Adeno-associated virus delivered CXCL9 sensitizes glioblastoma to anti-PD-1 immune checkpoint blockade

There are numerous mechanisms by which glioblastoma cells evade immunological detection, underscoring the need for strategic combinatorial treatments to achieve appreciable therapeutic effects. However, developing combination therapies is difficult due to dose-limiting toxicities, blood-brain-barrie...

Full description

Saved in:
Bibliographic Details
Published inNature communications Vol. 15; no. 1; pp. 5871 - 20
Main Authors von Roemeling, Christina A., Patel, Jeet A., Carpenter, Savannah L., Yegorov, Oleg, Yang, Changlin, Bhatia, Alisha, Doonan, Bently P., Russell, Rylynn, Trivedi, Vrunda S., Klippel, Kelena, Ryu, Daniel H., Grippin, Adam, Futch, Hunter S., Ran, Yong, Hoang-Minh, Lan B., Weidert, Frances L., Golde, Todd E., Mitchell, Duane A.
Format Journal Article
LanguageEnglish
Published London Nature Publishing Group UK 12.07.2024
Nature Publishing Group
Nature Portfolio
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:There are numerous mechanisms by which glioblastoma cells evade immunological detection, underscoring the need for strategic combinatorial treatments to achieve appreciable therapeutic effects. However, developing combination therapies is difficult due to dose-limiting toxicities, blood-brain-barrier, and suppressive tumor microenvironment. Glioblastoma is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment and activation. Herein, we develop a recombinant adeno-associated virus (AAV) gene therapy that enables focal and stable reconstitution of the tumor microenvironment with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for lymphocytes. By manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by cytotoxic lymphocytes, sensitizing glioblastoma to anti-PD-1 immune checkpoint blockade in female preclinical tumor models. These effects are accompanied by immunologic signatures evocative of an inflamed tumor microenvironment. These findings support AAV gene therapy as an adjuvant for reconditioning glioblastoma immunogenicity given its safety profile, tropism, modularity, and off-the-shelf capability. The limited infiltration and migration of T cells in the brain can hinder the success of immune checkpoint blockade in glioblastoma (GBM). Here the authors show that an adeno-associated virus-based gene therapy for the intratumor delivery of CXCL9 promotes T cell infiltration and enhances response to anti-PD1 in GBM preclinical models.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2041-1723
2041-1723
DOI:10.1038/s41467-024-49989-1