Your browser doesn't support javascript.
loading
Development of novel SUV39H2 inhibitors that exhibit growth suppressive effects in mouse xenograft models and regulate the phosphorylation of H2AX.
Vougiouklakis, Theodore; Saloura, Vassiliki; Park, Jae-Hyun; Takamatsu, Naofumi; Miyamoto, Takashi; Nakamura, Yusuke; Matsuo, Yo.
Affiliation
  • Vougiouklakis T; Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA.
  • Saloura V; Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA.
  • Park JH; Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA.
  • Takamatsu N; OncoTherapy Science Inc., Kawasaki, Japan.
  • Miyamoto T; OncoTherapy Science Inc., Kawasaki, Japan.
  • Nakamura Y; Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA.
  • Matsuo Y; Department of Surgery, The University of Chicago, Chicago, IL, USA.
Oncotarget ; 9(61): 31820-31831, 2018 Aug 07.
Article in En | MEDLINE | ID: mdl-30159125
ABSTRACT
Protein methyltransferase SUV39H2 was reported to methylate histone H2AX at lysine 134 and enhance the formation of phosphorylated H2AX (γ-H2AX), which causes chemoresistance of cancer cells. We found that a series of imidazo[1,2-a]pyridine compounds that we synthesized could inhibit SUV39H2 methyltransferase activity. One of the potent compounds, OTS193320, was further analyzed in in vitro studies. The compound decreased global histone H3 lysine 9 tri-methylation levels in breast cancer cells and triggered apoptotic cell death. Combination of OTS193320 with doxorubicin (DOX) resulted in reduction of γ-H2AX levels as well as cancer cell viability compared to a single agent OTS193320 or DOX. Further optimization of inhibitors and their in vivo analysis identified a compound, OTS186935, which revealed significant inhibition of tumor growth in mouse xenograft models using MDA-MB-231 breast cancer cells and A549 lung cancer cells without any detectable toxicity. Our results suggest that the SUV39H2 inhibitors sensitize cancer cells to DOX by reduction of γ-H2AX levels in cancer cells, and collectively demonstrate that SUV39H2 inhibition warrants further investigation as a novel anti-cancer therapy.
Key words