The Bromodomain Inhibitor JQ1 and the Histone Deacetylase Inhibitor Panobinostat Synergistically Reduce N-Myc Expression and Induce Anticancer Effects

Clin Cancer Res. 2016 May 15;22(10):2534-44. doi: 10.1158/1078-0432.CCR-15-1666. Epub 2016 Jan 5.

Abstract

Purpose: Patients with neuroblastoma associated with MYCN oncogene amplification experience a very poor prognosis. BET bromodomain inhibitors are among the most promising novel anticancer agents as they block BRD3 and BRD4 from activating oncogene transcription. However, treatment with BET bromodomain inhibitors alone does not result in cancer remission in many murine models.

Experimental design: MYCN-amplified neuroblastoma cells were treated with vehicle control, the BET bromodomain inhibitor JQ1, the histone deacetylase inhibitor panobinostat, or the combination of JQ1 and panobinostat. Genes modulated by JQ1, panobinostat, or the combination therapy were identified by Affymetrix microarray, and cell proliferation and apoptosis were examined by Alamar blue assays and flow cytometry analysis. Modulation of LIN28B promoter activity by BRD3 and BRD4 was examined by chromatin immunoprecipitation and luciferase assays. In addition, neuroblastoma-bearing mice were treated with vehicle control, JQ1, and/or panobinostat.

Results: LIN28B was one of the top genes synergistically reduced by JQ1 and panobinostat. BRD3 and BRD4 directly bound to the LIN28B gene promoter and activated LIN28B gene transcription, and knocking down LIN28B reduced the expression of N-Myc protein, but not N-Myc mRNA. JQ1 and panobinostat synergistically reduced LIN28B gene and N-Myc protein expression, and synergistically induced growth inhibition and apoptosis in neuroblastoma cells, but not normal nonmalignant cells in vitro In neuroblastoma-bearing mice, JQ1 and panobinostat synergistically and considerably reduced N-Myc protein expression in tumor tissues and blocked tumor progression.

Conclusions: Our findings have identified a novel strategy to reduce the N-Myc oncoprotein expression and a novel therapeutic approach for the treatment of aggressive neuroblastoma. Clin Cancer Res; 22(10); 2534-44. ©2016 AACR.

MeSH terms

  • Animals
  • Antineoplastic Agents / pharmacology*
  • Apoptosis / drug effects
  • Azepines / pharmacology*
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Female
  • Histone Deacetylase Inhibitors / pharmacology*
  • Humans
  • Hydroxamic Acids / pharmacology*
  • Indoles / pharmacology*
  • Mice
  • Mice, Inbred BALB C
  • Neuroblastoma / drug therapy
  • Neuroblastoma / metabolism
  • Nuclear Proteins / metabolism
  • Oncogene Proteins / metabolism
  • Panobinostat
  • Promoter Regions, Genetic / drug effects
  • Proteins / antagonists & inhibitors*
  • Proto-Oncogene Proteins c-myc / metabolism*
  • Transcription, Genetic / drug effects
  • Triazoles / pharmacology*
  • Xenograft Model Antitumor Assays / methods

Substances

  • (+)-JQ1 compound
  • Antineoplastic Agents
  • Azepines
  • Histone Deacetylase Inhibitors
  • Hydroxamic Acids
  • Indoles
  • Nuclear Proteins
  • Oncogene Proteins
  • Proteins
  • Proto-Oncogene Proteins c-myc
  • Triazoles
  • bromodomain and extra-terminal domain protein, human
  • Panobinostat