BRD4 modulates vulnerability of triple-negative breast cancer to targeting of integrin-dependent signaling pathways

Cell Oncol (Dordr). 2020 Dec;43(6):1049-1066. doi: 10.1007/s13402-020-00537-1. Epub 2020 Oct 2.

Abstract

Purpose: Stemming from a myriad of genetic and epigenetic alterations, triple-negative breast cancer (TNBC) is tied to poor clinical outcomes and aspires for individualized therapies. Here we investigated the therapeutic potential of co-inhibiting integrin-dependent signaling pathway and BRD4, a transcriptional and epigenetic mediator, for TNBC.

Methods: Two independent patient cohorts were subjected to bioinformatic and IHC examination for clinical association of candidate cancer drivers. The efficacy and biological bases for co-targeting these drivers were interrogated using cancer cell lines, a protein kinase array, chemical inhibitors, RNAi/CRISPR/Cas9 approaches, and a 4 T1-Balb/c xenograft model.

Results: We found that amplification of the chromosome 8q24 region occurred in nearly 20% of TNBC tumors, and that it coincided with co-upregulation or amplification of c-Myc and FAK, a key effector of integrin-dependent signaling. This co-upregulation at the mRNA or protein level correlated with a poor patient survival (p < 0.0109 or p < 0.0402, respectively). Furthermore, we found that 14 TNBC cell lines exhibited high vulnerabilities to the combination of JQ1 and VS-6063, potent pharmacological antagonists of the BRD4/c-Myc and integrin/FAK-dependent pathways, respectively. We also observed a cooperative inhibitory effect of JQ1 and VS-6063 on tumor growth and infiltration of Ly6G+ myeloid-derived suppressor cells in vivo. Finally, we found that JQ1 and VS-6063 cooperatively induced apoptotic cell death by altering XIAP, Bcl2/Bcl-xl and Bim levels, impairing c-Src/p130Cas-, PI3K/Akt- and RelA-associated signaling, and were linked to EMT-inducing transcription factor Snail- and Slug-dependent regulation.

Conclusion: Based on our results, we conclude that the BRD4/c-Myc- and integrin/FAK-dependent pathways act in concert to promote breast cancer cell survival and poor clinical outcomes. As such, they represent promising targets for a synthetic lethal-type of therapy against TNBC.

Keywords: BRD4; FAK; Integrin; Targeted therapy; Triple-negative breast cancer; c-Myc.

MeSH terms

  • Azepines / pharmacology
  • Bcl-2-Like Protein 11 / metabolism
  • Benzamides / pharmacology
  • Cell Cycle Proteins / metabolism*
  • Cell Death / drug effects
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Focal Adhesion Protein-Tyrosine Kinases / genetics
  • Focal Adhesion Protein-Tyrosine Kinases / metabolism
  • Gene Expression Regulation, Neoplastic
  • Genome, Human
  • Humans
  • Integrins / metabolism*
  • Proto-Oncogene Proteins c-myc / genetics
  • Proto-Oncogene Proteins c-myc / metabolism
  • Pyrazines / pharmacology
  • RNA, Messenger / genetics
  • RNA, Messenger / metabolism
  • Signal Transduction* / drug effects
  • Sulfonamides / pharmacology
  • Transcription Factors / metabolism*
  • Triazoles / pharmacology
  • Triple Negative Breast Neoplasms / genetics
  • Triple Negative Breast Neoplasms / metabolism*
  • Triple Negative Breast Neoplasms / pathology

Substances

  • (+)-JQ1 compound
  • Azepines
  • BRD4 protein, human
  • Bcl-2-Like Protein 11
  • Benzamides
  • Cell Cycle Proteins
  • Integrins
  • Proto-Oncogene Proteins c-myc
  • Pyrazines
  • RNA, Messenger
  • Sulfonamides
  • Transcription Factors
  • Triazoles
  • defactinib
  • Focal Adhesion Protein-Tyrosine Kinases