Irradiation-Modulated Murine Brain Microenvironment Enhances GL261-Tumor Growth and Inhibits Anti-PD-L1 Immunotherapy

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 res...

Full description

Bibliographic Details
Main Authors: Joel R. Garbow, Tanner M. Johanns, Xia Ge, John A. Engelbach, Liya Yuan, Sonika Dahiya, Christina I. Tsien, Feng Gao, Keith M. Rich, Joseph J. H. Ackerman
Format: Article
Language:English
Published: Frontiers Media S.A. 2021-06-01
Series:Frontiers in Oncology
Subjects:
MRI
Online Access:https://www.frontiersin.org/articles/10.3389/fonc.2021.693146/full
id doaj-df0d9d4ab43a4809bf80c2fe665a833b
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Joel R. Garbow
Joel R. Garbow
Tanner M. Johanns
Tanner M. Johanns
Xia Ge
John A. Engelbach
Liya Yuan
Sonika Dahiya
Christina I. Tsien
Feng Gao
Keith M. Rich
Joseph J. H. Ackerman
Joseph J. H. Ackerman
Joseph J. H. Ackerman
Joseph J. H. Ackerman
spellingShingle Joel R. Garbow
Joel R. Garbow
Tanner M. Johanns
Tanner M. Johanns
Xia Ge
John A. Engelbach
Liya Yuan
Sonika Dahiya
Christina I. Tsien
Feng Gao
Keith M. Rich
Joseph J. H. Ackerman
Joseph J. H. Ackerman
Joseph J. H. Ackerman
Joseph J. H. Ackerman
Irradiation-Modulated Murine Brain Microenvironment Enhances GL261-Tumor Growth and Inhibits Anti-PD-L1 Immunotherapy
Frontiers in Oncology
MRI
radiation
tumor
microenvironment
immunotherapy
checkpoint inhibitors
author_facet Joel R. Garbow
Joel R. Garbow
Tanner M. Johanns
Tanner M. Johanns
Xia Ge
John A. Engelbach
Liya Yuan
Sonika Dahiya
Christina I. Tsien
Feng Gao
Keith M. Rich
Joseph J. H. Ackerman
Joseph J. H. Ackerman
Joseph J. H. Ackerman
Joseph J. H. Ackerman
author_sort Joel R. Garbow
title Irradiation-Modulated Murine Brain Microenvironment Enhances GL261-Tumor Growth and Inhibits Anti-PD-L1 Immunotherapy
title_short Irradiation-Modulated Murine Brain Microenvironment Enhances GL261-Tumor Growth and Inhibits Anti-PD-L1 Immunotherapy
title_full Irradiation-Modulated Murine Brain Microenvironment Enhances GL261-Tumor Growth and Inhibits Anti-PD-L1 Immunotherapy
title_fullStr Irradiation-Modulated Murine Brain Microenvironment Enhances GL261-Tumor Growth and Inhibits Anti-PD-L1 Immunotherapy
title_full_unstemmed Irradiation-Modulated Murine Brain Microenvironment Enhances GL261-Tumor Growth and Inhibits Anti-PD-L1 Immunotherapy
title_sort irradiation-modulated murine brain microenvironment enhances gl261-tumor growth and inhibits anti-pd-l1 immunotherapy
publisher Frontiers Media S.A.
series Frontiers in Oncology
issn 2234-943X
publishDate 2021-06-01
description 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.
topic MRI
radiation
tumor
microenvironment
immunotherapy
checkpoint inhibitors
url https://www.frontiersin.org/articles/10.3389/fonc.2021.693146/full
work_keys_str_mv AT joelrgarbow irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT joelrgarbow irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT tannermjohanns irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT tannermjohanns irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT xiage irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT johnaengelbach irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT liyayuan irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT sonikadahiya irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT christinaitsien irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT fenggao irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT keithmrich irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT josephjhackerman irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT josephjhackerman irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT josephjhackerman irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
AT josephjhackerman irradiationmodulatedmurinebrainmicroenvironmentenhancesgl261tumorgrowthandinhibitsantipdl1immunotherapy
_version_ 1721361739262459904
spelling doaj-df0d9d4ab43a4809bf80c2fe665a833b2021-06-24T04:43:59ZengFrontiers Media S.A.Frontiers in Oncology2234-943X2021-06-011110.3389/fonc.2021.693146693146Irradiation-Modulated Murine Brain Microenvironment Enhances GL261-Tumor Growth and Inhibits Anti-PD-L1 ImmunotherapyJoel R. Garbow0Joel R. Garbow1Tanner M. Johanns2Tanner M. Johanns3Xia Ge4John A. Engelbach5Liya Yuan6Sonika Dahiya7Christina I. Tsien8Feng Gao9Keith M. Rich10Joseph J. H. Ackerman11Joseph J. H. Ackerman12Joseph J. H. Ackerman13Joseph J. H. Ackerman14Department of Radiology, Washington University, Saint Louis, MO, United StatesAlvin J. Siteman Cancer Center, Washington University, Saint Louis, MO, United StatesDepartment of Internal Medicine, Washington University, Saint Louis, MO, United StatesAlvin J. Siteman Cancer Center, Washington University, Saint Louis, MO, United StatesDepartment of Radiology, Washington University, Saint Louis, MO, United StatesDepartment of Radiology, Washington University, Saint Louis, MO, United StatesDepartment of Neurosurgery, Washington University, Saint Louis, MO, United StatesDivision of Neuropathology, Department of Pathology and Immunology, Washington University, Saint Louis, MO, United StatesDepartment of Radiation Oncology, Washington University, Saint Louis, MO, United StatesDepartment of Surgery, Washington University, Saint Louis, MO, United StatesDepartment of Neurosurgery, Washington University, Saint Louis, MO, United StatesDepartment of Radiology, Washington University, Saint Louis, MO, United StatesAlvin J. Siteman Cancer Center, Washington University, Saint Louis, MO, United StatesDepartment of Internal Medicine, Washington University, Saint Louis, MO, United StatesDepartment of Chemistry, Washington University, Saint Louis, MO, United StatesPurposeClinical 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.https://www.frontiersin.org/articles/10.3389/fonc.2021.693146/fullMRIradiationtumormicroenvironmentimmunotherapycheckpoint inhibitors