Your browser doesn't support javascript.
loading
Melanoma Cell Intrinsic GABAA Receptor Enhancement Potentiates Radiation and Immune Checkpoint Inhibitor Response by Promoting Direct and T Cell-Mediated Antitumor Activity.
Pomeranz Krummel, Daniel A; Nasti, Tahseen H; Kaluzova, Milota; Kallay, Laura; Bhattacharya, Debanjan; Melms, Johannes C; Izar, Benjamin; Xu, Maxwell; Burnham, Andre; Ahmed, Taukir; Li, Guanguan; Lawson, David; Kowalski, Jeanne; Cao, Yichun; Switchenko, Jeffrey M; Ionascu, Dan; Cook, James M; Medvedovic, Mario; Jenkins, Andrew; Khan, Mohammad K; Sengupta, Soma.
Afiliação
  • Pomeranz Krummel DA; Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.
  • Nasti TH; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia.
  • Kaluzova M; Emory University School of Medicine, Atlanta, Georgia.
  • Kallay L; Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.
  • Bhattacharya D; Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.
  • Melms JC; Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, New York.
  • Izar B; Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, New York.
  • Xu M; Johns Hopkins University, Baltimore, Maryland.
  • Burnham A; Emory University School of Medicine, Atlanta, Georgia.
  • Ahmed T; Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin.
  • Li G; Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin.
  • Lawson D; Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia.
  • Kowalski J; Department of Oncology, LIVESTRONG Cancer Institutes, Dell Medical School, University of Texas, Austin, Texas.
  • Cao Y; Biostatistics Shared Resource, Winship Cancer Institute of Emory University, Atlanta, Georgia.
  • Switchenko JM; Biostatistics Shared Resource, Winship Cancer Institute of Emory University, Atlanta, Georgia; Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia.
  • Ionascu D; Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
  • Cook JM; Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin.
  • Medvedovic M; Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio.
  • Jenkins A; Departments of Anesthesiology, Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia.
  • Khan MK; Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia.
  • Sengupta S; Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio. Electronic address: sengupsm@ucmail.uc.edu.
Int J Radiat Oncol Biol Phys ; 109(4): 1040-1053, 2021 03 15.
Article em En | MEDLINE | ID: mdl-33289666
ABSTRACT

PURPOSE:

Most patients with metastatic melanoma show variable responses to radiation therapy and do not benefit from immune checkpoint inhibitors. Improved strategies for combination therapy that leverage potential benefits from radiation therapy and immune checkpoint inhibitors are critical. METHODS AND MATERIALS We analyzed metastatic melanoma tumors in the TCGA cohort for expression of genes coding for subunits of type A γ-aminobutyric acid (GABA) receptor (GABAAR), a chloride ion channel and major inhibitory neurotransmitter receptor. Electrophysiology was used to determine whether melanoma cells possess intrinsic GABAAR activity. Melanoma cell viability studies were conducted to test whether enhancing GABAAR mediated chloride transport using benzodiazepine-impaired viability. A syngeneic melanoma mouse model was used to assay the effect of benzodiazepine on tumor volume and its ability to potentiate radiation therapy or immunotherapy. Treated tumors were analyzed for changes in gene expression by RNA sequencing and presence of tumor-infiltrating lymphocytes by flow cytometry.

RESULTS:

Genes coding for subunits of GABAARs express functional GABAARs in melanoma cells. By enhancing GABAAR-mediated anion transport, benzodiazepines depolarize melanoma cells and impair their viability. In vivo, benzodiazepine alone reduces tumor growth and potentiates radiation therapy and α-PD-L1 antitumor activity. The combination of benzodiazepine, radiation therapy, and α-PD-L1 results in near complete regression of treated tumors and a potent abscopal effect, mediated by increased infiltration of polyfunctional CD8+ T cells. Treated tumors show expression of cytokine-cytokine receptor interactions and overrepresentation of p53 signaling.

CONCLUSIONS:

This study identifies an antitumor strategy combining radiation and/or an immune checkpoint inhibitor with modulation of GABAARs in melanoma using benzodiazepine.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Linfócitos T / Receptores de GABA-A / Inibidores de Checkpoint Imunológico / Melanoma Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Linfócitos T / Receptores de GABA-A / Inibidores de Checkpoint Imunológico / Melanoma Idioma: En Ano de publicação: 2021 Tipo de documento: Article