and must be accompanied by strong immune activation to elicit anti-tumor immunity. We report here the creation of a novel replication-competent recombinant oncolytic herpes simplex virus type 1 (VG161) that carries genes coding for IL-12, IL-15, and IL-15 receptor alpha subunit, along with a peptide fusion protein capable of disrupting PD-1/PD-L1 interactions. The VG161 virus replicates efficiently and exhibits robust cytotoxicity in multiple tumor cell lines. Moreover, the encoded cytokines and the PD-L1 blocking peptide work cooperatively to boost immune cell function. In vivo testing in syngeneic CT26 and A20 tumor models reveals superior efficacy when compared to a backbone virus that does not express exogenous genes. Intratumoral injection of VG161 induces abscopal responses in non-injected distal tumors and grants resistance to tumor re-challenge. The robust anti-tumor effect of VG161 is associated with T cell and NK cell tumor infiltration, expression of Th1 associated genes in the injection site, and increased frequency of splenic tumor-specific T cells. VG161 also displayed a superb safety profile in GLP acute and repeated injection toxicity studies performed using cynomolgus monkeys. Overall, we demonstrate that VG161 can induce robust oncolysis and stimulate a robust anti-tumor immune response without sacrificing safety.

Highlights

  • A diverse range of oncolytic viruses (OVs) has shown efficacy in preclinical studies

  • Cells were infected with VG161 for 72 h at MOI ranging from 0.04 to 1, and cell viability was quantified by MTT assay

  • These results are consistent with cytotoxicity data for the parental virus HSV-345 (ICP34.5 deleted Herpes simplex virus type 1 (HSV-1) strain 17), which shows dramatically impaired cell killing in mouse tumor cell lines 4T1 and CT26 when compared to the human colon adenocarcinoma cell line LS174T (Figure S1A)

Read more

Summary

Introduction

A diverse range of oncolytic viruses (OVs) has shown efficacy in preclinical studies (reviewed in [1,2]). Only two OVs have cleared the hurdle of regulatory approval, consisting of the adenovirus H101 approved in China to treat head and neck cancer (4) and Talimogene laherparepvec (T-VEC) approved by the FDA for the treatment of advanced melanoma. It has become clear that direct infection and lysis of tumor cells is often not sufficient to generate a durable anti-tumor response. T-VEC was optimized for immunotherapy by expressing the cytokine granulocyte macrophage colony-stimulating factor (GM-CSF) and can generate systemic anti-tumor immunity as evidenced by observations of tumor regression in noninjected lesions [3,4,5,6,7]. It is apparent that a more robust immune response must be elicited in addition to potent oncolysis for an OV therapy to achieve long-lasing efficacy in the clinic

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call