Talimogene laherparepvec (T-VEC) and other oncolytic viruses for the treatment of melanoma

PK Bommareddy, A Patel, S Hossain… - American journal of …, 2017 - Springer
PK Bommareddy, A Patel, S Hossain, HL Kaufman
American journal of clinical dermatology, 2017Springer
Many mammalian viruses have properties that can be commandeered for the treatment of
cancer. These characteristics include preferential infection and replication in tumor cells, the
initiation of tumor cell lysis, and the induction of innate and adaptive anti-tumor immunity.
Furthermore, viruses can be genetically engineered to reduce pathogenicity and increase
immunogenicity resulting in minimally toxic therapeutic agents. Talimogene laherparepvec
(T-VEC; Imlygic™), is a genetically modified herpes simplex virus, type 1, and is the first …
Abstract
Many mammalian viruses have properties that can be commandeered for the treatment of cancer. These characteristics include preferential infection and replication in tumor cells, the initiation of tumor cell lysis, and the induction of innate and adaptive anti-tumor immunity. Furthermore, viruses can be genetically engineered to reduce pathogenicity and increase immunogenicity resulting in minimally toxic therapeutic agents. Talimogene laherparepvec (T-VEC; Imlygic™), is a genetically modified herpes simplex virus, type 1, and is the first oncolytic virus therapy to be approved for the treatment of advanced melanoma by the US FDA. T-VEC is attenuated by the deletion of the herpes neurovirulence viral genes and enhanced for immunogenicity by the deletion of the viral ICP47 gene. Immunogenicity is further supported by expression of the human granulocyte–macrophage colony-stimulating factor (GM-CSF) gene, which helps promote the priming of T cell responses. T-VEC demonstrated significant improvement in durable response rate, objective response rate, and progression-free survival in a randomized phase III clinical trial for patients with advanced melanoma. This review will discuss the optimal selection of patients for such treatment and describe how therapy is optimally delivered. We will also discuss future directions for oncolytic virus immunotherapy, which will likely include combination T-VEC clinical trials, expansion of T-VEC to other types of non-melanoma skin cancers, and renewed efforts at oncolytic virus drug development with other viruses.
Springer