[HTML][HTML] Irradiation-modulated murine brain microenvironment enhances GL261-tumor growth and inhibits Anti-PD-L1 immunotherapy

JR Garbow, TM Johanns, X Ge, JA Engelbach… - Frontiers in …, 2021 - frontiersin.org
JR Garbow, TM Johanns, X Ge, JA Engelbach, L Yuan, S Dahiya, CI Tsien, F Gao, KM Rich…
Frontiers in Oncology, 2021frontiersin.org
Purpose Clinical evidence suggests radiation induces changes in the brain
microenvironment that affect subsequent response to treatment. This study investigates the
effect of previous radiation, delivered six weeks prior to orthotopic tumor implantation, on
subsequent tumor growth and therapeutic response to anti-PD-L1 therapy in an intracranial
mouse model, termed the Radiation Induced Immunosuppressive Microenvironment (RI2M)
model. Method and Materials C57Bl/6 mice received focal (hemispheric) single-fraction, 30 …
Purpose
Clinical evidence suggests radiation induces changes in the brain microenvironment that affect subsequent response to treatment. This study investigates the effect of previous radiation, delivered six weeks prior to orthotopic tumor implantation, on subsequent tumor growth and therapeutic response to anti-PD-L1 therapy in an intracranial mouse model, termed the Radiation Induced Immunosuppressive Microenvironment (RI2M) model.
Method and Materials
C57Bl/6 mice received focal (hemispheric) single-fraction, 30-Gy radiation using the Leksell GammaKnife® Perfexion™, a dose that does not produce frank/gross radiation necrosis. Non-irradiated GL261 glioblastoma tumor cells were implanted six weeks later into the irradiated hemisphere. Lesion volume was measured longitudinally by in vivo MRI. In a separate experiment, tumors were implanted into either previously irradiated (30 Gy) or non-irradiated mouse brain, mice were treated with anti-PD-L1 antibody, and Kaplan-Meier survival curves were constructed. Mouse brains were assessed by conventional hematoxylin and eosin (H&E) staining, IBA-1 staining, which detects activated microglia and macrophages, and fluorescence-activated cell sorting (FACS) analysis.
Results
Tumors in previously irradiated brain display aggressive, invasive growth, characterized by viable tumor and large regions of hemorrhage and necrosis. Mice challenged intracranially with GL261 six weeks after prior intracranial irradiation are unresponsive to anti-PD-L1 therapy. K-M curves demonstrate a statistically significant difference in survival for tumor-bearing mice treated with anti-PD-L1 antibody between RI2M vs. non-irradiated mice. The most prominent immunologic change in the post-irradiated brain parenchyma is an increased frequency of activated microglia.
Conclusions
The RI2M model focuses on the persisting (weeks-to-months) impact of radiation applied to normal, control-state brain on the growth characteristics and immunotherapy response of subsequently implanted tumor. GL261 tumors growing in the RI2M grew markedly more aggressively, with tumor cells admixed with regions of hemorrhage and necrosis, and showed a dramatic loss of response to anti-PD-L1 therapy compared to tumors in non-irradiated brain. IHC and FACS analyses demonstrate increased frequency of activated microglia, which correlates with loss of sensitivity to checkpoint immunotherapy. Given that standard-of-care for primary brain tumor following resection includes concurrent radiation and chemotherapy, these striking observations strongly motivate detailed assessment of the late effects of the RI2M on tumor growth and therapeutic efficacy.
Frontiers