MET inhibitor PHA-665752 suppresses the hepatocyte growth factor-induced cell proliferation and radioresistance in nasopharyngeal carcinoma cells

https://doi.org/10.1016/j.bbrc.2014.04.147Get rights and content

Highlights

  • We demonstrated that irradiation induced MET overexpression and activation.

  • The aberrant MET signal mediated by HGF induced proliferation and radioresistance of NPC cells.

  • MET inhibitor PHA-665752 effectively suppressed HGF induced cell proliferation and radioresistance in NPC cells.

  • PHA-665752 suppressed the three downstream pathway of HGF/MET signal in a dose-dependent manner.

Abstract

Although ionizing radiation (IR) has provided considerable improvements in nasopharyngeal carcinoma (NPC), in subsets of patients, radioresistance is still a major problem in the treatment. In this study, we demonstrated that irradiation induced MET overexpression and activation, and the aberrant MET signal mediated by hepatocyte growth factor (HGF) induced radioresistance. We also found that MET inhibitor PHA-665752 effectively suppressed HGF induced cell proliferation and radioresistance in NPC cells. Further investigation indicated that PHA-665752 suppressed the phosphorylation of the Akt, ERK1/2, and STAT3 proteins in a dose-dependent manner. Our data indicated that the combination of IR with a MET inhibitor, such as PHA-665752, might be a promising therapeutic strategy for NPC.

Introduction

Nasopharyngeal carcinoma (NPC) is the most common cancer of the head and neck, and it is highly prevalent and endemic in Southern China and Southeast Asia, with an annual incidence rate of approximately 20 cases per 100,000 people in endemic areas [1]. The epidemiological evidence implies that environmental risk factors, Epstein-Barr virus (EBV) infection and genetic susceptibility play crucial roles in the carcinogenesis of NPC [2]. Whereas radiation therapy (RT) has remained the mainstay of treatment for early stage NPC, after primary treatment with radiotherapy or chemo-irradiation, more than 30% of patients will relapse with local or regional recurrence or distant metastasis [3]. Concurrent or palliative chemotherapy is given with radiotherapy for the treatment of locally advanced disease, such as stage III and non-metastatic stage IV disease [4], [5]. The overall survival after recurrence is poor, with median survival ranging from 7.2 to 22 months [6], [7], [8]. Because of the advent of molecular targeted therapy over the past decade, some reagents such as the anti-EGFR antibody, cetuximab, and the anti-VEGF antibody, bevacizumab, have been employed against NPC [9], [10]. Increasing evidence has shown that the combination of molecular target reagents and radiation might provide a more effective therapeutic strategy for NPC. However, the influence of molecular targeted therapy to the radiosensitivity of NPC has rarely been investigated.

MET, a transmembrane receptor tyrosine kinase, consists of a 45-kD a and a 150-kD subunit [11]. HGF, the only known ligand, binds to the extracellular region, leading to receptor dimerization and phosphorylation of the intracellular tyrosine kinase domains [11]. The activation/phosphorylation of the intracellular domain prompts activation of downstream signaling, predominantly through the phosphatidylinositide 3-kinase (PI3K)/Akt, mitogen-activated protein kinase (MAPK), and signal transducers and activators of the transcription (STAT) pathways [11], [12], [13], [14]. MET has important roles in the malignant transformation of cancer cells [11]. Targeting MET has shown promise for the treatment of various types of cancer and is currently being tested in phase III trials for non-small cell lung cancer (NSCLC). In NPC, high MET expression was statistically significant for negative prognostic factors of survival [15].

Many studies have shown that MET is an attractive candidate for targeted anticancer therapy, frequently in combination with chemotherapy [16]. MET plays important roles in cancer metastasis and DNA damage and repair pathways [17], [18]. Studies have shown that MET was involved in the radioresistance of tumors such as prostate cancer and brain metastasis of breast cancer [19], [20]. Ionizing radiation induces overexpression and activity of the MET oncogene, which promotes cell invasion and protects cells from apoptosis, thus supporting radioresistance [21]. Since the radiotherapy is essential in the treatments of NPC, the role of MET inhibition to the radiosensitivity of NPC should be evaluated specifically.

In this study, we investigated the anti-tumor and radiation-sensitizing effects of MET inhibitor PHA-665752 in NPC cells. Our findings suggested that the combination of MET inhibition and radiation might lead to important clinical benefits in NPC and provide the basis for further development of a targeted therapeutic strategy for NPC.

Section snippets

Cell culture and reagents

The NPC cell line CNE-2 was kindly provided by Prof. Xia Yunfei (Sun Yat-sen University Cancer Center, Guangzhou, China). The CNE-1, HONE-1, SUNE-1 and NP69 cell lines were maintained in our lab. The human immortalized nasopharyngeal epithelial cell line NP69 was cultured in Keratinocyte/serum-free medium (Invitrogen, Carlsbad, CA, USA) containing bovine pituitary extract (BD biosciences, San Jose, CA, USA). The other human NPC cell lines were maintained in RPMI-1640 containing 10% fetal bovine

Irradiation induces overexpression and activation of MET in NPC cells

We first determined the expression levels of the total and phosphorylated MET in four NPC cell lines and NP69 cells. As shown in Fig. 1A, the expression of the total and phosphorylated MET was upregulated in NPC cell lines compared with NP69 cells, indicating a high level of activation of the MET pathway in NPC cells. Then, we assessed the effect of irradiation on the activation and expression of MET in the NPC cells. We selected CNE-1 and HONE-1 cells for this study and further investigation

Discussion

In this study, we demonstrated that IR induced the overexpression and activation of MET in NPC cells. We also found that HGF stimulated the proliferation and radioresistance of NPC cells by activating MET, which could be overcome by MET inhibitor PHA-665752. Further investigation indicated that PHA-665752 suppressed the phosphorylation of the Akt, ERK1/2, and STAT3 proteins in a dose-dependent manner. Our results suggest that the MET inhibitor PHA-665752 could effectively sensitizes NPC cells

Conflict of interest

The authors declare no conflict of interest.

Acknowledgments

We are grateful to Prof. Xia Yunfei for providing the NPC cell lines. This study was supported by a National Natural Science Foundation of China Grant (W. Wang, 81172243; and Q. Li, 81201961).

References (33)

  • G. Xie et al.

    Mammosphere cells from high-passage MCF7 cell line show variable loss of tumorigenicity and radioresistance

    Cancer Lett.

    (2012)
  • A. Sallmyr et al.

    Genomic instability in myeloid malignancies: increased reactive oxygen species (ROS), DNA double strand breaks (DSBs) and error-prone repair

    Cancer Lett.

    (2008)
  • R.E. Meyn et al.

    Receptor signaling as a regulatory mechanism of DNA repair

    Radiother. Oncol.

    (2009)
  • H.H. Loong et al.

    Update on the management and therapeutic monitoring of advanced nasopharyngeal cancer

    Hematol. Oncol. Clin. North Am.

    (2008)
  • H. Ali et al.

    Chemotherapy in advanced nasopharyngeal cancer

    Oncology (Williston Park)

    (2000)
  • F. Caponigro et al.

    Treatment approaches to nasopharyngeal carcinoma: a review

    Anticancer Drugs

    (2010)
  • Cited by (14)

    • Combination of HGF/MET-targeting agents and other therapeutic strategies in cancer

      2021, Critical Reviews in Oncology/Hematology
      Citation Excerpt :

      Early studies reported that pretreatment of tumor cells with HGF, considerably reduces DNA fragmentation and damage followed by exposure to DDAs through the PI3K/Akt pathway (Fan et al., 1998, 2000). Later studies demonstrated that inhibition of MET pathway with the small molecule PHA665752, suppressed HGF-induced phosphorylation of the ERK1/2, Akt, and STAT3, and enhanced the formation of DNA double-strand breaks (Liu et al., 2014). Additional support for potential crosstalk between MET and DDR pathways came from a study by Dai and colleagues which showed that MET inhibitor-induced apoptosis induction is mediated through the increase in DNA damage and regulation of some DDR–related proteins in primary effusion lymphoma cells (Dai et al., 2015a).

    • Co-targeting c-Met and DNA double-strand breaks (DSBs): Therapeutic strategies in BRCA-mutated gastric carcinomas

      2017, Biochimie
      Citation Excerpt :

      Combinations of c-Met-targeted therapy and irradiation, increased γH2AX tyrosine phosphorylation levels and pro-apoptotic kinase JNK1 levels, providing an additional link to c-Met inhibitors and DDR machinery [72,73]. Liu et al. have revealed that such combinations can suppress HGF-induced cell proliferation and phosphorylation of the ERK1/2, AKT, and STAT3 [74], enhance the formation of DNA double strand breaks and probably alleviate tumor hypoxia [75]. Bacco and his research team have confirmed that radiation influence the activity and expression levels of c-Met via the ATM-NF-κB signaling pathway resulted in cell invasion.

    View all citing articles on Scopus
    1

    These authors equally contributed to this work.

    View full text