Anti–PD-1 induces M1 polarization in the glioma microenvironment and exerts therapeutic efficacy in the absence of CD8 cytotoxic T cells

G Rao, K Latha, M Ott, A Sabbagh, A Marisetty… - Clinical Cancer …, 2020 - AACR
G Rao, K Latha, M Ott, A Sabbagh, A Marisetty, X Ling, D Zamler, TA Doucette, Y Yang…
Clinical Cancer Research, 2020AACR
Purpose: Anti-programmed cell death protein 1 (PD-1) therapy has demonstrated
inconsistent therapeutic results in patients with glioblastoma (GBM) including those with
profound impairments in CD8 T-cell effector responses. Experimental Design: We ablated
the CD8α gene in BL6 mice and intercrossed them with Ntv-a mice to determine how CD8 T
cells affect malignant progression in forming endogenous gliomas. Tumor-bearing mice
were treated with PD-1 to determine the efficacy of this treatment in the absence of T cells …
Purpose
Anti-programmed cell death protein 1 (PD-1) therapy has demonstrated inconsistent therapeutic results in patients with glioblastoma (GBM) including those with profound impairments in CD8 T-cell effector responses.
Experimental Design
We ablated the CD8α gene in BL6 mice and intercrossed them with Ntv-a mice to determine how CD8 T cells affect malignant progression in forming endogenous gliomas. Tumor-bearing mice were treated with PD-1 to determine the efficacy of this treatment in the absence of T cells. The tumor microenvironment of treated and control mice was analyzed by IHC and FACS.
Results
We observed a survival benefit in immunocompetent mice with endogenously arising intracranial glioblastomas after intravenous administration of anti–PD-1. The therapeutic effect of PD-1 administration persisted in mice even after genetic ablation of the CD8 gene (CD8−/−). CD11b+ and Iba1+ monocytes and macrophages were enriched in the glioma microenvironment of the CD8−/− mice. The macrophages and microglia assumed a proinflammatory M1 response signature in the setting of anti–PD-1 blockade through the elimination of PD-1–expressing macrophages and microglia in the tumor microenvironment. Anti–PD-1 can inhibit the proliferation of and induce apoptosis of microglia through antibody-dependent cellular cytotoxicity, as fluorescently labeled anti–PD-1 was shown to gain direct access to the glioma microenvironment.
Conclusions
Our results show that the therapeutic effect of anti–PD-1 blockade in GBM may be mediated by the innate immune system, rather than by CD8 T cells. Anti–PD-1 immunologically modulates innate immunity in the glioma microenvironment—likely a key mode of activity.
AACR