Bommareddy et al. studied mechanisms underlying the efficacy of T-VEC, an oncolytic herpes simplex virus 1 encoding granulocyte-macrophage colony stimulating factor that was recently approved in advanced melanoma. In melanoma cell lines, STING expression positively correlated with T-VEC resistance; STING knockout in the most resistant line increased sensitivity and altered cytokine production. T-VEC induced immunogenic cell death. In mice with STINGlo melanoma tumors on opposite flanks, T-VEC injection into one tumor led to reduced growth, greater numbers of virus and melanoma-specific CD8+ T cells, and pro-immune gene profiles in both.

Contributed by Alex Najibi


Want to know more about oncolytic virus therapy?

We have partnered with ReFigure.org, a novel platform for collecting scientific figures into short-form insights. Click here to see complementary content that the ReFigure team has generated for this spotlight.

Successful immunotherapy for melanoma depends on the recruitment of effector CD8(+) T cells to the tumor microenvironment. Factors contributing to T cell regulation in melanoma have recently been recognized, including the stimulator of interferon genes (STING). Agents that can activate STING or enhance T cell infiltration into established tumors have become an important focus for further clinical development. Talimogene laherparepvec (T-VEC) is an oncolytic herpes simplex virus, type 1 (HSV-1) encoding granulocyte-macrophage colony stimulating factor (GM-CSF) and is approved for the treatment of melanoma and has shown therapeutic activity in murine tumors known to express high levels of STING. The mechanism of action for T-VEC has not been fully elucidated but is thought to include induction of immunogenic cell death (ICD) and activation of host anti-tumor immunity. Thus, we sought to investigate how T-VEC mediates anti-tumor activity in a melanoma model. To determine if T-VEC induced ICD we established the relative sensitivity of a panel of melanoma cell lines to T-VEC oncolysis. Following T-VEC infection in vitro, melanoma cell lines released of HMGB1, ATP, and translocated ecto-calreticulin. To identify potential mediators of this effect, we found that melanoma cell sensitivity to T-VEC was inversely related to STING expression. CRISPR/Cas9-STING knockout was also associated with increased T-VEC cell killing. In the D4M3A melanoma, which has low expression of STING and is resistant to PD-1 blockade therapy, T-VEC was able to induce therapeutic responses in both injected and non-injected tumors and demonstrated recruitment of viral- and tumor-antigen specific CD8(+) T cells, and induction of a pro-inflammatory gene signature at both injected and non-injected tumors. These data suggest that T-VEC induces ICD in-vitro and promotes tumor immunity and can induce therapeutic responses in anti-PD-1-refractory, low STING expressing melanoma.

Author Info: (1) School of Graduate Studies & Rutgers Cancer Institute, Rutgers University, Rutgers Universi, New Brunswick, NJ, USA. (2) School of Graduate Studies & Rutgers Cancer Institute,

Author Info: (1) School of Graduate Studies & Rutgers Cancer Institute, Rutgers University, Rutgers Universi, New Brunswick, NJ, USA. (2) School of Graduate Studies & Rutgers Cancer Institute, Rutgers University, Rutgers Universi, New Brunswick, NJ, USA. Division of Hematology, Oncology, and Cell Therapy, Department of Internal Medicine, Rush University Medical Center, Chicago, USA. (3) Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA. (4) Division of Surgical Oncology, Massachusetts General Hospital, Boston, MA, USA. Replimune, Inc., Woburn, MA, USA.