Abstract

Abstract Glioblastoma (GBM) has an extremely immunosuppressive microenvironment, primarily mediated by the recruitment of immature myeloid cells (IMCs) to the tumor. IMCs suppress T cell function and dampen the anti-tumor response. In this study, we sought to “reprogram” IMCs into antigen-presenting cells (APCs) to enhance anti-tumor T cell responses. To induce IMC differentiation into APCs, we utilized a retroviral replicating vector (RRV) expressing IRF8, a master regulator of type 1 conventional dendritic cell (cDC1) development. We evaluated the impact of IRF8 RRV on in vivo tumor growth kinetics, survival, immunosuppression, and immune microenvironment. Because RRV transduction requires the proliferation of target cells, we assessed the proliferative capacity of myeloid cells in murine and human GBM samples. We utilized the SB28 cell line, a poorly immunogenic murine GBM model. We pre-transduced SB28 cells with IRF8 RRV and implanted them intracranially into syngeneic C57BL/6 mice; controls expressed an empty vector. Intratumoral myeloid cells were proliferative in both murine and human samples. In vivo, tumor and myeloid cells are infected proportionately to their respective levels of proliferation. Mice with IRF8 RRV pre-transduced intracranial tumors had significantly longer survival (median 28 days) compared to controls (median 19 days) (p <0.0001). Additionally, tumor growth was delayed in IRF8 RRV mice. IRF8 RRV mice had significant enrichment of dendritic cells (p = 0.0145), cDC1s (p < 0.0001), T cells (p = 0.0025) and CD8+ T cells (p < 0.0001). Both monocytic and granulocytic IMCs had decreased expression of the immunosuppressive markers Arginase 1 and IDO1. Further, ex vivo co-culture of intra-tumoral myeloid cells with naïve T cells showed a functional decrease in immunosuppression in IRF8 RRV tumors compared to controls (p < 0.0001). To study the mechanism of action, we assessed whether IRF8 transduction of tumor cells alone or both tumor and immune cells, was necessary for the observed survival benefit. To inhibit RRV spread from infected cells to other cells, we treated mice with azidothymidine, a reverse transcription inhibitor. A robust survival benefit and tumor growth inhibition were seen only in mice where the RRV could spread freely (p = 0.0005). This finding shows that both tumor and immune cells must be infected to cause the phenotype and suggests that reprogramming immune cells is required for therapeutic efficacy. Our novel approach used RRV to deliver a myeloid transcriptional regulator in intracerebral GBM. This therapy achieved significant survival benefits and increased intratumoral cytotoxic T cells and APCs, while decreasing myeloid immunosuppression. Further studies will expand upon the mechanism of action and test combination therapies. Citation Format: Megan Montoya, Sara A. Collins, Pavlina Chuntova, Noriyuki Kasahara, Hideho Okada. IRF8 driven reprogramming of the immune microenvironment enhances anti-tumor adaptive immunity and reduces immunosuppression in murine glioblastoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 1417.

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