Your browser doesn't support javascript.
loading
IRF8-driven reprogramming of the immune microenvironment enhances anti-tumor adaptive immunity and reduces immunosuppression in murine glioblastoma.
Montoya, Megan; Collins, Sara A; Chuntova, Pavlina; Patel, Trishna S; Nejo, Takahide; Yamamichi, Akane; Kasahara, Noriyuki; Okada, Hideho.
Afiliación
  • Montoya M; Department of Neurological Surgery, University of California San Francisco, San Francisco, California.
  • Collins SA; Department of Neurological Surgery, University of California San Francisco, San Francisco, California.
  • Chuntova P; Department of Neurological Surgery, University of California San Francisco, San Francisco, California.
  • Patel TS; Department of Neurological Surgery, University of California San Francisco, San Francisco, California.
  • Nejo T; Department of Neurological Surgery, University of California San Francisco, San Francisco, California.
  • Yamamichi A; Department of Neurological Surgery, University of California San Francisco, San Francisco, California.
  • Kasahara N; Department of Neurological Surgery, University of California San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; Department of Radiation Oncology, University of California San Francisco, San Fra
  • Okada H; Department of Neurological Surgery, University of California San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; The Parker Institute for Cancer Immunotherapy.
bioRxiv ; 2024 Apr 03.
Article en En | MEDLINE | ID: mdl-38617245
ABSTRACT

Background:

Glioblastoma (GBM) has a highly immunosuppressive tumor immune microenvironment (TIME), largely mediated by myeloid-derived suppressor cells (MDSCs). Here, we utilized a retroviral replicating vector (RRV) to deliver Interferon Regulatory Factor 8 (IRF8), a master regulator of type 1 conventional dendritic cell (cDC1) development, in a syngeneic murine GBM model. We hypothesized that RRV-mediated delivery of IRF8 could "reprogram" intratumoral MDSCs into antigen-presenting cells (APCs) and thereby restore T-cell responses.

Methods:

Effects of RRV-IRF8 on survival and tumor growth kinetics were examined in the SB28 murine GBM model. Immunophenotype was analyzed by flow cytometry and gene expression assays. We assayed functional immunosuppression and antigen presentation by ex vivo T-cell-myeloid co-culture.

Results:

Mice with RRV-IRF8 pre-transduced intracerebral tumors had significantly longer survival and slower tumor growth compared to controls. RRV-IRF8 treated tumors exhibited significant enrichment of cDC1s and CD8+ T-cells. Additionally, myeloid cells derived from RRV-IRF8 tumors showed decreased expression of the immunosuppressive markers Arg1 and IDO1 and demonstrated reduced suppression of naïve T-cell proliferation in ex vivo co-culture, compared to controls. Furthermore, DCs from RRV-IRF8 tumors showed increased antigen presentation compared to those from control tumors. In vivo treatment with azidothymidine (AZT), a viral replication inhibitor, showed that IRF8 transduction in both tumor and non-tumor cells is necessary for survival benefit, associated with a reprogrammed, cDC1- and CD8 T-cell-enriched TIME.

Conclusions:

Our results indicate that reprogramming of glioma-infiltrating myeloid cells by in vivo expression of IRF8 may reduce immunosuppression and enhance antigen presentation, achieving improved tumor control.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: BioRxiv Año: 2024 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: BioRxiv Año: 2024 Tipo del documento: Article