BET protein inhibition sensitizes glioblastoma cells to temozolomide treatment by attenuating MGMT expression

Cell Death Dis. 2022 Dec 13;13(12):1037. doi: 10.1038/s41419-022-05497-y.

Abstract

Bromodomain and extra-terminal tail (BET) proteins have been identified as potential epigenetic targets in cancer, including glioblastoma. These epigenetic modifiers link the histone code to gene transcription that can be disrupted with small molecule BET inhibitors (BETi). With the aim of developing rational combination treatments for glioblastoma, we analyzed BETi-induced differential gene expression in glioblastoma derived-spheres, and identified 6 distinct response patterns. To uncover emerging actionable vulnerabilities that can be targeted with a second drug, we extracted the 169 significantly disturbed DNA Damage Response genes and inspected their response pattern. The most prominent candidate with consistent downregulation, was the O-6-methylguanine-DNA methyltransferase (MGMT) gene, a known resistance factor for alkylating agent therapy in glioblastoma. BETi not only reduced MGMT expression in GBM cells, but also inhibited its induction, typically observed upon temozolomide treatment. To determine the potential clinical relevance, we evaluated the specificity of the effect on MGMT expression and MGMT mediated treatment resistance to temozolomide. BETi-mediated attenuation of MGMT expression was associated with reduction of BRD4- and Pol II-binding at the MGMT promoter. On the functional level, we demonstrated that ectopic expression of MGMT under an unrelated promoter was not affected by BETi, while under the same conditions, pharmacologic inhibition of MGMT restored the sensitivity to temozolomide, reflected in an increased level of γ-H2AX, a proxy for DNA double-strand breaks. Importantly, expression of MSH6 and MSH2, which are required for sensitivity to unrepaired O6-methylguanine-lesions, was only briefly affected by BETi. Taken together, the addition of BET-inhibitors to the current standard of care, comprising temozolomide treatment, may sensitize the 50% of patients whose glioblastoma exert an unmethylated MGMT promoter.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Antineoplastic Agents, Alkylating / pharmacology
  • Cell Cycle Proteins / metabolism
  • Cell Line, Tumor
  • DNA / metabolism
  • DNA Methylation / genetics
  • DNA Modification Methylases / genetics
  • DNA Modification Methylases / metabolism
  • DNA Repair Enzymes / genetics
  • DNA Repair Enzymes / metabolism
  • Dacarbazine / pharmacology
  • Dacarbazine / therapeutic use
  • Gene Expression Regulation, Neoplastic
  • Glioblastoma* / drug therapy
  • Glioblastoma* / genetics
  • Glioblastoma* / pathology
  • Humans
  • Nuclear Proteins / metabolism
  • O(6)-Methylguanine-DNA Methyltransferase / genetics
  • O(6)-Methylguanine-DNA Methyltransferase / metabolism
  • O(6)-Methylguanine-DNA Methyltransferase / therapeutic use
  • Temozolomide / pharmacology
  • Temozolomide / therapeutic use
  • Transcription Factors / metabolism

Substances

  • Temozolomide
  • Dacarbazine
  • Nuclear Proteins
  • Antineoplastic Agents, Alkylating
  • Transcription Factors
  • DNA Modification Methylases
  • O(6)-Methylguanine-DNA Methyltransferase
  • DNA Repair Enzymes
  • DNA
  • BRD4 protein, human
  • Cell Cycle Proteins
  • MGMT protein, human