Abstract

PurposeClinical 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 MaterialsC57Bl/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.ResultsTumors 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.ConclusionsThe 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.

Highlights

  • Radiotherapy combined with immunotherapy is an active area of investigation in the treatment of brain tumors

  • We demonstrate that mice challenged intracranially with non-irradiated GL261 cells after prior intracranial irradiation with a dose eliciting no frank/gross evidence of radiation necrosis or other pathology are unresponsive to antiPD-1/PD-L1 directed therapy

  • Mice were anesthetized and restrained on a custom-built platform mounted to the stereotactic frame that attaches to the treatment couch of the Leksell GK PerfexionTM (Elekta, Stockholm, Sweden), a device used for stereotactic radiosurgery of patients with malignant brain tumors

Read more

Summary

Introduction

Radiotherapy combined with immunotherapy is an active area of investigation in the treatment of brain tumors. Studies of patients with metastatic brain tumors noted metastatic lesions that progress after initial irradiation are often less responsive to subsequent treatment. Similar observations were noted in a separate cohort of patients with melanoma brain metastases, in which the cohort with lesions that progressed following prior irradiation had a substantially lower response rate to immunotherapy compared to the cohort with irradiation-naïve lesions [6]. PD1 monotherapy and PD-1/CTLA-4 combination therapy alone failed to demonstrate clinical benefit or objective response rates [7,8,9] These patients were all treated previously with chemoradiotherapy per standard-of-care. Together, these clinical observations suggest that late effects of prior irradiation to the brain microenvironment may be associated with resistance to immune checkpoint inhibition (ICI) therapy

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.