Full length article
YC-1 potentiates the antitumor activity of gefitinib by inhibiting HIF-1α and promoting the endocytic trafficking and degradation of EGFR in gefitinib-resistant non-small-cell lung cancer cells

https://doi.org/10.1016/j.ejphar.2020.172961Get rights and content

Abstract

The tyrosine kinase inhibitor (TKI) gefitinib exerts good therapeutic effect on NSCLC patients with sensitive EGFR-activating mutations. However, most patients ultimately relapse due to the development of drug resistance after 6–12 months of treatment. Here, we showed that a HIF-1α inhibitor, YC-1, potentiated the antitumor efficacy of gefitinib by promoting EGFR degradation in a panel of human NSCLC cells with wild-type or mutant EGFRs. YC-1 alone had little effect on NSCLC cell survival but significantly enhanced the antigrowth and proapoptotic effects of gefitinib. In insensitive NSCLC cell lines, gefitinib efficiently inhibited the phosphorylation of EGFR but not the downstream signaling of ERK, AKT and STAT3; however, when combined with YC-1 treatment, these signaling pathways were strongly impaired. Gefitinib treatment induced EGFR arrest in the early endosome, and YC-1 treatment promoted delayed EGFR transport into the late endosome as well as receptor degradation. Moreover, the YC-1-induced reduction of HIF-1α protein was associated with the enhancement of EGFR degradation. HIF-1α knockdown promoted EGFR degradation, showing synergistic antigrowth and proapoptotic effects similar to those of the gefitinib and YC-1 combination treatment in NSCLC cells. Our findings provide a novel combination treatment strategy with gefitinib and YC-1 to extend the usage of gefitinib and overcome gefitinib resistance in NSCLC patients.

Introduction

Gefitinib is a well-known first-generation EGFR tyrosine kinase inhibitor (EGFR-TKI) that exhibits good antitumor activity in molecularly selected NSCLC patients with L858R/del19 mutations of EGFR (Rusch et al., 1997; Brabender et al., 2001; Jemal et al., 2011; Ferlay et al., 2013). However, despite the initial response and excellent disease control with EGFR-TKI therapy, acquired resistance is inevitable and raises an immense challenge in NSCLC therapy (Dancey, 2004; Perez-Soler et al., 2004; Pao and Chmielecki, 2010; Enting and Spicer, 2012; Rosell and Karachaliou, 2016). Among the mechanisms of acquired resistance, the emergence of a second substitution, T790M in exon 20 of EGFR, was responsible for approximately 50–60% of gefitinib-resistant cases (Sequist et al., 2011). The third-generation TKI osimertinib was approved by the FDA for its enhanced ability to bind and inhibit EGFR/T790M; however, new acquired resistance has been developed in a subset of patients with mutations, such as C797S (Thress et al., 2015). Moreover, many NSCLC patients who overexpress wild-type EGFR usually respond poorly to EGFR-TKI treatment (Stinchcom, 2016). This evidence highlights that innovative strategies are urgently needed to overcome both primary and acquired resistance to EGFR-TKIs in NSCLC patients.

EGFR-TKIs can efficiently inhibit the tyrosine kinase activity of EGFR, thus suppressing the unrestrained proliferation of cancer cells. However, accumulating evidence suggests that EGFR has many other functions beyond kinase activity that have been known to play an essential role in cancer pathology (Coker et al., 1994; Ewald et al., 2003). Many EGFR-dependent NSCLC patients display innate or acquired resistance to EGFR-TKI treatment despite the efficient inhibition of tyrosine kinase activity (Zhang et al., 2008; Tan et al., 2016). Studies have shown that EGFR-TKI treatment induced cellular stress and provoked noncanonical pathways of EGFR trafficking and signaling, which provides cancer cells with a survival advantage (Filosto et al., 2011; Orcutt et al., 2011; Filomeni, 2015; Zou et al., 2013). After long-term TKI treatment, the intracellular signaling of EGFR was prone to be shifted from its kinase-dependent to kinase-independent pattern, which was involved in the acquisition of TKI resistance (Engelman and Janne, 2008). Tan et al. reported that EGFR-TKIs elicited endosomal accumulation of inactive EGFR to initiate autophagy in a kinase-independent fashion, providing a survival advantage and facilitating TKI resistance in NSCLCs with wild-type EGFR (Tan et al., 2015). Menard et al. showed that the inhibition of kinase-independent EGFR signaling overcame TKI resistance in NSCLC cells with different EGFR mutations (Ménard et al., 2018). Inspired by these observations, we believe that cotargeting EGFR kinase-dependent and -independent functions may hold new promise for treating EGFR-TKI resistant cancers.

YC-1, 3-(5-hydroxymethyl-2-furyl)-1-benzyl indazole, was initially described as an activator of soluble guanylyl cyclase (sGC), prompting antiplatelet aggregation and vascular relaxation (Galle et al., 1999). More recent studies revealed the ability of YC-1 to inhibit tumor growth, suppress angiogenesis and enhance antitumor effects of radiation; these abilities have been associated with its activity to inhibit hypoxia-inducible factor 1α (HIF-1α) (Chun et al., 2004; Chen et al., 2008).

Recently, many studies showed that YC-1 could increase the sensitivity to cisplatin in cisplatin-resistant cancer cells and overcome the radioresistance of hypoxic cancer cells (Moon et al., 2009; Kong et al., 2014; Lee et al., 2017b; Tuttle et al., 2017). However, the detailed mechanism of YC-1 antitumor activity is largely unknown. In this study, we found for the first time that YC-1 exhibited an unexpected ability to sensitize gefitinib-resistant NSCLC cells to gefitinib treatment. YC-1 was capable of regulating the trafficking and degradation of EGFR protein by reducing HIF-1α protein levels and downregulating kinase-independent EGFR signaling in NSCLC cells.

Section snippets

Chemicals and cell lines

Gefitinib (Iressa) was purchased from Selleck Chemicals (Houston, TX, USA). YC-1 and E64D were purchased from Sigma-Aldrich (St. Louis, MO, USA). The compounds were dissolved in dimethyl sulfoxide (DMSO) at 100 mM, aliquoted and stored at −20 °C until use. The final working concentration of DMSO (plus compound) in all working assays was lower than 0.5%. All cell lines were purchased from American Typical Cell Collection (ATCC, Manassas, VA, USA). The cells were maintained in RPMI 1640 medium

YC-1 synergistically enhances gefitinib-induced growth inhibition in NSCLC cell lines with wild-type or L858R/T790M mutated EGFR

To identify the potential hits that increase the sensitivity of NSCLC cells to gefitinib, the LOPAC library from Sigma of 1280 pharmacologically active compounds was screened in the combination assay. Among these compounds, a HIF-1α inhibitor, YC-1, significantly decreased the cell viability of NCI-H226 cells at a concentration of 5 μM in the presence of gefitinib (5 μM, data not shown). Dose response curves were established to determine the IC50 values for gefitinib and YC-1 in two

Discussion

Innate and acquired resistance to gefitinib and other EGFR TKI therapies has become a great clinical burden in NSCLC therapy (Thress et al., 2015; Takeda and Nakagawa, 2019). Recently, the existence and activation of ligand-independent EGFR signaling pathways and functions has been proposed as one of the potential mechanisms that contributes to the emergence of innate and acquired resistance to EGFR TKIs (Coker et al., 1994; Ewald et al., 2003; Zhang et al., 2008; Tan et al., 2016).

CRediT authorship contribution statement

Hui Hu: Conceptualization, Investigation, Methodology, Formal analysis, Writing - original draft. Xiao-Kang Miao: Investigation, Methodology, Formal analysis, Funding acquisition. Jing-Yi Li: Conceptualization, Investigation, Methodology. Xiao-Wei Zhang: Investigation, Methodology. Jing-Jie Xu: Investigation, Methodology. Jing-Ying Zhang: Investigation, Methodology. Tian-Xiong Zhou: Investigation, Methodology. Ming-Ning Hu: Investigation, Methodology, Formal analysis. Wen-Le Yang: Formal

Declaration of competing interest

The authors declare that they have no conflicts of interest with the contents of this article.

Acknowledgements

We would like to acknowledge financial support from the National Natural Science Foundation of China (NSFC, Grant No. 81874315, 81302798, 21432003), the Program for Chang-jiang Scholars and Innovative Research Team in University (PCSIRT: No. IRT_15R27) and the Fundamental Research Funds for the Central Universities (lzujbky-2018-k9, lzujbky-2018-87).

References (58)

  • R. Rosell et al.

    Large-scale screening for somatic mutations in lung cancer

    Lancet

    (2016)
  • D.E. Swinson et al.

    Interactions between hypoxia and epidermal growth factor receptor in non–small-cell lung cancer

    Clin. Lung Canc.

    (2006)
  • X. Tan et al.

    Stress-induced EGFR trafficking: mechanisms, functions, and therapeutic implications

    Trends Cell Biol.

    (2016)
  • X. Tan et al.

    A kinase-independent role for EGF receptor in autophagy initiation

    Cell

    (2015)
  • A. Tomas et al.

    EGF receptor trafficking: consequences for signaling and cancer

    Trends Cell Biol.

    (2014)
  • T.R. Tuttle et al.

    Soluble guanylate cyclase stimulators increase sensitivity to cisplatin in head and neck squamous cell carcinoma cells

    Canc. Lett.

    (2017)
  • W. Yuan et al.

    CMTM3 decreases EGFR expression and EGF-mediated tumorigenicity by promoting Rab5 activity in gastric cancer

    Canc. Lett.

    (2017)
  • Y. Zou et al.

    The autophagy inhibitor chloroquine overcomes the innate resistance to erlotinib of non-small cell lung cancercells with wild-type EGFR

    J. Thorac. Oncol.

    (2013)
  • L. Bazzani et al.

    PGE2 mediates EGFR internalization and nuclear translocation via caveolin endocytosis promoting its transcriptional activity and proliferation in human NSCLC cells

    Oncotarget

    (2018)
  • J. Brabender et al.

    Epidermal growth factor receptor and HER2-neu mRNA expression in non-small cell lung cancer Is correlated with survival

    Clin. Canc. Res.

    (2001)
  • B.P. Ceresa et al.

    Molecular mechanisms that regulate epidermal growth factor receptor inactivation

    Clin. Med. Oncol.

    (2008)
  • P.H. Chen et al.

    Crosstalk between CLCb/Dyn1-mediated adaptive clathrin-mediated endocytosis and epidermal growth factor receptor signaling increases metastasis

    Dev. Cell

    (2017)
  • K.H. Cho et al.

    A ROS/STAT3/HIF-1α signaling cascade mediates EGF-induced TWIST1 expression and prostate cancer cell invasion

    Prostate

    (2014)
  • K.J. Coker et al.

    A kinase-negative epidermal growth factor receptor that retains the capacity to stimulate DNA synthesis

    Proc. Natl. Acad. Sci. U.S.A.

    (1994)
  • N. De Paulsen et al.

    Role of transforming growth factor-alpha in von Hippel-Lindau (VHL)-/-clear cell renal carcinoma cell proliferation: a possible mechanism coupling VHL tumor suppressor inactivation and tumorigenesis

    Proc. Natl. Acad. Sci. U.S.A.

    (2001)
  • J.A. Engelman et al.

    Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer

    Clin. Canc. Res.

    (2008)
  • D. Enting et al.

    EGFR inhibition and more: a new generation growing up

    J. Thorac. Dis.

    (2012)
  • J.A. Ewald et al.

    Ligand-and kinase activity-independent cell survival mediated by the epidermal growth factor receptor expressed in 32D cells

    Exp. Cell Res.

    (2003)
  • G. Filomeni

    Oxidative stress and autophagy: the clash between damage and metabolic needs

    Cell Death Differ.

    (2015)
  • Cited by (25)

    • Synthesis and evaluation of biarylquinoline derivatives as novel HIF-1α inhibitors

      2022, Bioorganic Chemistry
      Citation Excerpt :

      Increased HIF-1α expression stimulates glycolysis [3], angiogenesis [4], erythropoiesis [5], and metastasis [6,7], which enables tumor cells to adapt to the hypoxic environments [8,9] to obtain nutrition. Therefore, targeting HIF-1α has emerged as an attractive strategy for cancer therapy in recent years [10-14]. Multiple signaling pathways have been reported to regulate the expression of HIF-1α through distinct mechanisms (Fig. 1).

    • Inhibition of autophagy by YC-1 promotes gefitinib induced apoptosis by targeting FOXO1 in gefitinib-resistant NSCLC cells

      2021, European Journal of Pharmacology
      Citation Excerpt :

      Differences were considered statistically significant at P < 0.05. In our previous study, we observed that YC-1 could significantly potentiate the antitumor activity of gefitinib by promoting endocytic trafficking and degradation of EGFR in gefitinib-resistant NSCLC cells (Hu et al., 2020). The endosomal arrest of EGFR induced by EGFR TKIs is associated with the initiation of cytoprotective autophagy (Tan et al., 2016).

    View all citing articles on Scopus
    View full text