American Association for Cancer Research
Browse
crc-23-0424-s07.pdf (76.89 kB)

Supplementary Figure 5 from CAR T-cell Design-dependent Remodeling of the Brain Tumor Immune Microenvironment Modulates Tumor-associated Macrophages and Anti-glioma Activity

Download (76.89 kB)
journal contribution
posted on 2023-12-01, 02:20 authored by Dalia Haydar, Jorge Ibañez-Vega, Jeremy Chase Crawford, Ching-Heng Chou, Clifford S. Guy, Michaela Meehl, Zhongzhen Yi, Scott Perry, Jonathan Laxton, Trevor Cunningham, Deanna Langfitt, Peter Vogel, Christopher DeRenzo, Stephen Gottschalk, Martine F. Roussel, Paul G. Thomas, Giedre Krenciute

Supplementary Figure S5 shows murine CAR T cell expansion in media and after exposure to B7-H3-negative tumor cells.

Funding

HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)

HHS | NIH | National Cancer Institute (NCI)

the Katzen Foundation

History

ARTICLE ABSTRACT

Understanding the intricate dynamics between adoptively transferred immune cells and the brain tumor immune microenvironment (TIME) is crucial for the development of effective T cell–based immunotherapies. In this study, we investigated the influence of the TIME and chimeric antigen receptor (CAR) design on the anti-glioma activity of B7-H3–specific CAR T-cells. Using an immunocompetent glioma model, we evaluated a panel of seven fully murine B7-H3 CARs with variations in transmembrane, costimulatory, and activation domains. We then investigated changes in the TIME following CAR T-cell therapy using high-dimensional flow cytometry and single-cell RNA sequencing. Our results show that five out of six B7-H3 CARs with single costimulatory domains demonstrated robust functionality in vitro. However, these CARs had significantly varied levels of antitumor activity in vivo. To enhance therapeutic effectiveness and persistence, we incorporated 41BB and CD28 costimulation through transgenic expression of 41BBL on CD28-based CAR T-cells. This CAR design was associated with significantly improved anti-glioma efficacy in vitro but did not result in similar improvements in vivo. Analysis of the TIME revealed that CAR T-cell therapy influenced the composition of the TIME, with the recruitment and activation of distinct macrophage and endogenous T-cell subsets crucial for successful antitumor responses. Indeed, complete brain macrophage depletion using a CSF1R inhibitor abrogated CAR T-cell antitumor activity. In sum, our study highlights the critical role of CAR design and its modulation of the TIME in mediating the efficacy of adoptive immunotherapy for high-grade glioma. CAR T-cell immunotherapies hold great potential for treating brain cancers; however, they are hindered by a challenging immune environment that dampens their effectiveness. In this study, we show that the CAR design influences the makeup of the immune environment in brain tumors, underscoring the need to target specific immune components to improve CAR T-cell performance, and highlighting the significance of using models with functional immune systems to optimize this therapy.

Usage metrics

    Cancer Research Communications

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC