scholarly journals 555. Non-Engineered, Replication Competent Oncolytic Herpes Simplex Virus HSV-1 HF10: Unique Pathogenicity and Enhancement of Cancer Treatment in Combination with an HSV Amplicon System Expressing Granulocyte-Macrophage Colony-Stimulation Factor (GM-CSF)

2008 ◽  
Vol 16 ◽  
pp. S208
2010 ◽  
Vol 183 (4S) ◽  
Author(s):  
Hiroshi Fukuhara ◽  
Jiangang Hou ◽  
Yuzuri Tsurumaki ◽  
Yukio Homma ◽  
Yasushi Ino ◽  
...  

2018 ◽  
Vol 11 (1) ◽  
pp. 86-93 ◽  
Author(s):  
Mohammed G. Ghonime ◽  
Josh Jackson ◽  
Amish Shah ◽  
Justin Roth ◽  
Mao Li ◽  
...  

2009 ◽  
Vol 181 (4S) ◽  
pp. 153-153
Author(s):  
Hiroshi Fukuhara ◽  
Yuzuri Tsurumaki ◽  
Yasushi Ino ◽  
Tomoki Todo ◽  
Yukio Honma

2020 ◽  
Author(s):  
Ifeanyi Kingsley Uche ◽  
Natalie Fowlkes ◽  
Luan Vu ◽  
Tatiane Watanabe ◽  
Mariano Carossino ◽  
...  

Oncolytic virotherapy (OVT) is now understood to be an immunotherapy that uses viral infection to liberate tumor antigens in an immunogenic context to promote the development of anti-tumor immune responses. The only currently FDA approved oncolytic virotherapy, T-Vec™, is a modified herpes simplex virus type I (HSV-1). While T-Vec™ is associated with limited response rates its modest efficacy supports the continued development of novel OVT viruses. Herein, we test the efficacy of a recombinant HSV-1, VC2, as an OVT in a syngeneic B16F10-derived mouse model of melanoma. VC2 possesses mutations that block its ability to enter neurons via axonal termini. This greatly enhances its safety profile by precluding the virus’s ability to establish latent infection. VC2 has been shown to be a safe, effective vaccine against both HSV-1 and HSV-2 infection in mice, guinea pigs, and non-human primates. We found that VC2 slows tumor growth rates and that VC2 treatment significantly enhances survival of tumor-engrafted, VC2-treated mice over control treatments. VC2-treated mice that survived initial tumor engraftment were resistant to a second engraftment as well as colonization of lungs by intravenous introduction of tumor cells. We found that VC2 treatment induced substantial increases in intratumoral T-cells and a decrease in immunosuppressive T-regulatory cells. This immunity was critically dependent on CD8+ T-cells and less dependent on CD4+ T-cells. Our data provide significant support for the continued development of VC2 as an OVT for the treatment of human and animal cancers. Importance Current oncolytic virotherapies possess limited response rates. However, when certain patient selection criteria are used, oncolytic virotherapy response rates have been shown to increase. This, in addition to the increased response rates of oncolytic virotherapy in combination with other immunotherapies, suggests that oncolytic viruses possess significant therapeutic potential for the treatment of cancer. As such, it is important to continue to develop novel oncolytic viruses as well as support basic research into their mechanisms of efficacy. Our data demonstrate significant clinical potential for VC2, a novel Type 1 oncolytic herpes simplex virus. Additionally, due to the high rates of survival and the dependence on CD8+ T-cells for efficacy, our model will enable study of the immunological correlates of protection for VC2 oncolytic virotherapy and oncolytic virotherapy in general. Understanding the mechanisms of efficacious oncolytic virotherapy will inform the rational design of improved oncolytic virotherapies.


Sign in / Sign up

Export Citation Format

Share Document