1. Academic Validation
  2. The SMARCA2/4 ATPase Domain Surpasses the Bromodomain as a Drug Target in SWI/SNF-Mutant Cancers: Insights from cDNA Rescue and PFI-3 Inhibitor Studies

The SMARCA2/4 ATPase Domain Surpasses the Bromodomain as a Drug Target in SWI/SNF-Mutant Cancers: Insights from cDNA Rescue and PFI-3 Inhibitor Studies

  • Cancer Res. 2015 Sep 15;75(18):3865-3878. doi: 10.1158/0008-5472.CAN-14-3798.
Bhavatarini Vangamudi 1 Thomas A Paul  # 2 Parantu K Shah  # 1 Maria Kost-Alimova 1 Lisa Nottebaum 2 Xi Shi 1 Yanai Zhan 1 Elisabetta Leo 1 Harshad S Mahadeshwar 1 Alexei Protopopov 1 Andrew Futreal 3 Trang N Tieu 1 Mike Peoples 1 Timothy P Heffernan 1 Joseph R Marszalek 1 Carlo Toniatti 1 Alessia Petrocchi 1 Dominique Verhelle 2 Dafydd R Owen 4 Giulio Draetta 1 Philip Jones 1 Wylie S Palmer 1 Shikhar Sharma 2 Jannik N Andersen 1
Affiliations

Affiliations

  • 1 Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, TX.
  • 2 Pfizer Oncology Research Unit, La Jolla, CA.
  • 3 Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, TX.
  • 4 Pfizer Worldwide Medicinal Chemistry, Cambridge, MA.
  • # Contributed equally.
Abstract

The SWI/SNF multisubunit complex modulates chromatin structure through the activity of two mutually exclusive catalytic subunits, SMARCA2 and SMARCA4, which both contain a bromodomain and an ATPase domain. Using RNAi, cancer-specific vulnerabilities have been identified in SWI/SNF-mutant tumors, including SMARCA4-deficient lung cancer; however, the contribution of conserved, druggable protein domains to this Anticancer phenotype is unknown. Here, we functionally deconstruct the SMARCA2/4 paralog dependence of Cancer cells using bioinformatics, genetic, and pharmacologic tools. We evaluate a selective SMARCA2/4 bromodomain inhibitor (PFI-3) and characterize its activity in chromatin-binding and cell-functional assays focusing on cells with altered SWI/SNF complex (e.g., lung, synovial sarcoma, leukemia, and rhabdoid tumors). We demonstrate that PFI-3 is a potent, cell-permeable probe capable of displacing ectopically expressed, GFP-tagged SMARCA2-bromodomain from chromatin, yet contrary to target knockdown, the inhibitor fails to display an antiproliferative phenotype. Mechanistically, the lack of pharmacologic efficacy is reconciled by the failure of bromodomain inhibition to displace endogenous, full-length SMARCA2 from chromatin as determined by in situ cell extraction, chromatin immunoprecipitation, and target gene expression studies. Furthermore, using inducible RNAi and cDNA complementation (bromodomain- and ATPase-dead constructs), we unequivocally identify the ATPase domain, and not the bromodomain of SMARCA2, as the relevant therapeutic target with the catalytic activity suppressing defined transcriptional programs. Taken together, our complementary genetic and pharmacologic studies exemplify a general strategy for multidomain protein drug-target validation and in case of SMARCA2/4 highlight the potential for drugging the more challenging helicase/ATPase domain to deliver on the promise of synthetic-lethality therapy.

Figures
Products