Cyclic pentapeptide cRGDfK enhances the inhibitory effect of sunitinib on TGF-β1-induced epithelial-to-mesenchymal transition in human non-small cell lung cancer cells

PLoS One. 2020 Aug 18;15(8):e0232917. doi: 10.1371/journal.pone.0232917. eCollection 2020.

Abstract

In human lung cancer progression, the EMT process is characterized by the transformation of cancer cells into invasive forms that migrate to other organs. Targeting to EMT-related molecules is emerging as a novel therapeutic approach for the prevention of lung cancer cell migration and invasion. Traf2- and Nck-interacting kinase (TNIK) has recently been considered as an anti-proliferative target molecule to regulate the Wnt signaling pathway in several types of cancer cells. In the present study, we evaluated the inhibitory effect of a tyrosine kinase inhibitor sunitinib and the integrin-αⅤβ3 targeted cyclic peptide (cRGDfK) on EMT in human lung cancer cells. Sunitinib strongly inhibited the TGF-β1-activated EMT through suppression of Wnt signaling, Smad and non-Smad signaling pathways. In addition, the cRGDfK also inhibited the expression of TGFβ1-induced mesenchymal marker genes and proteins. The anti-EMT effect of sunitinib was enhanced when cRGDfK was treated together. When sunitinib was treated with cRGDfK, the mRNA and protein expression levels of mesenchymal markers were decreased compared to the treatment with sunitinib alone. Co-treatment of cRGDfK has shown the potential to improve the efficacy of anticancer agents in combination with therapeutic agents that may be toxic at high concentrations. These results provide new and improved therapies for treating and preventing EMT-related disorders, such as lung fibrosis and cancer metastasis, and relapse.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • A549 Cells
  • Adenosine Triphosphate / metabolism
  • Antineoplastic Combined Chemotherapy Protocols / administration & dosage
  • Biomarkers, Tumor / genetics
  • Biomarkers, Tumor / metabolism
  • Carcinoma, Non-Small-Cell Lung / drug therapy*
  • Carcinoma, Non-Small-Cell Lung / metabolism
  • Carcinoma, Non-Small-Cell Lung / pathology
  • Catalytic Domain
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Drug Synergism
  • Epithelial-Mesenchymal Transition / drug effects*
  • Epithelial-Mesenchymal Transition / genetics
  • Epithelial-Mesenchymal Transition / physiology
  • Humans
  • Integrin alphaVbeta3 / antagonists & inhibitors
  • Lung Neoplasms / drug therapy*
  • Lung Neoplasms / metabolism
  • Lung Neoplasms / pathology
  • Molecular Docking Simulation
  • Neoplasm Invasiveness / pathology
  • Neoplasm Invasiveness / prevention & control
  • Peptides, Cyclic / administration & dosage*
  • Protein Serine-Threonine Kinases / antagonists & inhibitors
  • Protein Serine-Threonine Kinases / chemistry
  • Protein Serine-Threonine Kinases / metabolism
  • Smad Proteins / metabolism
  • Sunitinib / administration & dosage*
  • Transforming Growth Factor beta1 / antagonists & inhibitors*
  • Wnt Signaling Pathway / drug effects

Substances

  • Biomarkers, Tumor
  • Integrin alphaVbeta3
  • Peptides, Cyclic
  • Smad Proteins
  • TGFB1 protein, human
  • Transforming Growth Factor beta1
  • cyclic (arginyl-glycyl-aspartyl-phenylalanyl-lysyl)
  • Adenosine Triphosphate
  • Protein Serine-Threonine Kinases
  • TNIK protein, human
  • Sunitinib

Grants and funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (Ministry of Science and NCT; 2020R1A2C1006416 and Ministry of Education; 2017R1D1A1B03027968). This work has not received any financial investment by CHA Meditech, Co., Ltd. The funder provided support in the form of salaries for authors [KYP], but did not have any additional role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. The specific roles of these authors are articulated in the ‘Author Contributions’ section.