Passaro et al. modified an engineered oncolytic herpes simplex virus (NG34, which expresses the human GADD34 gene to promote replication in glioblastoma [GBM] cells) by adding a single-chain antibody against PD-1 (scFvPD-1) to provide local checkpoint blockade. In vitro, NG34scFvPD-1-infected murine or human GBM cells produced and secreted scFvPD-1, which bound to PD-1 from either species. In two orthotopic models of murine GBM, intratumorally administered NG34scFvPD-1 prolonged survival and provided protection against tumor rechallenge in a manner dependent on the presence of a T cell immune response.

PURPOSE: Glioblastoma (GBM) is resistant to standard of care. Immune checkpoints inhibitors (such as anti-PD-1 monoclonal antibodies) efficiently restore anti-tumor T-cell activity. We engineered a new oncolytic herpes simplex virus (oHSV) expressing a single-chain antibody against PD-1 (scFvPD-1) to evaluate its efficacy in mouse models of GBM. Experimental plan: NG34scFvPD-1expresses the human GADD34 gene transcriptionally controlled by the Nestin promoter to allow replication in GBM cells and a scFvPD-1 cDNA transcriptionally controlled by the CMV promoter. ELISA assays were performed to detect binding of scFvPD-1 to mouse and human PD-1. In vitro cytotoxicity and replication assays were performed to measure NG34scFvPD-1 oncolysis, and scFvPD-1 expression and secretion were determined. In vivo survival studies using orthotopic mouse GBM models were performed to evaluate the therapeutic potency of NG34scFvPD-1. RESULTS: NG34scFvPD-1 infected GBM cells express and secrete scFvPD-1 that binds mouse PD-1. The introduction of the scFvPD-1 sequence in the viral backbone does not alter the oncolytic properties of NG34scFvPD-1. In situ NG34scFvPD-1 treatment improved the survival with a tail of durable survivorship in two syngeneic immunocompetent mouse models of GBM. Mice that survived the first GBM challenge rejected the second challenge of GBM when implanted in the contralateral hemisphere. However, this was not true when athymic mice were employed as the recipients of the second challenge, consistent with the need for an intact immune system to obtain a memory response. CONCLUSION: NG34scFvPD-1 treatment induces a durable anti-tumor response in two preclinical mouse models of GBM with evidence for antitumor memory.

Author Info: (1) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (2) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (3) Depart

Author Info: (1) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (2) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (3) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (4) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (5) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (6) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (7) Neurosurgery, Brigham and Women's Hospital. (8) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School. (9) Neurosurgery, Brigham and Women's Hospital. (10) Neurosurgery, Brigham & Women's Hospital. (11) Medical Oncology, Dana-Farber Cancer Institute. (12) Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine. (13) Center for Biostatistics, The Ohio State University. (14) Center for Biostatistics, Ohio State Unversity. (15) Center for Neuro-onology, Dana Farber Harvard Cancer Center. (16) Department of Medical Oncology, Dana-Farber Cancer Institute. (17) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School EAChiocca@partners.org. (18) Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School.