Src Inhibition Can Synergize with Gemcitabine and Reverse Resistance in Triple Negative Breast Cancer Cells via the AKT/c-Jun Pathway

PLoS One. 2016 Dec 30;11(12):e0169230. doi: 10.1371/journal.pone.0169230. eCollection 2016.

Abstract

Purpose: Gemcitabine-based chemotherapy remains one of the standards in management of metastatic breast cancer. However, intrinsic and acquired resistance to gemcitabine inevitably occurs. The aims of this study were to assess the efficacy of the combination of src inhibition and gemcitabine in gemcitabine-resistant breast cancer cells.

Methods and results: By using colony formation, sphere forming, flow cytometry, cell counting kit-8 and transwell assays, 231/GEM-res (gemcitabine-resistant) cell line, which was 10 times more resistant, was shown to have elevated drug tolerance, enhanced proliferative and self-renewal abilities, compared with its parental cells. Inhibition of src by both saracatinib (AZD0530) and siRNA could partially reverse gemcitabine resistance and attenuate resistance-associated anti-apoptosis, migration and stem cell capacities. In addition, the combination of src inhibition and gemcitabine had synergistic antitumor effects. Western blot analysis revealed up-regulation of pro-apoptotic protein BAX, along with the down-regulation of anti-apoptotic proteins (BCL-XL, Survivin), migration associated proteins (p-FAK, MMP-3) and cancer stem cell (CSC) markers (CD44, Oct-4), which was probably mediated by AKT/c-Jun pathway.

Conclusion: In highly gemcitabine-resistant 231 cells, src inhibition can synergize with gemcitabine, reverse drug resistance, inhibit tumor growth/metastasis/stemness of cancer stem cells, possibly via the AKT/c-Jun pathway.

MeSH terms

  • Antimetabolites, Antineoplastic / pharmacology*
  • Antineoplastic Combined Chemotherapy Protocols / pharmacology
  • Apoptosis / drug effects
  • Benzodioxoles / pharmacology*
  • Cell Movement / drug effects
  • Cell Proliferation / drug effects
  • Deoxycytidine / analogs & derivatives*
  • Deoxycytidine / pharmacology
  • Drug Resistance, Neoplasm / genetics*
  • Drug Synergism
  • Enzyme Inhibitors / pharmacology*
  • Female
  • Focal Adhesion Kinase 1 / biosynthesis
  • Gemcitabine
  • Gene Expression Regulation, Neoplastic
  • Humans
  • Hyaluronan Receptors / biosynthesis
  • Inhibitor of Apoptosis Proteins / biosynthesis
  • JNK Mitogen-Activated Protein Kinases / metabolism*
  • Matrix Metalloproteinase 3 / biosynthesis
  • Octamer Transcription Factor-3 / biosynthesis
  • Proto-Oncogene Proteins c-akt / metabolism*
  • Quinazolines / pharmacology*
  • RNA Interference
  • RNA, Small Interfering / genetics
  • Survivin
  • Triple Negative Breast Neoplasms / drug therapy*
  • Triple Negative Breast Neoplasms / genetics
  • Triple Negative Breast Neoplasms / pathology
  • Tumor Cells, Cultured
  • bcl-2-Associated X Protein / biosynthesis
  • bcl-X Protein / biosynthesis
  • src-Family Kinases / antagonists & inhibitors*
  • src-Family Kinases / genetics

Substances

  • Antimetabolites, Antineoplastic
  • BAX protein, human
  • BCL2L1 protein, human
  • BIRC5 protein, human
  • Benzodioxoles
  • CD44 protein, human
  • Enzyme Inhibitors
  • Hyaluronan Receptors
  • Inhibitor of Apoptosis Proteins
  • Octamer Transcription Factor-3
  • POU5F1 protein, human
  • Quinazolines
  • RNA, Small Interfering
  • Survivin
  • bcl-2-Associated X Protein
  • bcl-X Protein
  • Deoxycytidine
  • saracatinib
  • Focal Adhesion Kinase 1
  • PTK2 protein, human
  • src-Family Kinases
  • Proto-Oncogene Proteins c-akt
  • JNK Mitogen-Activated Protein Kinases
  • MMP3 protein, human
  • Matrix Metalloproteinase 3
  • Gemcitabine

Grants and funding

Xi-Chun Hu received the support from “an Investigator Sponsored Study Programme” of AstraZeneca. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.