Combination of gefitinib and DNA methylation inhibitor decitabine exerts synergistic anti-cancer activity in colon cancer cells

PLoS One. 2014 May 29;9(5):e97719. doi: 10.1371/journal.pone.0097719. eCollection 2014.

Abstract

Despite recent advances in the treatment of human colon cancer, the chemotherapy efficacy against colon cancer is still unsatisfactory. In the present study, effects of concomitant inhibition of the epidermal growth factor receptor (EGFR) and DNA methyltransferase were examined in human colon cancer cells. We demonstrated that decitabine (a DNA methyltransferase inhibitor) synergized with gefitinib (an EGFR inhibitor) to reduce cell viability and colony formation in SW1116 and LOVO cells. However, the combination of the two compounds displayed minimal toxicity to NCM460 cells, a normal human colon mucosal epithelial cell line. The combination was also more effective at inhibiting the AKT/mTOR/S6 kinase pathway. In addition, the combination of decitabine with gefitinib markedly inhibited colon cancer cell migration. Furthermore, gefitinib synergistically enhanced decitabine-induced cytotoxicity was primarily due to apoptosis as shown by Annexin V labeling that was attenuated by z-VAD-fmk, a pan caspase inhibitor. Concomitantly, cell apoptosis resulting from the co-treatment of gefitinib and decitabine was accompanied by induction of BAX, cleaved caspase 3 and cleaved PARP, along with reduction of Bcl-2 compared to treatment with either drug alone. Interestingly, combined treatment with these two drugs increased the expression of XIAP-associated factor 1 (XAF1) which play an important role in cell apoptosis. Moreover, small interfering RNA (siRNA) depletion of XAF1 significantly attenuated colon cancer cells apoptosis induced by the combination of the two drugs. Our findings suggested that gefitinib in combination with decitabine exerted enhanced cell apoptosis in colon cancer cells were involved in mitochondrial-mediated pathway and induction of XAF1 expression. In conclusion, based on the observations from our study, we suggested that the combined administration of these two drugs might be considered as a novel therapeutic regimen for treating colon cancer.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adaptor Proteins, Signal Transducing
  • Antineoplastic Agents / pharmacology*
  • Apoptosis / drug effects
  • Apoptosis Regulatory Proteins / genetics
  • Apoptosis Regulatory Proteins / metabolism
  • Azacitidine / analogs & derivatives*
  • Azacitidine / pharmacology
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Colonic Neoplasms / genetics*
  • Colonic Neoplasms / metabolism
  • DNA Methylation / drug effects*
  • Decitabine
  • Dose-Response Relationship, Drug
  • Drug Synergism
  • Gefitinib
  • Humans
  • Intracellular Signaling Peptides and Proteins / genetics
  • Intracellular Signaling Peptides and Proteins / metabolism
  • Neoplasm Proteins / genetics
  • Neoplasm Proteins / metabolism
  • Protein Kinase Inhibitors / pharmacology
  • Proto-Oncogene Proteins c-akt / metabolism
  • Quinazolines / pharmacology*
  • Signal Transduction / drug effects
  • TOR Serine-Threonine Kinases / metabolism

Substances

  • Adaptor Proteins, Signal Transducing
  • Antineoplastic Agents
  • Apoptosis Regulatory Proteins
  • Intracellular Signaling Peptides and Proteins
  • Neoplasm Proteins
  • Protein Kinase Inhibitors
  • Quinazolines
  • XAF1 protein, human
  • Decitabine
  • Proto-Oncogene Proteins c-akt
  • TOR Serine-Threonine Kinases
  • Azacitidine
  • Gefitinib

Grants and funding

The work was supported by the South China Normal University Science Fund for Young Scholars (Grant No. 13KJ19 to Zheng-zhi Zou), the Bureau of Dongguan city science and Technology 2010 key project Fund (Grant No. 201028 to Xiao-yong Luo). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.