학술논문

IDH mutant gliomas escape natural killer cell immune surveillance by downregulation of NKG2D ligand expression.
Document Type
article
Source
Neuro-Oncology. 18(10)
Subject
Biomedical and Clinical Sciences
Oncology and Carcinogenesis
Immunology
Cancer
Brain Disorders
Brain Cancer
Genetics
Rare Diseases
Neurosciences
2.1 Biological and endogenous factors
Aetiology
Antineoplastic Agents
Azacitidine
Brain Neoplasms
DNA Methylation
Decitabine
Down-Regulation
GPI-Linked Proteins
Gene Expression Regulation
Neoplastic
Glioma
Humans
Immunologic Surveillance
Intercellular Signaling Peptides and Proteins
Isocitrate Dehydrogenase
Killer Cells
Natural
Mutation
Real-Time Polymerase Chain Reaction
Transcriptome
Tumor Escape
IDH mutation
glioma
Natural Killer cells
NKG2D ligands
immune escape
immunotherapy
Oncology & Carcinogenesis
Oncology and carcinogenesis
Language
Abstract
BackgroundDiffuse gliomas are poorly immunogenic, fatal brain tumors. The basis for insufficient antitumor immunity in diffuse gliomas is unknown. Gain-of-function mutations in isocitrate dehydrogenases (IDH1 and IDH2) promote diffuse glioma formation through epigenetic reprogramming of a number of genes, including immune-related genes. Here, we identify epigenetic dysregulation of natural killer (NK) cell ligand genes as significant contributors to immune escape in glioma.MethodsWe analyzed the database of The Cancer Genome Atlas for immune gene expression patterns in IDH mutant or wild-type gliomas and identified differentially expressed immune genes. NKG2D ligand expression levels and NK cell-mediated lysis were measured in IDH mutant and wild-type patient-derived glioma stem cells and genetically engineered astrocytes. Finally, we assessed the impact of hypomethylating agent 5-aza-2'deoxycytodine (decitabine) as a potential NK cell sensitizing agent in IDH mutant cells.ResultsIDH mutant glioma stemlike cell lines exhibited significantly lower expression of NKG2D ligands compared with IDH wild-type cells. Consistent with these findings, IDH mutant glioma cells and astrocytes are resistant to NK cell-mediated lysis. Decitabine increases NKG2D ligand expression and restores NK-mediated lysis of IDH mutant cells in an NKG2D-dependent manner.ConclusionsIDH mutant glioma cells acquire resistance to NK cells through epigenetic silencing of NKG2D ligands ULBP1 and ULBP3. Decitabine-mediated hypomethylation restores ULBP1 and ULBP3 expression in IDH mutant glioma cells and may provide a clinically useful method to sensitize IDH mutant gliomas to NK cell-mediated immune surveillance in patients with IDH mutated diffuse gliomas.