Association between the HGF/c‑MET signaling pathway and tumorigenesis, progression and prognosis of hepatocellular carcinoma (Review)

  • Authors:
    • Wei Meng
    • Tao Chen
  • View Affiliations

  • Published online on: July 13, 2021     https://doi.org/10.3892/or.2021.8142
  • Article Number: 191
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is one of the most aggressive and lethal malignancies with a rising incidence, and is characterized by rapid progression, frequent metastasis, late diagnosis, high postoperative recurrence and poor prognosis. Therefore, novel treatment strategies for HCC, particularly advanced HCC, are urgently required. The hepatocyte growth factor (HGF)/c‑mesenchymal‑epithelial transition receptor (c‑MET) axis is a key signaling pathway in HCC and is strongly associated with its highly malignant features. Available treatments based on HGF/c‑MET inhibition may prolong the lifespan of patients with HCC; however, they do not achieve the desired therapeutic effects. The aim of the present article was to review the basic knowledge regarding the role of the HGF/c‑MET signaling pathway in HCC, and examine the association between the HGF/c‑MET signaling pathway and the tumorigenesis, progression and prognosis of HCC.

Introduction

The liver is a critical organ involved in numerous physiological processes, including immune system support, blood volume regulation, endocrine control of growth signaling pathways and macronutrient metabolism. Due to the livers unique metabolism and relationship with the gastrointestinal tract, hepatocytes are exposed to several toxins, which may result in several anomalies, such as primary liver cancer (1). Primary liver cancer comprises hepatocellular carcinoma (HCC; 75–85%) and intrahepatic cholangiocarcinoma (10–15%), as well as other rare types (2). HCC, the most common hepatobiliary malignancy, ranks as the sixth most common type and the fourth most common primary cause of cancer-associated mortality worldwide (2). HCC is associated with strong propensity for invasion and metastasis, poor prognosis and a persistently high mortality due to early metastasis and late diagnosis (3). The most relevant risk factors of HCC include chronic infection with hepatitis B or hepatitis C virus, aflatoxin exposure, excessive alcohol intake, metabolic syndrome, obesity, smoking and type 2 diabetes (4).

The Barcelona Clinic Liver Cancer staging system, established in 1999, is the primary means of evaluating prognosis and selecting the appropriate treatment for HCC (5,6). The survival rate of patients with HCC is determined by the stage of diagnosis and treatment. In general, surgical resection, local ablation and liver transplantation are the preferred methods for early-stage HCC, while patients with intermediate-stage HCC may benefit from chemoembolization. The prognosis of HCC has not considerably improved over the past few years, despite the advances of classical therapies. Patients with advanced HCC or extensive extrahepatic disease usually have a survival of <3 months and, thus, pain and symptom control should be the primary focus for improving their quality of life (7,8). Overall, the 5-year survival rate of patients with HCC remains relatively poor at 20% (9), while that of patients with advanced HCC is <10% (10). The high mortality rate is due to the lack of effective treatments and increased resistance to conventional chemotherapy and radiotherapy (11). Therefore, the development of novel, tolerable and effective therapeutic agents is a pressing issue. The emergence of targeted therapies and the approval of the kinase inhibitor sorafenib have offered some hope for the future. Based on their physiological and pathological significance, growth factors and their receptors have become targets for drug discovery and development. Selective inhibitors against ligand growth factors and their growth factor receptors are currently indispensable in the molecular-targeted therapy of malignant tumors. Even if the molecular mechanisms responsible for the onset and progression of HCC remain largely unknown, novel therapeutic targets have recently become the focus of attention. One of these targets is the hepatocyte growth factor (HGF)/c-mesenchymal-epithelial transition receptor (c-MET) signaling pathway, which is known to promote tumor growth and metastasis in a number of human organs.

HGF/c-MET signaling pathway

Structure and function of HGF

HGF is a large multidomain heterodimeric protein, which belongs to the peptidase S1 family of serine proteases. HGF was discovered in 1989 and was cloned and expressed shortly after (12). In 1991, the scatter factor and the tumor cytotoxic factor were identified as HGF (13,14). The HGF gene is located on chromosome 7q21 and contains 18 exons and 17 introns (15). HGF is secreted in the form of a single-chain precursor HGF and processed into a double-chain mature HGF by extracellular proteases. The active state of HGF consists of an α chain (69 kDa) and a β chain (34 kDa), which are connected by a disulfide bridge. The α chain contains an N-terminal hairpin domain (N) and four Kringle domains (K1-K4). The first two Kringle domains and hairpin domain are crucial for HGF to perform its biological function. The β chain contains a serine protease homology domain, which is the binding site of c-MET (16). Serine proteases, such as matriptase and HGF-activator, are involved in the processing of HGF (17).

HGF is a multifunctional cytokine and acts on a great variety of epithelial cells as a mitogen (stimulation of cell proliferation), a motogen (stimulation of cell motility) and a morphogen (induction of multicellular tissue-like structure) (18). Due to these functions, previous studies have demonstrated that HGF serves a crucial biological and pathophysiological role in tissue regeneration, tumorigenesis, tumor invasion and epithelial wound healing (19,20).

Structure and function of c-MET

c-MET was initially identified as the fusion gene (tpr-met), which is the receptor of HGF encoded by c-MET proto-oncogene, in a chemically transformed human osteosarcoma cell line (21). It was identified as the transmembrane receptor tyrosine kinase (RTK) in 1991 (22,23). The gene encoding c-MET is located on human chromosome 7q21-31 and contains 21 exons and 20 introns. The encoded protein is ~120 kDa in size (24). c-MET is generated as a single-chain precursor and converted by post-translational modification. Mature c-MET is a disulfide-linked heterodimer consisting of a 50-kDa highly glycosylated extracellular α-chain and a 140-kDa transmembrane-spanning β-chain (25). The transmembrane chain includes a Sema homology region (SEMA domain), a PSI domain (plexin-semaphorin-integrin; PSI), four immunoglobulin-like regions in plexins and transcription factors (IPT domains), a transmembrane domain, a juxtamembrane domain, a tyrosine kinase domain and a c-terminal docking site (carboxyl terminal; CT). Among them, SEMA is the site where HGF binds directly to c-MET, and PSI can stabilize this interaction (2629). c-MET activation is pleiotropic, since its cytoplasmic domain can interact with various proteins involved in multiple cellular signaling pathways. Upon binding to HGF, c-MET becomes active and drives a complex biological program, contributing to the promotion of several biological activities, including cell proliferation, cell invasion and protection from apoptosis. By controlling the epithelial-mesenchymal transition (EMT) of myogenic progenitor cells, c-MET promotes the development of placental tissue, liver and neuronal precursors, as well as the migration and development of muscle tissue (30,31). Inappropriate c-MET activation promotes the onset, proliferation, invasion and metastasis of multiple cancer types, including HCC (3234).

HGF/c-MET axis (Fig. 1)

The binding of HGF and c-MET triggers dimerization of two subunits, resulting in autophosphorylation on the tyrosine residues Y1234 and Y1235 at the tyrosine kinase domain. Subsequently, it activates autophosphorylation of Y1349 and Y1356 residues near the COOH terminus, which has been identified to be able to recruit intracellular adapters via Src and to activate downstream signaling events (35). Numerous adaptor proteins, such as Shc, Src, the adaptor protein growth factor receptor-bound protein 2 (Grb2) and the p85 regulatory subunit of the effector molecules, such as phosphoinositol 3-kinase (PI3K), bind directly or indirectly to c-MET (36,37). Most of them consist of a Src homologous 2 domain interacting with c-MET and a Src homologous 3 domain binding to downstream signaling molecules (38). This recruitment triggers activation of certain important intracellular signaling pathways, such as the Src, signal transducer and activator of transcription 3 (STAT3), Ras-MAPK, PI3K-AKT and phospholipase C (PLCγ) signaling pathways (39,40). Through these intermediary pathways, HGF-induced c-MET activation induces a variety of cellular responses, including proliferation, survival, motility, invasion, angiogenesis, EMT and branching morphogenesis (41,42).

However, inappropriate amplification of the c-MET gene and activation of the HGF/c-MET axis, as well as eventually elevated protein expression and constitutive kinase activation, may contribute to proliferation, survival, invasion, migration, drug resistance and angiogenesis in a variety of tumors, and are associated with poor clinical outcomes (43,44). It has been reported that HGF and c-MET expression can be upregulated by interleukin (IL)-1, IL-6, basic fibroblast growth factor (bFGF), oncostatin M (OSM), tumor necrosis factor (TNF)-α and several other cytokines (45). The HGF/c-MET axis triggers a series of signaling cascades and is associated with higher proliferation, cell dissociation, poor prognosis, prevention of apoptosis and loss of intercellular junctions. During the past three decades, its abnormal activity has been documented as a common characteristic in a wide range of human malignancies, including colorectal cancer (46), breast cancer (47), bladder cancer (48), gastric cancer (49) and cervical cancer (50), as well as HCC.

Role of HGF/c-MET in HCC

Over the past few years, the availability of data regarding signaling pathways regulating the growth and progression of HCC has improved, and several studies have proposed that HGF/c-MET signaling is one of the most promising molecular signaling pathways with which to guide the treatment of HCC (51,52). Under physiological conditions, the HGF/c-MET signaling pathway mediates multiple cellular responses, such as tissue regeneration and cell migration. HGF-induced c-MET activation is strictly regulated; however, aberrant activation of the signaling pathway leads to increased cell motility and cell cycle progression, as well as numerous other protumorigenic signaling events, which has been demonstrated to be involved in multiple malignant neoplasms, including HCC (35).

HGF/c-MET expression in HCC

c-MET amplification, c-MET point mutations, exon 14 skipping mutations and excessive autocrine/paracrine HGF secretion may be the molecular mechanisms that cause aberration in the HGF/c-MET signaling pathway. Upregulation of c-MET expression at the transcriptional level can be caused by activation of hypoxia-inducible factor (HIF) and transcription factors, and downregulation of inhibitor microRNAs. Due to the mutation of deoxyadenosine tract element in the HGF gene promoter, HGF expression can be upregulated by transcriptional upregulation (53).

c-MET amplification is almost undetectable in patients with HCC and may lead to upregulation of protein expression and structural activation of c-MET receptor kinase (5456). The incidence of upregulation of c-MET expression has been observed in multiple studies. In one study, high expression levels of c-MET were observed in 25.4% of 59 patients with HCC (54). Another study revealed upregulation of c-MET expression in 80.6% of 93 patients with HCC (57). Furthermore, 282 c-MET+ patients (54.2%) were identified among a total of 520 patients with HCC, and upregulation of c-MET was associated with a shorter 7-year overall survival (OS) time (58). Compared with those in adjacent normal hepatic tissues, c-MET transcription levels in HCC tissues are increased by 30–100%, and protein levels are increased by 25–100% (59). Ang et al (60) evaluated the expression levels of c-MET and HGF in 49 patients with early HCC, and analyzed the results in terms of their association with the characteristics of disease and survival, revealing that the expression levels of c-MET and HGF are upregulated in well-characterized early disease; however, these are not associated with survival.

Association of HGF/c-MET with invasion and metastasis of HCC

Modern medical studies have demonstrated that the tumor microenvironment (TME) provides conditions for tumor metastasis, and EMT induced by different components in the TME is a key factor for distant metastasis of HCC (61). HGF can cause metastatic changes, including EMT, enhancement of motility and invasiveness of various tumor cells, and thus can be referred to as a ‘metastatic growth factor’. Among the metastatic factors, HGF serves a significant role in HCC progression (62). A number of studies have reported that the high expression levels of c-MET in HCC can improve the motility of cells, and generate a variety of signaling molecules via downstream signaling pathways, such as vascular epidermal growth factor (VEGF) and Rac signaling molecules, thereby promoting tumor neovascularization and tumor cell metastasis (63).

One study has demonstrated that HGF-induced c-MET activation can induce the invasion and migration of HCC cells (64). RNA interference (small interfering RNA) silencing of c-MET expression in the MHCC97-H HCC cell line decreased the proliferation, motility and invasion of MHCC97-H cells, indicating that the upregulation of c-MET protein expression serves a crucial role in the invasion of HCC cells (65). Zhang et al (66) aimed to explore how HGF/c-MET regulates EMT and the metastasis of HCC cells at the epigenetic level, and revealed that HGF/c-MET signaling can induce HCC cells to express high levels of sumo-specific protease1 (Senp1). Senp1 silencing can reduce HGF-mediated proliferation and migration of HCC cells, and Senp1 inhibition can also induce apoptosis and growth arrest. Liu et al (51) used HGF to treat Hep3B HCC cells with p53 deficiency, and revealed that it could promote the invasion and metastasis of Hep3B cells, as well as the occurrence of EMT. Additionally, they revealed that p53 defects could enhance the HGF/c-MET signaling pathway, thus upregulating the transcription factor Snail to promote the invasion of HCC.

HGF/c-MET levels as a diagnostic, prognostic and predictive biomarker

The poor survival rate and the increasing incidence of HCC emphasize the importance of detecting novel screening biomarkers in the early treatable stage to move towards personalized medicine (67). Different tissue-based and circulation-based aberrations of HGF/c-MET may be considered as tumor diagnostic, prognostic and predictive biomarkers in HCC.

Circulating HGF level as a diagnostic biomarker

HCC develops along different clinical backgrounds, including alcoholism, cirrhosis and chronic hepatitis. All these factors result in continuous inflammation and regeneration of hepatocytes, which is challenging for the early diagnosis of HCC. The diagnostic value of serum HGF in HCC has been demonstrated (68). A study revealed that the serum HGF levels of patients with HCC were higher than those of patients without HCC, suggesting that the evaluation of HGF levels is of clinical significance in the diagnosis of HCC (69). Karabulut et al (70) investigated the relationship between serum HGF levels and tumor progression in patients with HCC and revealed that the baseline levels of serum HGF were higher in patients with HCC than in controls, demonstrating the diagnostic value of serum HGF. However, Unic et al (71) demonstrated that the evidence for the diagnostic value of HGF was inadequate. It is clear that these issues must be addressed in larger randomized clinical trials.

HGF or c-MET aberrant activation as a prognostic biomarker

Several studies have demonstrated the prognostic value of circulating HGF levels, and most studies revealed a negative association between HGF levels and survival in patients with different types of cancer (72). Pretreatment plasma HGF has been reported to be a useful biomarker for predicting the susceptibility to radiation-induced liver dysfunction and survival after radiotherapy and liver transplantation (73). Rimassa et al (74) reported that patients with higher baseline HGF had a distinctly shorter survival, indicating that the circulating HGF level may be a prognostic biomarker in secondary HCC. However, no prognostic value was identified in another study on patients with HCC (70).

Multiple studies have demonstrated the prognostic value of c-MET levels on tumor grade, tumor recurrence, intrahepatic metastases and overall survival, as well as portal vein invasion or thrombosis (7578). It has been demonstrated that upregulation of c-MET expression is an adverse prognostic marker for recurrence and survival in HCC. In a meta-analysis, patients with HCC with high c-MET expression had worse relapse-free survival and OS compared with patients with HCC with low c-MET expression (79). Zhuang et al (80) revealed that the expression levels of c-MET protein in HCC tissues are higher than those in adjacent normal tissues, further supporting the prognostic value of c-MET protein expression in patients with HCC. In a retrospective study of 194 patients with HCC treated by hepatic resection or microwave ablation, upregulation of c-MET expression was revealed to be associated with poor survival outcomes (81).

c-MET aberrant activation as a predictive biomarker

The c-MET status of patients may serve as a potential biomarker for predicting the response to HGF/c-MET-targeted inhibitors. Several clinical studies have addressed the predictive role of c-MET dysregulation as an actionable target in HCC. A review discussed the importance of accurate HGF/c-MET signaling detection as a predictive biomarker for the selection of c-MET-targeted therapies and considered the c-MET status of patients as a predictor of the clinical response to HGF/c-MET inhibitors (82). Xiang et al (83) proposed that the activation of c-MET in HCC might predict the efficacy of cabozantinib therapy. Chu et al (57) explored the clinical and prognostic significance of c-MET expression in patients with HCC, suggesting that high c-MET expression may be a promising efficacy-predicting biomarker for the prediction of the response to sorafenib treatment, thus achieving individualized therapy for patients with HCC.

HCC therapy via targeting of the HGF/c-MET signaling pathway

Despite extensive efforts and recent advances made in the diagnosis and treatment of HCC, the prognosis of patients with HCC is still relatively poor, and definitive treatments remain to be improved, particularly for patients with advanced HCC (84). RTK c-MET expression is frequently upregulated in tumor tissues, and the activation of c-MET signaling is associated with drug resistance and the processes of carcinogenesis, invasion and metastasis. Furthermore, inhibiting HGF/c-MET signaling appears to be safe, considering that neutralization of HGF or c-MET has no noticeable adverse effects (25). Therefore, c-MET and HGF are considered to be primary targets for the development of anticancer drugs. Therapeutic strategies for the prevention of HCC tumor progression via targeting of the HGF/c-MET signaling pathway have been intensively investigated. Due to its indisputable role in HCC, the HGF/c-MET axis has been considered as a promising therapeutic target for the development of novel anticancer treatments (85). The binding of HGF to the RTK c-MET is involved in the malignant process of HCC and disrupting this interaction may represent a promising therapeutic strategy. On the premise of understanding the HGF/c-MET signaling pathway, several strategies have been designed to interfere with this signaling pathway: i) c-MET kinase inhibitors (inhibiting c-MET kinase activity); ii) c-MET adaptor inhibitors (inhibiting c-MET and adaptor interaction); and iii) antibody-based inhibitors (blocking ligand and receptor interaction) (86). Numerous preclinical and clinical studies have been devoted to developing anticancer drugs targeting the HGF/c-MET signaling pathway (87,88).

c-MET kinase inhibitors

RTKs are key regulators in normal growth, development and regeneration of mammals. Dysregulation of RTKs is attributed to gene mutation, gene amplification, chromosome rearrangement, transcriptional upregulation, etc., which have been identified as causative factors in the development of human malignancies (89). Most small molecule-kinase inhibitors can block the phosphorylation of the catalytic domain within the receptor by competitively antagonizing the occupation of the intracellular ATP binding sites, thus impeding the recruitment of signal transducers and mediators, and consequently preventing the transmission of downstream signals. To date, numerous small-molecule kinase inhibitors targeting c-MET have entered clinical trials, with hundreds of trials completed or ongoing, either as a single drug or in conjunction with other cancer drugs (90), which represents a potential promising target for the treatment of patients with HCC with an active HGF/c-MET signaling pathway (91).

Sorafenib is a multi-kinase inhibitor of VEGF, and is the first approved systemic therapy for unresectable and advanced HCC and the first-line clinical treatment choice (92,93). However, the survival benefits of sorafenib are limited and development of novel effective therapies is urgently required (10). The overall results are far from satisfactory, with the overall survival improved by <1 year (94). Furthermore, acquired drug resistance and side effects of sorafenib have also raised concerns (95). Targeted molecular therapies other than sorafenib are still being pursued and may be the second-line drugs for patients who fail or cannot tolerate sorafenib. The candidates are listed in Table I. Foretinib, a multikinase inhibitor with potent activity against c-MET, recepteur dorigine nantais (RON), AXL receptor tyrosine kinase (AXL), tyrosine-protein kinase receptor Tie-2 and vascular endothelial growth factor receptor 2 (VEGFR2), is currently in a phase I/II trial to estimate its safety and tolerability in patients with advanced HCC (96). Cabozantinib is an oral inhibitor of c-MET, VEGFR and ret proto-oncogene, and a phase II randomized discontinuation trial has been performed on 41 patients with advanced HCC and Child-Pugh cirrhosis (97). Furthermore, a phase III trial is ongoing (98). Tivantinib, a staurosporine derivative, has been reported to exhibit promising activity in a variety of phase I/II clinical trials, particularly in highly selective cancers targeting c-MET-driven signaling (99). Unfortunately, it failed to reach the primary endpoint in a phase III clinical trial (85).

Table I.

c-MET kinase inhibitors in hepatocellular carcinoma.

Table I.

c-MET kinase inhibitors in hepatocellular carcinoma.

AgentTargetsPhaseMechanism(Refs.)
Foretinibc-MET, AXL, RON, VEGFR-2, TIE-2I/IIATP competitive   (96)
Gefitinibc-MET, EGFR, HGFIIATP competitive(104)
Cabozantinibc-MET,VEGFR2, RET,IIIATP competitive(105)
BMS-777607c-MET, RONI/IIATP competitive(106)
Golvantinibc-MET,VEGFR-2I/IIATP competitive(107)
Crizotinibc-MET, ALK, ROS1IIATP competitive(108)
MK2461c-MET, RON, TIE-2,I/IIATP competitive(109)
AMG337c-METIATP competitive(110)
Tepotinibc-METIb/IIATP competitive(111)
Capmatinibc-METIIATP competitive(112)
Tivantinibc-METIIINon-ATP competitive(113)

[i] c-MET, c-mesenchymal-epithelial transition receptor; HGF, hepatocyte growth factor; RON, recepteur d'origine nantais; VEGFR2, vascular endothelial growth factor receptor 2; AXL, anexelekto; EGFR, epidermal growth factor receptor; TIE-2, tyrosine kinase receptor 2; RET, rearranged during transfection; ALK, anaplastic lymphoma kinase; ROS1, c-ros oncogene 1.

All these small-molecule c-MET kinase inhibitors have exhibited antitumor activity in nonclinical studies of HCC; however, most of them are still in the clinical trial phase due to other efficacy, tolerance and safety issues (100102). Firstly, it has been demonstrated that c-MET inhibitors have numerous side effects, such as anemia, liver and bone marrow toxicity, and neutropenia. Furthermore, the efficacy of individual c-MET-based target therapy is limited for patients with HCC, particularly for patients with c-MET-negative HCC. Finally, advanced HCC is often accompanied by fibrosis and cirrhosis, resulting in the reduction of hepatic drug clearance and metabolic enzyme activity (103).

c-MET adaptor inhibitors

Adaptor proteins, such as Grb1, Grb2 and PI3K, are important components of the HGF/c-MET signaling pathway. Multiple tumor effects induced by HGF/c-MET signaling can be inhibited by blocking the binding of one or more adaptor proteins to c-MET or by inhibiting the activation of these adaptor proteins. Grb2 is regarded as a key protein in the HGF/c-MET axis, since it interacts with Y1356 of c-MET to transduce HGF signaling to the cytoplasm and connects to several signaling transducers, such as Ras, Son of Sevenless (SOS) and GRB2 associated binding protein 1 (GAB1) (85). As a small selective antagonist of Grb2, C90 can effectively block the cell motility and matrix invasion of gastric cancer cells in vitro and reduce the metastasis of major solid tumors produced by human prostate adenocarcinoma cells in vivo (114,115). The efficacy of c-MET adaptor inhibitors in the treatment of HCC still requires further study.

Antibody-based inhibitors

In terms of the progress of humanized antibody drugs, anti-HGF and anti-MET antibodies have entered the clinical trial stage. At least eight anti-MET [onartuzumab (116), emibetuzumab (117), LY3164530, JNJ-61186372, SAIT301, ABT-700, ARGX-111 and DN30] and four anti-HGF [rilotumumab (AMG102), ficlatuzumab (AV-299), HuL2G7 (TAK-701) and YYB-101] antibodies have been examined or are being examined in clinical trials (Table II). The mechanism of these antibodies is blocking the binding of ligand HGF and receptor c-MET to achieve an antitumor therapeutic effect. Compared with small-molecule chemical drugs, antibody drugs have the characteristics of strong targeting specificity, high activity, long half-life and low toxicity. Furthermore, antibody drugs have excellent predictability in pharmacology, thus their safety is better.

Table II.

Antibodies targeting the HGF/c-MET axis.

Table II.

Antibodies targeting the HGF/c-MET axis.

Anti-MET antibodyPhaseFormat(Refs.)
OnartuzumabII/IIImAb (IgG1)(118)
EmibetuzumabIImAb (IgG1)(119)
AmivantamabIbsAb (DuoBody)(120)
LY3164530IbsAb(121)
SAIT301ImAb(122)
TelisotuzumabImAb(123)
ARGX-111ImAb(124)
DN30 InvestigationalFab(125)

Anti-HGF antibody

RilotumumabII/IIImAb (IgG2)(126)
FiclatuzumabIImAb (IgG1)(127)
HuL2G7ImAb (IgG1)(128)
YYB-101ImAb(129)

[i] HGF, hepatocyte growth factor; c-MET, c-mesenchymal-epithelial transition receptor; mAb, monoclonal antibody; bsAb, bi-specific antibody; Fab, antigen-binding fragment.

Using herbal medicinal antagonists to achieve more effective and safer HGF/c-MET targeting

Using natural herbs instead of synthetic chemicals is another strategy to avoid the issues of HGF/c-MET-targeted therapy. For example, the medicinal peptide LZ8 (also known as Ling-Zhi or Reishi), a well-known traditional Chinese medicine extracted from Ganoderma lucidum, has antitumor activity against cervical cancer, breast cancer, lung cancer and HCC (130132). LZ8 can inhibit the progression of HCC by blocking both the c-MET-dependent and -independent MAPK signaling pathway (133). Resveratrol, one of the major polyphenols found in red wine, inhibits the progression of HCC via downregulation of HGF/c-MET signaling (134). Furthermore, our previous study revealed that the traditional Chinese medicine compound QHF has an anti-angiogenic effect on H22 cells, and it can suppress tumor growth by prohibiting the HGF/c-MET signaling pathway (135). Zhang et al (136) reported the synergistic effect of astragaloside IV and curcumin on the inhibition of tumor growth and angiogenesis in HCC mouse models, and revealed that this synergistic effect is associated with HGF.

Future directions and conclusions

HCC is a complicated pathology with interconnected regulatory networks. Due to the limited clinical efficacy of current therapeutic drugs and the lack of biomarkers for early diagnosis, HCC is among the cancers with the worst prognosis. In recent years, great achievements have been made in understanding the role of HGF/c-MET signaling in overexpression, invasion, migration and metastasis of HCC. Additionally, the discovery of novel drugs related to the HGF/c-MET signaling pathway provides a promising clinical approach for designing improved specific individualized therapy. Accordingly, targeting the HGF/c-MET signaling pathway has become a research ‘hotspot’ for efficient targeted therapy and prognosis of patients with HCC. In terms of biomarker development and drug combination, more efforts are required for novel strategies to ensure optimal use of the drugs in the treatment of HCC. Therefore, it is worthwhile to explore the detailed mechanisms of HGF/c-MET signaling and the complex multistep process of HCC, which will help to promote the development of preventive measures, early diagnostic methods and more appropriate targets, in order to develop more effective and safer therapeutic strategies.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The information provided in this review is documented by relevant references.

Authors contributions

WM designed and wrote this review. TC reviewed the literature data and edited the manuscript. Both authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Waller LP, Deshpande V and Pyrsopoulos N: Hepatocellular carcinoma: A comprehensive review. World J Hepatol. 7:2648–2663. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Forner A, Reig M and Bruix J: Hepatocellular carcinoma. Lancet. 391:1301–1314. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Li L and Wang H: Heterogeneity of liver cancer and personalized therapy. Cancer Lett. 379:191–197. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Llovet JM, Bru C and Bruix J: Prognosis of hepatocellular carcinoma: The BCLC staging classification. Semin Liver Dis. 19:329–338. 1999. View Article : Google Scholar : PubMed/NCBI

6 

de Lope CR, Tremosini S, Forner A, Reig M and Bruix J: Management of HCC. J Hepatol. 56 (Suppl 1):S75–S87. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Bruix J and Sherman M; American Association for the Study of Liver Diseases, : Management of hepatocellular carcinoma: An update. Hepatology. 53:1020–1022. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M and Gores G: Hepatocellular carcinoma. Nat Rev Dis Primers. 2:160182016. View Article : Google Scholar : PubMed/NCBI

9 

Grandhi MS, Kim AK, Ronnekleiv-Kelly SM, Kamel IR, Ghasebeh MA and Pawlik TM: Hepatocellular carcinoma: From diagnosis to treatment. Surg Oncol. 25:74–85. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Goyal L, Muzumdar MD and Zhu AX: Targeting the HGF/c-MET pathway in hepatocellular carcinoma. Clin Cancer Res. 19:2310–2318. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Whittaker S, Marais R and Zhu AX: The role of signaling pathways in the development and treatment of hepatocellular carcinoma. Oncogene. 29:4989–5005. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Nakamura T, Nishizawa T, Hagiya M, Seki T, Shimonishi M, Sugimura A, Tashiro K and Shimizu S: Molecular cloning and expression of human hepatocyte growth factor. Nature. 342:440–443. 1989. View Article : Google Scholar : PubMed/NCBI

13 

Furlong RA, Takehara T, Taylor WG, Nakamura T and Rubin JS: Comparison of biological and immunochemical properties indicates that scatter factor and hepatocyte growth factor are indistinguishable. J Cell Sci. 100:173–177. 1991. View Article : Google Scholar : PubMed/NCBI

14 

Shima N, Nagao M, Ogaki F, Tsuda E, Murakami A and Higashio K: Tumor cytotoxic factor/hepatocyte growth factor from human fibroblasts: Cloning of its cDNA, purification and characterization of recombinant protein. Biochem Biophys Res Commun. 180:1151–1158. 1991. View Article : Google Scholar : PubMed/NCBI

15 

Seki T, Hagiya M, Shimonishi M, Nakamura T and Shimizu S: Organization of the human hepatocyte growth factor-encoding gene. Gene. 102:213–219. 1991. View Article : Google Scholar : PubMed/NCBI

16 

Zhang Y, Xia M, Jin K, Wang S, Wei H, Fan C, Wu Y, Li X, Li X, Li G, et al: Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer. 17:452018. View Article : Google Scholar : PubMed/NCBI

17 

Sakai K, Aoki S and Matsumoto K: Hepatocyte growth factor and Met in drug discovery. J Biochem. 157:271–284. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Nakamura T, Sakai K, Nakamura T and Matsumoto K: Hepatocyte growth factor twenty years on: Much more than a growth factor. J Gastroenterol Hepatol. 26 (Suppl 1):S188–S202. 2011. View Article : Google Scholar

19 

Barrow-McGee R and Kermorgant S: Met endosomal signalling: In the right place, at the right time. Int J Biochem Cell Biol. 49:69–74. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Imamura R and Matsumoto K: Hepatocyte growth factor in physiology and infectious diseases. Cytokine. 98:97–106. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Cooper CS, Park M, Blair DG, Tainsky MA, Huebner K, Croce CM and Vande Woude GF: Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature. 311:29–33. 1984. View Article : Google Scholar : PubMed/NCBI

22 

Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF and Aaronson SA: Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science. 251:802–804. 1991. View Article : Google Scholar : PubMed/NCBI

23 

Naldini L, Vigna E, Narsimhan RP, Gaudino G, Zarnegar R, Michalopoulos GK and Comoglio PM: Hepatocyte growth factor (HGF) stimulates the tyrosine kinase activity of the receptor encoded by the proto-oncogene c-MET. Oncogene. 6:501–504. 1991.PubMed/NCBI

24 

Li Y, Liu H and Chen J: Dysregulation of HGF/c-Met signal pathway and their targeting drugs in lung cancer. Zhongguo Fei Ai Za Zhi. 17:625–634. 2014.(In Chinese). PubMed/NCBI

25 

Kim KH and Kim H: Progress of antibody-based inhibitors of the HGF-cMET axis in cancer therapy. Exp Mol Med. 49:e3072017. View Article : Google Scholar : PubMed/NCBI

26 

Safaie Qamsari E, Safaei Ghaderi S, Zarei B, Dorostkar R, Bagheri S, Jadidi-Niaragh F, Somi MH and Yousefi M: The c-Met receptor: Implication for targeted therapies in colorectal cancer. Tumour Biol. 39:10104283176991182017. View Article : Google Scholar : PubMed/NCBI

27 

Yin B, Liu Z, Wang Y, Wang X, Liu W, Yu P, Duan X, Liu C, Chen Y, Zhang Y, et al: RON and c-Met facilitate metastasis through the ERK signaling pathway in prostate cancer cells. Oncol Rep. 37:3209–3218. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Hass R, Jennek S, Yang Y and Friedrich K: c-Met expression and activity in urogenital cancers-novel aspects of signal transduction and medical implications. Cell Commun Signal. 15:102017. View Article : Google Scholar : PubMed/NCBI

29 

Hu CT, Wu JR, Cheng CC and Wu WS: The therapeutic targeting of HGF/c-Met signaling in hepatocellular carcinoma: Alternative approaches. Cancers. 9:582017. View Article : Google Scholar : PubMed/NCBI

30 

Giordano S and Columbano A: Met as a therapeutic target in HCC: Facts and hopes. J Hepatol. 60:442–452. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Birchmeier C, Birchmeier W, Gherardi E and Vande Woude GF: Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 4:915–925. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Szturz P, Raymond E, Abitbol C, Albert S, de Gramont A and Faivre S: Understanding c-MET signalling in squamous cell carcinoma of the head & neck. Crit Rev Oncol Hematol. 111:39–51. 2017. View Article : Google Scholar : PubMed/NCBI

33 

International Cancer Genome Consortium PedBrain Tumor Project, . Recurrent MET fusion genes represent a drug target in pediatric glioblastoma. Nat Med. 22:1314–1320. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Kim B, Jung N, Lee S, Sohng JK and Jung HJ: Apigenin inhibits cancer stem cell-like phenotypes in human glioblastoma cells via suppression of c-Met signaling. Phytother Res. 30:1833–1840. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Blumenschein GR Jr, Mills GB and Gonzalez-Angulo AM: Targeting the hepatocyte growth factor-cMET axis in cancer therapy. J Clin Oncol. 30:3287–3296. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Gherardi E, Birchmeier W, Birchmeier C and Vande Woude G: Targeting MET in cancer: Rationale and progress. Nat Rev Cancer. 12:89–103. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Spina A, De Pasquale V, Cerulo G, Cocchiaro P, Della Morte R, Avallone L and Pavone LM: HGF/c-MET axis in tumor microenvironment and metastasis formation. Biomedicines. 3:71–88. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Gui Y, Yeganeh M, Donates YC, Tobelaim WS, Chababi W, Mayhue M, Yoshimura A, Ramanathan S, Saucier C and Ilangumaran S: Regulation of MET receptor tyrosine kinase signaling by suppressor of cytokine signaling 1 in hepatocellular carcinoma. Oncogene. 34:5718–5728. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Liu X, Newton RC and Scherle PA: Developing c-MET pathway inhibitors for cancer therapy: Progress and challenges. Trends Mol Med. 16:37–45. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Sierra JR and Tsao MS: c-MET as a potential therapeutic target and biomarker in cancer. Ther Adv Med Oncol. 3 (Suppl 1):S21–S35. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Jafarnejad M, Sove RJ, Danilova L, Mirando AC, Zhang Y, Yarchoan M, Tran PT, Pandey NB, Fertig EJ and Popel AS: Mechanistically detailed systems biology modeling of the HGF/Met pathway in hepatocellular carcinoma. NPJ Syst Biol Appl. 5:292019. View Article : Google Scholar : PubMed/NCBI

42 

Zhang YW: Promise and challenges on the horizon of MET-targeted cancer therapeutics. World J Biol Chem. 6:16–27. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Cao HH, Cheng CY, Su T, Fu XQ, Guo H, Li T, Tse AK, Kwan HY, Yu H and Yu ZL: Quercetin inhibits HGF/c-Met signaling and HGF-stimulated melanoma cell migration and invasion. Mol Cancer. 14:1032015. View Article : Google Scholar : PubMed/NCBI

44 

Matsumoto K, Umitsu M, De Silva DM, Roy A and Bottaro DP: Hepatocyte growth factor/MET in cancer progression and biomarker discovery. Cancer Sci. 108:296–307. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Czyz M: HGF/c-MET signaling in melanocytes and melanoma. Int J Mol Sci. 19:38442018. View Article : Google Scholar : PubMed/NCBI

46 

Parizadeh SM, Jafarzadeh-Esfehani R, Fazilat-Panah D, Hassanian SM, Shahidsales S, Khazaei M, Parizadeh SMR, Ghayour-Mobarhan M, Ferns GA and Avan A: The potential therapeutic and prognostic impacts of the c-MET/HGF signaling pathway in colorectal cancer. IUBMB Life. 71:802–811. 2019.PubMed/NCBI

47 

Liu S: HGF-MET as a breast cancer biomarker. Aging. 7:150–151. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Matsumoto R, Tsuda M, Wang L, Maishi N, Abe T, Kimura T, Tanino M, Nishihara H, Hida K, Ohba Y, et al: Adaptor protein CRK induces epithelial-mesenchymal transition and metastasis of bladder cancer cells through HGF/c-Met feedback loop. Cancer Sci. 106:709–717. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Huang KH, Sung IC, Fang WL, Chi CW, Yeh TS, Lee HC, Yin PH, Li AF, Wu CW, Shyr YM and Yang MH: Correlation between HGF/c-Met and Notch1 signaling pathways in human gastric cancer cells. Oncol Rep. 40:294–302. 2018.PubMed/NCBI

50 

Boromand N, Hasanzadeh M, ShahidSales S, Farazestanian M, Gharib M, Fiuji H, Behboodi N, Ghobadi N, Hassanian SM, Ferns GA and Avan A: Clinical and prognostic value of the C-Met/HGF signaling pathway in cervical cancer. J Cell Physiol. 233:4490–4496. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Liu WT, Jing YY, Yu GF, Chen H, Han ZP, Yu DD, Fan QM, Ye F, Li R, Gao L, et al: Hepatic stellate cell promoted hepatoma cell invasion via the HGF/c-Met signaling pathway regulated by p53. Cell Cycle. 15:886–894. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Tan S, Li R, Ding K, Lobie PE and Zhu T: miR-198 inhibits migration and invasion of hepatocellular carcinoma cells by targeting the HGF/c-MET pathway. FEBS Lett. 585:2229–2234. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Moosavi F, Giovannetti E, Saso L and Firuzi O: HGF/MET pathway aberrations as diagnostic, prognostic, and predictive biomarkers in human cancers. Crit Rev Clin Lab Sci. 56:533–566. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Kondo S, Ojima H, Tsuda H, Hashimoto J, Morizane C, Ikeda M, Ueno H, Tamura K, Shimada K, Kanai Y and Okusaka T: Clinical impact of c-Met expression and its gene amplification in hepatocellular carcinoma. Int J Clin Oncol. 18:207–213. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Wang K, Lim HY, Shi S, Lee J, Deng S, Xie T, Zhu Z, Wang Y, Pocalyko D, Yang WJ, et al: Genomic landscape of copy number aberrations enables the identification of oncogenic drivers in hepatocellular carcinoma. Hepatology. 58:706–717. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, Calderaro J, Bioulac-Sage P, Letexier M, Degos F, et al: Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 44:694–698. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Chu JS, Ge FJ, Zhang B, Wang Y, Silvestris N, Liu LJ, Zhao CH, Lin L, Brunetti AE, Fu YL, et al: Expression and prognostic value of VEGFR-2, PDGFR-β, and c-Met in advanced hepatocellular carcinoma. J Exp Clin Cancer Res. 32:162013. View Article : Google Scholar : PubMed/NCBI

58 

Ke AW, Shi GM, Zhou J, Wu FZ, Ding ZB, Hu MY, Xu Y, Song ZJ, Wang ZJ, Wu JC, et al: Role of overexpression of CD151 and/or c-Met in predicting prognosis of hepatocellular carcinoma. Hepatology. 49:491–503. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Lee SJ, Lee J, Sohn I, Mao M, Kai W, Park CK and Lim HY: A survey of c-MET expression and amplification in 287 patients with hepatocellular carcinoma. Anticancer Res. 33:5179–5186. 2013.PubMed/NCBI

60 

Ang CS, Sun MY, Huitzil-Melendez DF, Chou JF, Capanu M, Jarnagin W, Fong Y, Dematteo RP, DAngelica M, Allen P, et al: c-MET and HGF mRNA expression in hepatocellular carcinoma: Correlation with clinicopathological features and survival. Anticancer Res. 33:3241–3245. 2013.PubMed/NCBI

61 

He C, Zhou Z, Jiang H, Yin Z, Meng S, Zhang J, Huang P, Xu K, Bian L, Xiao Z and Wang J: Epithelial-mesenchymal transition is superior to vessels-encapsulate tumor cluster in promoting metastasis of hepatocellular carcinoma: A morphological evidence. J Cancer. 8:39–47. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Trusolino L, Bertotti A and Comoglio PM: MET signalling: Principles and functions in development, organ regeneration and cancer. Nat Rev Mol Cell Biol. 11:834–848. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Menard L, Parker PJ and Kermorgant S: Receptor tyrosine kinase c-Met controls the cytoskeleton from different endosomes via different pathways. Nat Commun. 5:39072014. View Article : Google Scholar : PubMed/NCBI

64 

Bozkaya G, Korhan P, Cokakli M, Erdal E, Sağol O, Karademir S, Korch C and Atabey N: Cooperative interaction of MUC1 with the HGF/c-Met pathway during hepatocarcinogenesis. Mol Cancer. 11:642012. View Article : Google Scholar : PubMed/NCBI

65 

Xie B, Xing R, Chen P, Gou Y, Li S, Xiao J and Dong J: Down-regulation of c-Met expression inhibits human HCC cells growth and invasion by RNA interference. J Surg Res. 162:231–238. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Zhang W, Sun H, Shi X, Wang H, Cui C, Xiao F, Wu C, Guo X and Wang L: SENP1 regulates hepatocyte growth factor-induced migration and epithelial-mesenchymal transition of hepatocellular carcinoma. Tumour Biol. 37:7741–7748. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Hass HG, Jobst J, Scheurlen M, Vogel U and Nehls O: Gene expression analysis for evaluation of potential biomarkers in hepatocellular carcinoma. Anticancer Res. 35:2021–2028. 2015.PubMed/NCBI

68 

Alkozai EM, Porte RJ, Adelmeijer J, Zanetto A, Simioni P, Senzolo M and Lisman T: Levels of angiogenic proteins in plasma and platelets are not different between patients with hepatitis B/C-related cirrhosis and patients with cirrhosis and hepatocellular carcinoma. Platelets. 26:577–582. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Yamagamim H, Moriyama M, Matsumura H, Aoki H, Shimizu T, Saito T, Kaneko M, Shioda A, Tanaka N and Arakawa Y: Serum concentrations of human hepatocyte growth factor is a useful indicator for predicting the occurrence of hepatocellular carcinomas in C-viral chronic liver diseases. Cancer. 95:824–834. 2002. View Article : Google Scholar : PubMed/NCBI

70 

Karabulut S, Tas F, Akyuz F, Ormeci AC, Serilmez M, Soydinç HO, Vatansever S and Yasasever V: Clinical significance of serum hepatocyte growth factor (HGF) levels in hepatocellular carcinoma. Tumour Biol. 35:2327–2333. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Unic A, Derek L, Duvnjak M, Patrlj L, Rakić M, Kujundžić M, Renjić V, Štoković N, Dinjar P, Jukic A and Grgurević I: Diagnostic specificity and sensitivity of PIVKAII, GP3, CSTB, SCCA1 and HGF for the diagnosis of hepatocellular carcinoma in patients with alcoholic liver cirrhosis. Ann Clin Biochem. 55:355–362. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Dreikhausen L, Blank S, Sisic L, Heger U, Weichert W, Jäger D, Bruckner T, Giese N, Grenacher L, Falk C, et al: Association of angiogenic factors with prognosis in esophageal cancer. BMC cancer. 15:1212015. View Article : Google Scholar : PubMed/NCBI

73 

Hong TS, Grassberger C, Yeap BY, Jiang W, Wo JY, Goyal L, Clark JW, Crane CH, Koay EJ, Dima S, et al: Pretreatment plasma HGF as potential biomarker for susceptibility to radiation-induced liver dysfunction after radiotherapy. NPJ Precis Oncol. 2:222018. View Article : Google Scholar : PubMed/NCBI

74 

Rimassa L, Abbadessa G, Personeni N, Porta C, Borbath I, Daniele B, Salvagni S, Van Laethem JL, Van Vlierberghe H, Trojan J, et al: Tumor and circulating biomarkers in patients with second-line hepatocellular carcinoma from the randomized phase II study with tivantinib. Oncotarget. 7:72622–72633. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Yan S, Jiao X, Zou H and Li K: Prognostic significance of c-Met in breast cancer: A meta-analysis of 6010 cases. Diagn Pathol. 10:622015. View Article : Google Scholar : PubMed/NCBI

76 

Pyo JS, Kang G, Cho WJ and Choi SB: Clinicopathological significance and concordance analysis of c-MET immunohistochemistry in non-small cell lung cancers: A meta-analysis. Pathol Res Pract. 212:710–716. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Pyo JS, Kang G and Cho H: Clinicopathological significance and diagnostic accuracy of c-MET expression by immunohistochemistry in gastric cancer: A meta-analysis. J Gastric Cancer. 16:141–151. 2016. View Article : Google Scholar : PubMed/NCBI

78 

Liu Y, Yu XF, Zou J and Luo ZH: Prognostic value of c-Met in colorectal cancer: A meta-analysis. World J Gastroenterol. 21:3706–3710. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Kim JH, Kim HS, Kim BJ, Jang HJ and Lee J: Prognostic value of c-Met overexpression in hepatocellular carcinoma: A meta-analysis and review. Oncotarget. 8:90351–90357. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Zhuang PH, Xu L, Gao L, Lu W, Ruan LT and Yang J: Correlations of microvascular blood flow of contrast-enhanced ultrasound and HGF/c-Met signaling pathway with clinicopathological features and prognosis of patients with hepatocellular carcinoma. Onco Targets Ther. 10:847–857. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Wang ZL, Liang P, Dong BW, Yu XL and Yu DJ: Prognostic factors and recurrence of small hepatocellular carcinoma after hepatic resection or microwave ablation: A retrospective study. J Gastrointest Surg. 12:327–337. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Zhang Y, Du Z and Zhang M: Biomarker development in MET-targeted therapy. Oncotarget. 7:37370–37389. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Xiang Q, Chen W, Ren M, Wang J, Zhang H, Deng DY, Zhang L, Shang C and Chen Y: Cabozantinib suppresses tumor growth and metastasis in hepatocellular carcinoma by a dual blockade of VEGFR2 and MET. Clin Cancer Res. 20:2959–2970. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Forner A, Reig ME, de Lope CR and Bruix J: Current strategy for staging and treatment: The BCLC update and future prospects. Semin Liver Dis. 30:61–74. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Garcia-Vilas JA and Medina MA: Updates on the hepatocyte growth factor/c-Met axis in hepatocellular carcinoma and its therapeutic implications. World J Gastroenterol. 24:3695–3708. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Gao J, Inagaki Y, Song P, Qu X, Kokudo N and Tang W: Targeting c-Met as a promising strategy for the treatment of hepatocellular carcinoma. Pharmacol Res. 65:23–30. 2012. View Article : Google Scholar : PubMed/NCBI

87 

Gao JJ, Inagaki Y, Xue X, Qu XJ and Tang W: c-Met: A potential therapeutic target for hepatocellular carcinoma. Drug Discov Ther. 5:2–11. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Garber K: MET inhibitors start on road to recovery. Nat Rev Drug Discov. 13:563–565. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Lemmon MA and Schlessinger J: Cell signaling by receptor tyrosine kinases. Cell. 141:1117–1134. 2010. View Article : Google Scholar : PubMed/NCBI

90 

Munshi N, Jeay S, Li Y, Chen CR, France DS, Ashwell MA, Hill J, Moussa MM, Leggett DS and Li CJ: ARQ 197, a novel and selective inhibitor of the human c-Met receptor tyrosine kinase with antitumor activity. Mol Cancer Ther. 9:1544–1553. 2010. View Article : Google Scholar : PubMed/NCBI

91 

You H, Ding W, Dang H, Jiang Y and Rountree CB: c-Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinoma. Hepatology. 54:879–889. 2011. View Article : Google Scholar : PubMed/NCBI

92 

Brunetti O, Gnoni A, Licchetta A, Longo V, Calabrese A, Argentiero A, Delcuratolo S, Solimando AG, Casadei-Gardini A and Silvestris N: Predictive and prognostic factors in HCC patients treated with Sorafenib. Medicina (Kaunas). 55:7072019. View Article : Google Scholar : PubMed/NCBI

93 

Kuczynski EA, Lee CR, Man S, Chen E and Kerbel RS: Effects of sorafenib dose on acquired reversible resistance and toxicity in hepatocellular carcinoma. Cancer Res. 75:2510–2519. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Zhu AX: Molecularly targeted therapy for advanced hepatocellular carcinoma in 2012: Current status and future perspectives. Semin Oncol. 39:493–502. 2012. View Article : Google Scholar : PubMed/NCBI

95 

Zhai B and Sun XY: Mechanisms of resistance to sorafenib and the corresponding strategies in hepatocellular carcinoma. World J Hepatol. 5:345–352. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Yau TCC, Lencioni R, Sukeepaisarnjaroen W, Chao Y, Yen CJ, Lausoontornsiri W, Chen PJ, Sanpajit T, Camp A, Cox DS, et al: A phase I/II multicenter study of single-agent foretinib as first-line therapy in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 23:2405–2413. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Kelley RK, Verslype C, Cohn AL, Yang TS, Su WC, Burris H, Braiteh F, Vogelzang N, Spira A, Foster P, et al: Cabozantinib in hepatocellular carcinoma: Results of a phase 2 placebo-controlled randomized discontinuation study. Ann Oncol. 28:528–534. 2017. View Article : Google Scholar : PubMed/NCBI

98 

Rimassa L, Cheng A, Braiteh F, Benzaghou F, Hazra S, Borgman A, Sinha R, Kayali Z, Zhu A and Kelley R: Phase 3 (COSMIC-312) study of cabozantinib in combination with atezolizumab vs. sorafenib in patients with advanced hepatocellular carcinoma (aHCC) who have not received previous systemic anticancer therapy. Ann Oncol. 30 (Suppl 4):iv65–iv66. 2019. View Article : Google Scholar

99 

Santoro A, Rimassa L, Borbath I, Daniele B, Salvagni S, Van Laethem JL, Van Vlierberghe H, Trojan J, Kolligs FT, Weiss A, et al: Tivantinib for second-line treatment of advanced hepatocellular carcinoma: A randomised, placebo-controlled phase 2 study. Lancet Oncol. 14:55–63. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Wagner AJ, Goldberg JM, Dubois SG, Choy E, Rosen L, Pappo A, Geller J, Judson I, Hogg D, Senzer N, et al: Tivantinib (ARQ 197), a selective inhibitor of MET, in patients with microphthalmia transcription factor-associated tumors: Results of a multicenter phase 2 trial. Cancer. 118:5894–5902. 2012. View Article : Google Scholar : PubMed/NCBI

101 

Yoshioka H, Azuma K, Yamamoto N, Takahashi T, Nishio M, Katakami N, Ahn MJ, Hirashima T, Maemondo M, Kim SW, et al: A randomized, double-blind, placebo-controlled, phase III trial of erlotinib with or without a c-Met inhibitor tivantinib (ARQ 197) in Asian patients with previously treated stage IIIB/IV nonsquamous nonsmall-cell lung cancer harboring wild-type epidermal growth factor receptor (ATTENTION study). Ann Oncol. 26:2066–2072. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Yap TA, Olmos D, Brunetto AT, Tunariu N, Barriuso J, Riisnaes R, Pope L, Clark J, Futreal A, Germuska M, et al: Phase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studies. J Clin Oncol. 29:1271–1279. 2011. View Article : Google Scholar : PubMed/NCBI

103 

Dietrich CG, Gotze O and Geier A: Molecular changes in hepatic metabolism and transport in cirrhosis and their functional importance. World J Gastroenterol. 22:72–88. 2016. View Article : Google Scholar : PubMed/NCBI

104 

Tong Y, Wang M, Huang H, Zhang J, Huang Y, Chen Y and Pan H: Inhibitory effects of genistein in combination with gefitinib on the hepatocellular carcinoma Hep3B cell line. Exp Ther Med. 18:3793–3800. 2019.PubMed/NCBI

105 

Abou-Alfa GK, Meyer T, Cheng AL, El-Khoueiry AB, Rimassa L, Ryoo BY, Cicin I, Merle P, Chen Y, Park JW, et al: Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med. 379:54–63. 2018. View Article : Google Scholar : PubMed/NCBI

106 

Zhang W, Ai J, Shi D, Peng X, Ji Y, Liu J, Geng M and Li Y: Discovery of novel type II c-Met inhibitors based on BMS-777607. Eur J Med Chem. 80:254–266. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Molife LR, Dean EJ, Blanco-Codesido M, Krebs MG, Brunetto AT, Greystoke AP, Daniele G, Lee L, Kuznetsov G, Myint KT, et al: A phase I, dose-escalation study of the multitargeted receptor tyrosine kinase inhibitor, golvatinib, in patients with advanced solid tumors. Clin Cancer Res. 20:6284–6294. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Sun CY, Zhu Y, Li XF, Tang LP, Su ZQ, Wang XQ, Li CY, Yang HM, Zheng GJ and Feng B: Norcantharidin alone or in combination with crizotinib induces autophagic cell death in hepatocellular carcinoma by repressing c-Met-mTOR signaling. Oncotarget. 8:114945–114955. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Katz JD, Jewell JP, Guerin DJ, Lim J, Dinsmore CJ, Deshmukh SV, Pan BS, Marshall CG, Lu W, Altman MD, et al: Discovery of a 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one (MK-2461) inhibitor of c-Met kinase for the treatment of cancer. J Med Chem. 54:4092–4108. 2011. View Article : Google Scholar : PubMed/NCBI

110 

Hughes PE, Rex K, Caenepeel S, Yang Y, Zhang Y, Broome MA, Kha HT, Burgess TL, Amore B, Kaplan-Lefko PJ, et al: In Vitro and in vivo activity of AMG 337, a potent and selective MET kinase inhibitor, in MET-dependent cancer models. Mol Cancer Ther. 15:1568–1579. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Decaens T, Barone C, Assenat E, Wermke M, Fasolo A, Merle P, Blanc JF, Grando V, Iacobellis A, Villa E, et al: Phase 1b/2 trial of tepotinib in sorafenibpretreated advanced hepatocellular carcinoma with MET overexpression. Br J Cancer. Apr 6–2021.(Epub ahead of print). doi: 10.1038/s41416-021-01334-9. View Article : Google Scholar

112 

Qin S, Chan SL, Sukeepaisarnjaroen W, Han G, Choo SP, Sriuranpong V, Pan H, Yau T, Guo Y, Chen M, et al: A phase II study of the efficacy and safety of the MET inhibitor capmatinib (INC280) in patients with advanced hepatocellular carcinoma. Ther Adv Med Oncol. 11:17588359198890012019. View Article : Google Scholar : PubMed/NCBI

113 

Kudo M, Morimoto M, Moriguchi M, Izumi N, Takayama T, Yoshiji H, Hino K, Oikawa T, Chiba T, Motomura K, et al: A randomized, double-blind, placebo-controlled, phase 3 study of tivantinib in Japanese patients with MET-high hepatocellular carcinoma. Cancer Sci. 111:3759–3769. 2020. View Article : Google Scholar : PubMed/NCBI

114 

Giubellino A, Gao Y, Lee S, Lee MJ, Vasselli JR, Medepalli S, Trepel JB, Burke TR Jr and Bottaro DP: Inhibition of tumor metastasis by a growth factor receptor bound protein 2 Src homology 2 domain-binding antagonist. Cancer Res. 67:6012–6016. 2007. View Article : Google Scholar : PubMed/NCBI

115 

Atabey N, Gao Y, Yao ZJ, Breckenridge D, Soon L, Soriano JV, Burke TR Jr and Bottaro DP: Potent blockade of hepatocyte growth factor-stimulated cell motility, matrix invasion and branching morphogenesis by antagonists of Grb2 Src homology 2 domain interactions. J Biol Chem. 276:14308–14314. 2001. View Article : Google Scholar : PubMed/NCBI

116 

Prell RA, Dybdal N, Arima A, Chihaya Y, Nijem I and Halpern W: Placental and fetal effects of onartuzumab, a Met/HGF signaling antagonist, when administered to pregnant cynomolgus monkeys. Toxicol Sci. 165:186–197. 2018. View Article : Google Scholar : PubMed/NCBI

117 

Harding JJ, Zhu AX, Bauer TM, Choueiri TK, Drilon A, Voss MH, Fuchs CS, Abou-Alfa GK, Wijayawardana SR, Wang XA, et al: A Phase Ib/II study of ramucirumab in combination with emibetuzumab in patients with advanced cancer. Clin Cancer Res. 25:5202–5211. 2019. View Article : Google Scholar : PubMed/NCBI

118 

Kim BJ, Kim D, Kim JH, Kim HS and Jang HJ: The efficacy and safety of onartuzumab in patients with solid cancers: A meta-analysis of randomized trials. Indian J Cancer. 58:232–240. 2021.PubMed/NCBI

119 

Scagliotti G, Moro-Sibilot D, Kollmeier J, Favaretto A, Cho EK, Grosch H, Kimmich M, Girard N, Tsai CM, Hsia TC, et al: A randomized-controlled phase 2 study of the MET antibody emibetuzumab in combination with erlotinib as first-line treatment for EGFR mutation-positive NSCLC patients. J Thorac Oncol. 15:80–90. 2020. View Article : Google Scholar : PubMed/NCBI

120 

Vijayaraghavan S, Lipfert L, Chevalier K, Bushey BS, Henley B, Lenhart R, Sendecki J, Beqiri M, Millar HJ, Packman K, et al: Amivantamab (JNJ-61186372), an Fc enhanced EGFR/cMet bispecific antibody, induces receptor downmodulation and antitumor activity by Monocyte/macrophage trogocytosis. Mol Cancer Ther. 19:2044–2056. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Patnaik A, Gordon M, Tsai F, Papadopoulos KP, Rasco D, Beeram M, Fu S, Janku F, Hynes SM, Gundala SR, et al: A phase I study of LY3164530, a bispecific antibody targeting MET and EGFR, in patients with advanced or metastatic cancer. Cancer Chemother Pharmacol. 82:407–418. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Lee BS, Kang S, Kim KA, Song YJ, Cheong KH, Cha HY and Kim CH: Met degradation by SAIT301, a Met monoclonal antibody, reduces the invasion and migration of nasopharyngeal cancer cells via inhibition of EGR-1 expression. Cell Death Dis. 5:e11592014. View Article : Google Scholar : PubMed/NCBI

123 

Strickler JH, LoRusso P, Salgia R, Kang YK, Yen CJ, Lin CC, Ansell P, Motwani M, Wong S, Yue H, et al: Phase I dose-escalation and -expansion study of telisotuzumab (ABT-700), an anti-c-Met antibody, in patients with advanced solid tumors. Mol Cancer Ther. 19:1210–1217. 2020. View Article : Google Scholar : PubMed/NCBI

124 

Hultberg A, Morello V, Huyghe L, De Jonge N, Blanchetot C, Hanssens V, De Boeck G, Silence K, Festjens E, Heukers R, et al: Depleting MET-expressing tumor cells by ADCC provides a therapeutic advantage over inhibiting HGF/MET signaling. Cancer Res. 75:3373–3383. 2015. View Article : Google Scholar : PubMed/NCBI

125 

Vigna E, Chiriaco C, Cignetto S, Fontani L, Basilico C, Petronzelli F and Comoglio PM: Inhibition of ligand-independent constitutive activation of the Met oncogenic receptor by the engineered chemically-modified antibody DN30. Mol Oncol. 9:1760–1772. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Affronti ML, Jackman JG, McSherry F, Herndon JE II, Massey EC Jr, Lipp E, Desjardins A, Friedman HS, Vlahovic G, Vredenburgh J and Peters KB: Phase II study to evaluate the efficacy and safety of Rilotumumab and Bevacizumab in subjects with recurrent malignant glioma. Oncologist. 23:889–e98. 2018. View Article : Google Scholar : PubMed/NCBI

127 

Mok TS, Geater SL, Su WC, Tan EH, Yang JC, Chang GC, Han M, Komarnitsky P, Payumo F, Garrus JE, et al: A randomized phase 2 study comparing the combination of ficlatuzumab and gefitinib with gefitinib alone in Asian patients with advanced stage pulmonary adenocarcinoma. J Thorac Oncol. 11:1736–1744. 2016. View Article : Google Scholar : PubMed/NCBI

128 

Houghton PJ, Kurmasheva RT, Kolb EA, Wu J, Gorlick R, Maris JM and Smith MA: Initial testing (stage 1) of TAK-701, a humanized hepatocyte growth factor binding antibody, by the pediatric preclinical testing program. Pediatr Blood Cancer. 61:380–382. 2014. View Article : Google Scholar : PubMed/NCBI

129 

Kim HJ and Heo K: YYB-101, a humanized antihepatocyte growth factor monoclonal antibody, inhibits ovarian cancer cell motility and proliferation. Anticancer Res. 41:671–678. 2021. View Article : Google Scholar : PubMed/NCBI

130 

Martinez-Montemayor MM, Acevedo RR, Otero-Franqui E, Cubano LA and Dharmawardhane SF: Ganoderma lucidum (Reishi) inhibits cancer cell growth and expression of key molecules in inflammatory breast cancer. Nutr Cancer. 63:1085–1094. 2011. View Article : Google Scholar : PubMed/NCBI

131 

Wang PH, Yang SF, Chen GD, Han CP, Chen SC, Lin LY and Ko JL: Human nonmetastatic clone 23 type 1 gene suppresses migration of cervical cancer cells and enhances the migration inhibition of fungal immunomodulatory protein from Ganoderma tsugae. Reprod Sci. 14:475–485. 2007. View Article : Google Scholar : PubMed/NCBI

132 

Liao CH, Hsiao YM, Hsu CP, Lin MY, Wang JC, Huang YL and Ko JL: Transcriptionally mediated inhibition of telomerase of fungal immunomodulatory protein from Ganoderma tsugae in A549 human lung adenocarcinoma cell line. Mol Carcinog. 45:220–229. 2006. View Article : Google Scholar : PubMed/NCBI

133 

Wu JR, Hu CT, You RI, Ma PL, Pan SM, Lee MC and Wu WS: Preclinical trials for prevention of tumor progression of hepatocellular carcinoma by LZ-8 targeting c-Met dependent and independent pathways. PLoS One. 10:e1144952015.

134 

Gao F, Deng G, Liu W, Zhou K and Li M: Resveratrol suppresses human hepatocellular carcinoma via targeting HGF-c-Met signaling pathway. Oncol Rep. 37:1203–1211. 2017. View Article : Google Scholar : PubMed/NCBI

135 

Chen T, Yuan SJ, Wang J and Hu W: Mechanism of QHF-cisplatin against hepatocellular carcinoma in a mouse model. World J Gastroenterol. 21:10126–10136. 2015. View Article : Google Scholar : PubMed/NCBI

136 

Zhang S, Tang D, Zang W, Yin G, Dai J, Sun YU, Yang Z, Hoffman RM and Guo X: Synergistic inhibitory effect of traditional Chinese medicine astragaloside IV and curcumin on tumor growth and angiogenesis in an orthotopic nude-mouse model of human hepatocellular carcinoma. Anticancer Res. 37:465–473. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2021
Volume 46 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Meng W and Meng W: Association between the HGF/c‑MET signaling pathway and tumorigenesis, progression and prognosis of hepatocellular carcinoma (Review). Oncol Rep 46: 191, 2021
APA
Meng, W., & Meng, W. (2021). Association between the HGF/c‑MET signaling pathway and tumorigenesis, progression and prognosis of hepatocellular carcinoma (Review). Oncology Reports, 46, 191. https://doi.org/10.3892/or.2021.8142
MLA
Meng, W., Chen, T."Association between the HGF/c‑MET signaling pathway and tumorigenesis, progression and prognosis of hepatocellular carcinoma (Review)". Oncology Reports 46.3 (2021): 191.
Chicago
Meng, W., Chen, T."Association between the HGF/c‑MET signaling pathway and tumorigenesis, progression and prognosis of hepatocellular carcinoma (Review)". Oncology Reports 46, no. 3 (2021): 191. https://doi.org/10.3892/or.2021.8142