Next Article in Journal
Regulation of the Tumor-Suppressor Function of the Class III Phosphatidylinositol 3-Kinase Complex by Ubiquitin and SUMO
Previous Article in Journal
Dynamic Changes in Numbers and Properties of Circulating Tumor Cells and Their Potential Applications
Previous Article in Special Issue
Characterization of HGF/Met Signaling in Cell Lines Derived From Urothelial Carcinoma of the Bladder
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Met in Urological Cancers

Department of Urology, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
*
Author to whom correspondence should be addressed.
Cancers 2014, 6(4), 2387-2403; https://doi.org/10.3390/cancers6042387
Submission received: 19 June 2014 / Revised: 3 December 2014 / Accepted: 4 December 2014 / Published: 16 December 2014
(This article belongs to the Special Issue MET in Cancer)

Abstract

:
Met is a tyrosine kinase receptor that is considered to be a proto-oncogene. The hepatocyte growth factor (HGF)-Met signaling system plays an important role in tumor growth, invasion, and metastasis in many types of malignancies. Furthermore, Met expression has been reported to be a useful predictive biomarker for disease progression and patient survival in these malignancies. Many studies have focused on the clinical significance and prognostic role of Met in urological cancers, including prostate cancer (PCa), renal cell carcinoma (RCC), and urothelial cancer. Several preclinical studies and clinical trials are in progress. In this review, the current understanding of the pathological role of Met in cancer cell lines, its clinical significance in cancer tissues, and its predictive value in patients with urological cancers are summarized. In particular, Met-related malignant behavior in castration-resistant PCa and the different pathological roles Met plays in papillary RCC and other histological types of RCC are the subjects of focus. In addition, the pathological significance of phosphorylated Met in these cancers is shown. In recent years, Met has been recognized as a potential therapeutic target in various types of cancer; therapeutic strategies used by Met-targeted agents in urological cancers are summarized in this review.

1. Introduction

Met is a member of cell surface tyrosine kinase (TK) family that was isolated from a chemically transformed human malignant cell line in the 1980s [1]. Hepatocyte growth factor (HGF, also known as scatter factor) is the only endogenous ligand of MET, and HGF/Met signaling can regulate various cellular processes, such as proliferation, apoptosis, and migration [2,3]. Therefore, this signaling is important in physiological activities including maintenance of homeostasis and wound repair [3,4,5].
Since cell proliferation, migration, and angiogenesis are important steps in malignant cell dissemination, many investigators have paid special attention to the relationship between Met overexpression and malignant aggressiveness, including tumor invasion and metastasis. A recent review indicated that enhanced HGF/Met signaling is found in various types of cancers [6]. In addition, Met expression has been reported to be positively associated with tumor growth, invasion, and metastasis in cancer patients [7]. Indeed, our previous reports showed that Met played a crucial role in tumor growth, metastasis, and prognosis in patients with bladder cancer and renal cell carcinoma [8,9].
Many investigators have suggested that evaluation of the expression and biological activity of Met is important in understanding the pathological characteristics, malignant behavior, and strategies of observation and treatment for patients with malignancies. However, it should be noted that there is a wide range of variability in the quantification and pathological significance of Met in previous reports. In particular, variability has been demonstrated in the immunohistochemical technique most commonly used for evaluation of Met expression in animal and human tissues. Possible reasons that have been suggested for such discrepancies include differences in sample size, antibodies used, methods of evaluation, and condition of the tissue samples [8]. It has subsequently been suggested that quantification of phosphorylated Met could be a useful marker of Met signaling activity and Met-related pathological functions; this is discussed in greater detail below. Aberrant MET signaling is the cause of the pathological behavior and malignant aggressiveness of Met in human malignancies. As shown in Figure 1, it is regulated through various mechanisms.
The common mechanism of MET signaling activation is transcriptional deregulation, resulting in MET overexpression in malignant cells. In addition, MET gene mutation and amplification of the MET gene locus, resulting in receptor overexpression, are also characterized aberrancies. Mutation of the MET gene is thought to be a rare event in cancer, but it is often found in papillary renal cell carcinoma (RCC) and childhood hepatocellular carcinoma [9,10]. Conversely, MET gene copy number gain has been reported to be common and is associated with a poor prognosis in patients with lung cancer [11]. The fact that the activation and up-regulation mechanisms of Met in malignancies depend on the type of cancer and background of the patient should be noted when discussing the clinical significance of Met.
HGF-Met signaling has been reported to interact with various cancer-related molecules that promote tumor growth and metastasis. For example, HGF-Met signaling can promote angiogenesis though interaction with the vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) signaling system [3,12]. In addition to VEGF, HGF-Met signaling has been known to mediate angiogenesis through regulation of thrombospondin (TSP)-1, which is a known anti-angiogenic factor [13]. Interestingly, HGF-Met signaling has been found to mediate angiogenesis through positive regulation of VEGF and negative regulation of TSP-1 [13]. Furthermore, Met is reported to be associated with cancer-related activities through various mechanisms including carcinogenesis, cell migration, and chemoresistance [14,15,16].
Figure 1. In cancer, HGF-Met signaling is up-regulated by various systems, including over-expression or mutation of Met and increased HGF production.
Figure 1. In cancer, HGF-Met signaling is up-regulated by various systems, including over-expression or mutation of Met and increased HGF production.
Cancers 06 02387 g001
There are three different strategies to inhibit Met signaling: antagonism of receptor-ligand interaction; inhibition of the TK catalytic activity; and inhibition of the interaction between the receptor and intracellular signaling effectors. By understanding the detailed structure, function, and regulative mechanisms of HGF-Met signaling, it has been possible to develop various Met inhibitors (Table 1). At present, Met inhibitors are divided into two major classes: monoclonal antibodies that compete with HGF for binding to Met and Met TK inhibitors (TKIs). In Table 1, Met inhibitors for various types of malignancies are shown, including those for urological cancers; some of these drugs are in preclinical development and others are in or have completed clinical trials. However, as shown in Table 1, the number of Met-targeted therapies in urological cancer clinical trials is limited. Therefore, the pathological role, clinical significance, and predictive value of Met, and the clinical development of Met-targeted therapy in urological cancers, including PCa, RCC, and urothelial cancer, are reviewed in this manuscript.
Table 1. Met-targeting therapies for malignancies including urological cancers.
Table 1. Met-targeting therapies for malignancies including urological cancers.
Type of MalignancyUrological Cancers (Phase)
Anti-Met mAbs
LY-2875358Lymphoma
OnartuzumabBreast, Colon, Lung, Stomach
Small anti-MET TKIs
Cabozantinib (XL 184)Brain, Breast, Lung, Pancreas, Thyroid *Prostate (II/III), Renal (II/III)
Crizotinib Lung *, Lymphoma
Foretinib (XL 880)Breast, Head and neck, Lung, Stomach Papillary renal (II)
Golvatinib (E7050)Brain, Head and neck, Liver, Stomach
MGCD 265Lung
Tivantinib (ARQ 197)Breast, Colon, Liver, Lung, MyelomaProstate (II), Renal (I/II)
* Approved by U.S. Food and Drug Administration (PDA).

2. Prostate Cancer

PCa is the most common cancer in men. In the United States, it is estimated that 29,000 patients died from PCa in 2013 [17]. Numerous studies have investigated the pathological characteristics and cancer-related changes to the molecules and genes of PCa cells in vivo and in vitro. As mentioned above, the Met TK receptor has been shown to play a role in the proliferation and progression of many types of malignancy, including PCa. In this section, the pathological significance, prognostic roles, and possible therapeutic use of targeting Met in PCa are discussed. In addition, the pathological role of Met expression in castration-resistant PCa (CRPC) is shown.

2.1. The Function of Met Expression in Prostate Cancer Cell Lines

Numerous studies have demonstrated that in human PCa cell lines the MET signaling system plays an important role in cell survival by enhancing cell proliferation and suppressing apoptosis [18,19]. In addition, knockout of the Met gene inhibits tumor cell metastasis [20]. Recent studies have shown that the expression of the tumor suppressor miR-34 is inversely correlated with MET expression in PC-3 cells [21]. In addition, it has been suggested that miR-34 may inhibit the migration of PCa cells by regulating Met activity. Thus, several reported suggested that Met plays a critical role in the malignant aggressiveness of PCa cells.
In discussing the pathological mechanisms of Met in PCa, it is important to consider in vivo and in vitro androgen-sensitivity. PCa cells initially depend upon androgen stimulation, through androgen receptor signaling, for cell survival and malignant behavior. In fact, androgen deprivation is the standard treatment for patients with advanced PCa. However, as the cells become androgen-independent, they become more aggressive. Most recurrent and metastatic tumors that occur after androgen-deprivation therapy include castration-resistant PCa cells [22]. There is currently no curative therapy for CRPC, and patients tend to die due to invasion and metastasis-related pathological conditions.
Among the most widely used human PCa cell lines, MET protein is highly expressed in androgen receptor-negative cell lines (PC-3 and DU145) but only minimally expressed in androgen receptor-positive cell lines (LNCaP, LAPC-4, CWR22, and LUCaP) [23]. In these cell lines, the MET mRNA expression levels are reported to be similar to the protein levels. The androgen receptor-negative PCa cells, PC3 and DU145, have a higher malignant potential, including increased cell proliferation and migration, suppressed apoptosis, and a higher rate of angiogenesis when compared to androgen-receptor-positive cells. Based on these findings, it has been hypothesized that the loss of androgen receptor expression and increased Met expression contribute to the malignant potential of CRPC. Interestingly, continuous androgen-deprivation of the androgen-sensitive PCa cell line, LNCaP, results in decreased androgen receptor expression and increased Met expression [24]. In addition to these in vitro studies, the pathological activity of Met in CRPC has also been confirmed using CRPC cell lines and animal xenograft models [23,25]. From these findings, Met is speculated to be potential therapeutic target for CRPC. In fact, Met inhibitors have been shown to suppress cell proliferation and tumor growth of androgen receptor-negative (androgen-insensitive) PCa cell lines [26].
Furthermore, the importance of cancer stem cells (CSCs) in prostate cancer should be emphasized. According to the CSC hypothesis, tumor growth is sustained by a subpopulation of cancer stem/progenitor-like cells. Like other malignancies, human prostate cancer tissue contains neoplastic cells with self-renewal and clonogenic potential, which can be enriched and perpetuated in prostaspheres [27]. When MET is activated in prostate cancer cells, a stem-like phenotype is induced. MET is thought to regulate tumor infiltration in surrounding tissues by acquisition of a stem-like phenotype [28].

2.2. Expression/Activation of MET in Prostate Cancer and Correlation with Clinicopathological Characteristics

Some investigators have showed the relationships between Met expression and clinicopathological features in patients with prostate cancer [29,30,31,32,33,34]. We list a summary of their results as Table 2. In regard to Gleason score, several reports have indicated MET expression had a positive correlation [29,31,33,34]. On the other hand, there is little information regarding its correlation with TNM stage. Indeed, there is one study that indicates that the expression of MET is not significantly associated with tumor stage, nodal stage, or surgical margin status [32]. Furthermore, although MET expression in metastatic tumors is higher than in primary PCa, it is not a significant predictive factor in patients with a Gleason score of 6 or 7 [31]. Thus, further detailed studies are necessary to decisively determine the clinical significance and pathological role of MET in PCa patients.
Table 2. Relationship between Met expression and pathological features in prostate cancer.
Table 2. Relationship between Met expression and pathological features in prostate cancer.
YearNo. ptsClinicopathological FeaturesRef.
High GSHigh T StagePresence of LN MetastasisPresence of Distant Metastasis
199573p < 0.01[29]
1995128NSp < 0.05[30]
199936p < 0.05[31]
200286NS[32]
200466p < 0.05[33]
20133378p < 0.01NSNS[34]
No. pts; number of patients, GS; Gleason score, LN; lymph node, NS; not significance, Ref; reference.
For CRPC in particular, though, MET expression has been reported to be upregulated and is thought to play a role in angiogenesis, cell invasion, and metastasis to distant tissues [35]. As mentioned previously, in vitro studies have shown a significant correlation between increased Met expression and androgen-independent growth [23,24]. These in vitro and in vivo findings suggest that the expression of Met is correlated with the emergence of castration-resistant tumor growth, and that Met is a potential therapeutic target in CRPC.

2.3. MET-Targeting Therapies for Patients with Prostate Cancer

2.3.1. Cabozantinib

Cabozantinib (XL184) is an orally bioavailable TKI with potent activity against MET and VEGFR2. Cabozantinib blocked the progression of osteolytic and osteoblastic lesions in a xenograft model of CRPC in bone [36]. A phase II, randomized, discontinuation trial has been conducted in patients with advanced PCa, showing that cabozantinib has clinical efficacy in men with CRPC [37]. The trial showed that cabozantinib led to a reduction in soft tissue lesions, improved progression-free survival (PFS), improved resolution of bone scans, decreased levels of bone turnover markers, less pain, and reduced narcotic use. Two phase III clinical trials were done in men with metastatic CRPC. The first is the Cabozantinib Met Inhibition CRPC Efficacy Trial (COMET)-1 (NCT01605227), which aims to evaluate the efficacy of cabozantinib compared to prednisone. In this study, the overall survival of men previously treated for metastatic CRPC with bone-dominant disease, who have experienced disease progression on docetaxel-containing chemotherapy, abiraterone or MDV3100, was assessed. The second study is the Cabozantinib Met Inhibition CRPC Efficacy Trial (COMET)-2 (NCT01522443). This study aims to evaluate the effect of cabozantinib versus mitoxantrone plus prednisone on pain response and bone scan response in men with CRPC. However, COMET-1 did not meet demonstrating a statistically significant increase in overall survival, and based on the outcome of COMET-1, enrollment in COMET-2 was halted.

2.3.2. Tivantinib

Another small molecule inhibitor of Met, tivantinib (formerly ARQ 197), is being studied in PCa. A phase I trial of ARQ 197 was conducted to evaluate the safety of tivantinib. The study showed that tivantinib safely inhibited intratumoral c-MET signaling, but no responses, according to the Response Evaluation Criteria in Solid Tumors, were observed [38]. A phase II trial is ongoing in men with asymptomatic or minimally symptomatic metastatic CRPC (NCT01519414). The purpose of this trial is to investigate the efficacy of tivantinib compared to placebo for treating patients with metastatic PCa.

2.3.3. Sorafenib

Sorafenib is a multikinase inhibitor targeting Raf kinase (serine-threonine kinase) and tyrosine kinases, including VEGFR, platelet-derived growth factor receptor (PDGFR), c-kit, c-Met. A phase II trial of sorafenib monotherapy for CRPC reported relative tolerance of this agent. However, although initially 2 of 22 patients treated with sorafenib showed evidence of improved bony metastatic lesions, most patients (21 of 22) had disease progression [39].

2.3.4. Sunitinib

Sunitinib is an oral multitargeted inhibitor of VEGFR, PDGFR, c-kit, FLT-3, and c-Met. A phase III trial that evaluated angiogenesis-targeted sunitinib therapy in a randomized, double-blind trial of metastatic CRPC concluded that the addition of sunitinib to prednisone did not improve OS compared with placebo in docetaxel-refractory metastatic CRPC [40].

2.3.5. Cause of Failure in MET-Targeting Therapies

As mentioned above, various types of MET inhibitors have been studied in clinical trials. However, unfortunately, almost of them showed “negative” results. As reason of such failure, several factors including study design, patient’s selection, and unexpected side effect are speculated. In this review, we emphasize the fact that such molecular-targeting therapies used for patients with CRPC. In addition, in many cases, CRPC patients were previously treated with docetaxel prior to such therapies. From these facts, we believe that more detailed information regarding pathological roles of Met expression in patients with CRPC are essential to improve the anti-tumor effect.

3. Renal Cell Carcinoma

Renal cell carcinoma (RCC) is the third most frequent cancer originating from the genitourinary organs. It is estimated that 65,000 patients were diagnosed with RCC in the United States in 2013 [17]. RCC originates from the proximal tubule of the kidney or the collecting duct and is classified into four major histological types: clear cell (conventional), papillary, chromophobe, and collecting duct. Among these, clear cell (conventional) RCC is the most common histological type and accounts for 75%–80% of all cases. Papillary RCC is the second most common histological type and accounts for 10%–15% of cases. Papillary RCC can be further divided into two morphological subtypes; type 1 consists of predominantly basophilic cells and type 2 consists of mostly eosinophilic cells. In recent years, cytogenetic and molecular studies have indicated that these different RCC pathological types possess different characteristics and respond differently to therapies. For example, papillary RCC is more frequent in male patients and tends to be of a low stage compared to clear cell or chromophobe RCC [41]. Therefore, it is necessary to discuss the pathological role of Met according to the different subtypes. Inactivation of the von Hippel-Lindau (VHL) gene should also be considered in this discussion of the pathological characteristics and molecular mechanisms of RCC.

3.1. The Function of Met Expression in Papillary RCC

The clinical significance and pathological role of MET has been most widely investigated, and is best understood, in papillary RCC. Several activating missense mutations of the MET gene have been described in individuals with papillary RCC [42]. Several mutations in the MET TK domain, both in the germline of hereditary papillary RCC families (M1131T, V1188L, D1228H, Y1230C) and in a subset of sporadic papillary RCC patients (L1195V, D1228H, Y1230H, M1250T), have been described [43]. Similar trends in hereditary papillary RCC have also been reported in another study [44]. Research on MET and papillary RCC reported the interaction between c-MET and VEGF; the development of resistance to VEGF-targeted therapy correlated with the up-regulation of c-MET expression. This result suggests that the acquired resistance to VEGF pathway inhibitors is frequently mediated by the activation of alternative signaling pathway (angiogenic escape), which induces c-MET overexpression [45].

3.2. The Function of Met Expression in Clear Cell RCC and Other RCCs

The most well-known form of inherited clear cell RCC is that associated with VHL syndrome. Inactivation of VHL gene function by mutation or methylation is found in patients with sporadic clear cell RCC, although it is rare in other histological RCC types. Several reports have found that Met protein is detectable in human clear cell RCC tissues [46,47,48,49]. VHL expression in RCC cells suppresses MET/β-catenin signaling, and a loss of VHL in conventional RCC has been shown to enable MET/β-catenin signaling in vitro [50]. Thus, it has been suggested that Met plays important roles in the carcinogenesis and malignant behavior of clear cell RCC via direct and indirect mechanisms.

3.3. Expression/Activation of MET in Renal Cell Carcinomas and Correlation with Clinicopathological Features

Strong expression of MET is observed in almost all papillary RCC and collecting duct carcinoma, however, it is uncommon in clear cell RCC and chromophobe RCC [48]. Gibney et al. have also reported that MET expression in papillary RCC and sarcomatous RCC is significantly higher than in clear cell RCC subtypes [51]. Thus, Met overexpression is speculated to be a characteristic of papillary RCC. It has been reported that MET expression is positively correlated with the nuclear grade, sarcomatoid component, and lymphatic invasion in patients with clear cell RCC [48]. Furthermore, the presence of phosphorylated MET is reported to be significantly correlated with malignancy aggressiveness and prognosis in clear cell RCC [52]. The clinical significance and prognostic role of MET in other types of RCC, including chromophobe and collecting duct carcinoma, is not fully understood. A summary of the correlations between MET expression and the clinicopathological features including malignant potential and survival of patients with RCC is shown in Table 3.
Table 3. Relationship between MET expression and grade, tumor stage, and prognosis for patients with renal cell carcinoma.
Table 3. Relationship between MET expression and grade, tumor stage, and prognosis for patients with renal cell carcinoma.
YearNo. ptsPathological FeaturesSurvivalRef.
High GradepT StageLymph Node MetastasisDistant Metastasis
2006114NSNSNSNSNS[52]
200696p < 0.01NSNSNS-[48]
200766-P < 0.01-p < 0.05p < 0.01[47]
2013317p < 0.01---p < 0.05[51]
No. pts; number of patients, NS; not significant, Ref.; reference.

3.4. Met-Targeted Therapies for Patients with Renal Cell Carcinoma

3.4.1. Foretinib (XL880)

The drug foretinib, formerly XL880, is a multi-targeted receptor TKI that targets the MET, VEGFR2, platelet derived growth factor receptor (PDGFR) β, Tie-2, recepteur d'origine nantais (RON), KIT, and FLT3 receptors. A phase I study of foretinib has been completed, and two out of four patients with papillary RCC showed a partial response (PR) [53]. A phase II study of foretinib in patients with papillary RCC was open-label and nonrandomized [54]. In this study, 74 patients were enrolled and an overall response rate of 13.5% with a median PFS of 9.3 months was demonstrated. The most frequent adverse events in patients with any grade RCC were fatigue, hypertension, gastrointestinal toxicities, and pulmonary emboli. Interestingly, among patients with germline MET mutations, five (50%) of ten experienced a PR, and the remaining five (50%) patients had stable disease as the best response. A PR was seen in only five (9%) of 57 patients without germline MET mutations. Further investigations are necessary to clarify these results.

3.4.2. Tivantinib (ARQ197)

Tivantinib (ARQ197) is an inhibitor of Met receptor TK activity. In vitro studies indicate that tivantinib inhibits colony formation in ACHN and 769P cells [51]. Following completion of a phase I dose-finding study [55], phase II trials are ongoing in patients with metastatic papillary RCC (NCT00345423).

3.4.3. Cabozantinib (XL184)

Recently, a phase I study of cabozantinib (XL184), a small molecule inhibitor of TKs including MET and VEGFR2, was performed [56]. The study enrolled 25 patients with metastatic clear cell RCC for whom standard therapy had failed. Common adverse events included fatigue, diarrhea, nausea, proteinuria, decreased appetite, palmar-plantar erythrodysesthesia, and vomiting. PR was reported in seven patients (28%) with a median PFS of 12.9 months and a median overall survival of 15.0 months. From these preliminary data, a phase II study randomizing patients to cabozantinib or sunitinib in the first-line setting for mRCC has been initiated [45]. In papillary RCC, a special role for c-MET inhibitors may emerge because of the critical role of activating c-MET mutations [46].

4. Urothelial Cancer

Urothelial carcinoma (UC), especially bladder cancer, is the second most common genitourinary malignancy; it accounts for almost 5% of all newly diagnosed cancers. It is estimated that 72,570 patients were newly diagnosed with UC of the bladder in the United States in 2013. More than 90% of cases pathologically originate from urothelial cells. Papillary UCs are typically superficial at first presentation and are often multifocal. These tumors recur with a high frequency (>60%), and 10%–15% of them will progress to life-threatening malignancies over a longer period of time [17,57]. Several risk factors can be used to predict tumor progression, including histological grading, stage classification, tumor morphology, and the size and number of tumors [58,59,60,61,62,63]. The number of tumors and their classification stage are considered the most important prognostic factors for recurrence [59,60,61,62,63,64]. Patients with invasive and/or metastatic lesions have a poor prognosis. Tumor invasion and metastasis are regulated by complex mechanisms, including tumor growth, degeneration of stromal tissues, cell migration, and angiogenesis. Moreover, bladder cancer is characterized by distinct molecular characteristics. For example, FGR3 mutations are associated with low-stage and low-grade tumors, whereas p53 mutations are associated with high-stage and high-grade tumors [65,66]. Therefore, detailed information regarding these mechanisms is essential to formulate appropriate treatment and observation strategies for patients with UC [67]. In addition to MET expression, interaction with the Axl and PDGFR-α pathways contributes to bladder cancer progression [68]. RON, a macrophage-stimulating protein, is a distinct receptor TK in the Met proto-oncogene family [69]. Co-expression of RON and MET is significantly associated with decreased overall and metastasis-free survival [70]. RON-associated signaling may also play an important role in the progression of human bladder cancer. Some researchers have suggested that cross-talk exists between the RON and Met pathways [71,72]. Evaluation of RON and Met expression status may help to identify a subset of bladder cancer patients who require more intensive treatment.

4.1. Function of Met Expression in UC

In UC cell lines, Met mRNA has been detected by northern blot analysis [73] and reverse transcriptase polymerase chain reaction [74]. In the absence of point mutations, the expression of Met tends to positively correlate with cancer cell line differentiation [59]. However, since most of these studies have been in vitro, a detailed understanding of the pathological significance and regulative mechanism of MET in UC cells has not been achieved.

4.2. Expression/Activation of Met in UC and Correlation with Clinicopathological Characteristics

It was initially reported that Met expression could not be detected in the normal urothelium or in cases of UC of the kidney [75]. However, more recently, Met expression in urothelial cells, endothelial cells, and smooth muscle cells of the urinary organs has been detected [76]. A previous study had reported that the expression of Met in UC cells of the urinary bladder was positively associated with histological grading, non-papillary contour, tumor size, and muscle invasion [77]. In another study, antibody arrays were used to demonstrate that MET was strongly expressed in high-grade cases of bladder cancer [78]. However, some researchers have found contradictory results (Table 4). Thus, there is no general agreement on the relationship between MET expression and pathological features in patients with UC. However, in recent years, the consensus is that Met may play an important role in the carcinogenesis and malignancy aggressiveness in patients with UC [76,79,80]. Strong MET expression has been demonstrated in an invasive phenotype of bladder cancer [76], although varying levels of MET immunostaining are consistently detected in UC of the bladder [59]. In regard to correlation between MET expression and outcome in patients with bladder cancer, several reports have indicated that MET levels do correlate with the progression and several studies have shown that its overexpression is associated with shortened metastasis-free and overall survival in patients with bladder cancer [68,77,78]. These pathological significances and predictive roles of MET expression were showed in Table 4.
Table 4. Relationship between MET expression and pathological features and survival in urothelial carcinoma.
Table 4. Relationship between MET expression and pathological features and survival in urothelial carcinoma.
YearNo. ptsClinicopathological FeaturesOutcomeRef.
GradeT StageN StageM StagePDSurvival
199849NSNSNS- NS[79]
2002142p < 0.01p < 0.01p < 0.01-p < 0.01p < 0.05[59]
2005183p < 0.01p < 0.01-NSp < 0.05[69]
2006173p < 0.01p < 0.01p < 0.01- p < 0.05[78]
2009133p < 0.01p < 0.01p = 0.01 NSNS[52]
201175NSNSNSNS p < 0.01[68]
No. pts; number of patients, PD, progression of disease, Ref; reference, NS; not significant.
The biological activities of Met are exerted by phosphorylation, and phosphorylation of Met is essential for the stimulation of downstream signaling pathways. For this reason, Miyata et al. [52] investigated the clinical significance and pathological function of MET phosphorylation in human bladder cancer tissues and found that MET expression was not a significant predictor of metastasis-free or cause-specific survival. However, phosphorylated MET was significantly associated with tumor aggressiveness and prognosis in patients with bladder cancer patients. In addition, among the different MET phosphorylation sites, phosphorylation of Y1349 was thought to play an important role in metastasis and survival. Phospho-Y1349 MET was correlated with the expression of matrix metalloproteinase-2 and -7, and E-cadherin [52].

4.3. Met-Targeted Therapy for Patients with Urothelial Carcinoma

Based on the pathological significance and prognostic role of MET in UC cells, many investigators have suggested that Met is a potential novel therapeutic target for these patients. SU-11274, a selective MET kinase inhibitor, is reported to affect the growth of bladder cancer [81]. In addition, several reports have shown that tumor growth and invasion by UC is inhibited by regulation of the HGF-Met signaling system [82,83]. However, to our knowledge, no clinical trial using a Met inhibitor is ongoing at present. Furthermore, a combination of TKIs could induce a synergistic antitumor effect, and improve therapeutic efficacy [84]. In the future, detection of Met co-expression molecules, combined with more selective, or multi-targeting, TKIs could enable individualized therapy in UC.

5. Conclusions

In this review, we have summarized the pathological role, clinical significance, and predictive value of Met in urological cancer in vivo and in vitro. In addition, we have reviewed the preclinical studies and clinical trials of Met-targeted therapies in patients with urological cancers. Numerous investigators have focused on the functions of Met at a genetic, molecular, and clinical level. However, much is still unknown. Based on previous reports and current studies, we believe that Met has the potential to be used as a predictive factor and therapeutic target in patients with urological cancers.

Acknowledgements

This review was supported by Grant-in-Aid for Scientific research from the Japanese Government to Yasuyoshi Miyata (No. 25462487). However, it was not supported financially by any commercial entity.

Author Contributions

Yasuyoshi Miyata: Literature review and preparation of the manuscript; Akihiro Asai: Literature review and preparation of the manuscript; Kensuke Mitsunari: Literature review and preparation of the manuscript; Tomohiro Matsuo: Literature review and preparation of the manuscript, Kojiro Ohba: Literature review and preparation of the manuscript; Yasushi Mochizuki: Preparation of the manuscript; Hideki Sakai: Preparation of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cooper, C.S.; Park, M.; Blair, D.G.; Tainsky, M.A.; Huebner, K.; Croce, C.M.; Vande Woude, G.F. Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature 1984, 311, 29–33. [Google Scholar] [CrossRef] [PubMed]
  2. Birchmeier, C.; Birchmeier, W.; Gherardi, E.; Vande Wounde, G.F. Met, metastasis, mortality and more. Nat. Rev. Mol. Cell Biol. 2003, 4, 915–925. [Google Scholar] [CrossRef] [PubMed]
  3. Trusolino, L.; Bertotti, A.; Comoglio, P.M. MET signaling: Principles and functions in development, organ regeneration and cancer. Nat. Rev. Mol. Cell Biol. 2010, 11, 834–848. [Google Scholar] [CrossRef] [PubMed]
  4. Bussolino, F.; di Renzo, M.F.; Ziche, M.; Bocchietto, E.; Olivero, M.; Naldini, L.; Gaudino, G.; Tamagnone, L.; Coffer, A.; Comoglio, P.M. Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell motility and growth. J. Cell Biol. 1992, 119, 629–641. [Google Scholar] [CrossRef] [PubMed]
  5. Kagoshima, M.; Kinoshita, T.; Matsumoto, K.; Nakamura, T. Developmental changes in hepatocyte growth factor mRNA and its receptor in rat liver, kidney and lung. Eur. J. Biochem. 1992, 210, 375–380. [Google Scholar] [CrossRef] [PubMed]
  6. Yano, S.; Nakagawa, T. The current state of molecularly targeted drugs tragting HGF/Met. Jpn. J. Clin. Oncol. 2014, 44, 9–12. [Google Scholar] [CrossRef] [PubMed]
  7. Scagliotti, G.V.; Novello, S.; von Pawel, J. The emerging role of MET/HGF inhibitors in oncology. Cancer Treat. Rev. 2013, 39, 793–801. [Google Scholar] [CrossRef] [PubMed]
  8. Koeppen, H.; Rost, S.; Yauch, R. Developing biomarkers to predict benefit from HGF/MET pathway inhibitors. J. Pathol. 2014, 232, 210–218. [Google Scholar] [CrossRef] [PubMed]
  9. Lubensky, I.A.; Schmidt, L.; Zhuang, Z.; Weirich, G.; Pack, S.; Zambrano, N.; Walther, M.M.; Choyke, P.; Linehan, W.M.; Zbar, B. Hereditary and sporadic papillary renal cell carcinoma with c-met mutations share a distinct morphological phenotype. Am. J. Pathol. 1999, 155, 517–526. [Google Scholar] [CrossRef] [PubMed]
  10. Park, W.S.; Dong, S.M.; Kim, S.Y.; Na, E.Y.; Shin, M.S.; Pi, J.H.; Kim, B.J.; Bae, J.H.; Hong, Y.K.; Lee, K.S.; et al. Somatic mutations in the kinase domain of the Met/hepatocyte growth factor receptor gene in childhood hepatocellular carcinomas. Cancer Res. 1999, 59, 307–310. [Google Scholar]
  11. Toschi, L.; Cappuzzo, F. Clinical implications of MET gene copy number in lung cancer. Future Oncol. 2010, 6, 239–247. [Google Scholar] [CrossRef] [PubMed]
  12. Grant, D.S.; Kleinman, H.K.; Goldberg, I.D.; Bhargava, M.M.; Nickoloff, B.J.; Kinsella, J.L.; Polverini, P.; Rosen, E.M. Scatter factor induces blood vessel formation in vivo. Proc. Natl. Acad. Sci. USA 1993, 90, 1937–1941. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, Y.W.; Su, Y.; Volpert, O.V.; Vande Wounde, G.F. Hepatocyte growth factor/scatter factor mediates angiogenesis through positive VEGF and negative thrombospondin 1 regulation. Proc. Natl. Acad. Sci. USA 2003, 100, 12718–12723. [Google Scholar] [CrossRef] [PubMed]
  14. Delitto, D.; Vertes-George, E.; Hughes, S.J.; Behrns, K.E.; Trevino, J.G. c-Met signaling in the development of tumorgenesis and chemoresitance: Potential applications in pancreatic cancer. World J. Gastroentrol. 2014, 20, 8458–8470. [Google Scholar] [CrossRef]
  15. Peng, Y.; Guo, J.J.; Liu, Y.M.; Wu, X.L. MicroRNA-34A inhibits the growth, invasion and metastasis of gastric cancer by targeting PDGFR and MET expression. Biosci. Rep. 2014, 34, 247–256. [Google Scholar] [CrossRef]
  16. Ueki, R.; Sando, S. A DNA adaptor to c-Met inhibits cancer cell migration. Chem. Commun. 2014, 50, 13131–13134. [Google Scholar] [CrossRef]
  17. Siegel, R.; Naishadham, D.; Jemal, A. Cancer statistics, 2013. CA Cancer J. Clin. 2013, 63, 11–30. [Google Scholar] [CrossRef] [PubMed]
  18. Yue, D.; Wang, Y.; Ma, P.; Li, Y.Y.; Chen, H.; Wang, P.; Ren, C.S. Effects of transferred NK4 gene on proliferation, migration, invasion and apoptosis of human prostate cancer DU145 cells. Asian J. Androl. 2010, 12, 381–389. [Google Scholar] [CrossRef] [PubMed]
  19. Sankpal, U.T.; Abdelrahim, M.; Connelly, S.F.; Lee, C.M.; Madero-Visbal, R.; Colon, J.; Smith, J.; Safe, S.; Maliakal, P.; Basha, R. Small molecule tolfenamic acid inhibits PC-3 cell proliferation and invasion in vitro, and tumor growth in orthotopic mouse model for prostate cancer. Prostate 2012, 72, 1648–1658. [Google Scholar] [CrossRef] [PubMed]
  20. Nishida, S.; Hirohashi, Y.; Torigoe, T.; Inoue, R.; Kitamura, H.; Tanaka, T.; Takahashi, A.; Asanuma, H.; Masumori, N.; Tsukamoto, T.; et al. Prostate cancer stem-like cells/cancer-initiating cells have an autocrine system of hepatocyte growth factor. Cancer Sci. 2013, 104, 431–436. [Google Scholar]
  21. Hagman, Z.; Haflidadottir, B.S.; Ansari, M.; Persson, M.; Bjartell, A.; Edsjö, A.; Ceder, Y. The tumour suppressor miR-34c targets MET in prostate cancer cells. Br. J. Cancer 2013, 109, 1271–1278. [Google Scholar] [CrossRef] [PubMed]
  22. Niu, Y.; Altuwaijri, S.; Lai, K.P.; Wu, C.T.; Ricke, W.A.; Messing, E.M.; Yao, J.; Yeh, S.; Chang, C. Androgen receptor is a tumor suppressor and proliferator in prostate cancer. Proc. Natl. Acad. Sci. USA 2008, 105, 12182–12187. [Google Scholar] [CrossRef] [PubMed]
  23. Verras, M.; Lee, J.; Xue, H.; Li, T.H.; Wang, Y.; Sun, Z. The androgen receptor negatively regulates the expression of c-Met: Implications for a novel mechanism of prostate cancer progression. Cancer Res. 2007, 67, 967–975. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, T.; Mendes, D.E.; Berkman, C.E. From AR to c-Met: Androgen deprivation leads to a signaling pathway switch in prostate cancer cells. Int. J. Oncol. 2013, 43, 1125–1130. [Google Scholar] [PubMed]
  25. Knudsen, B.S.; Edlund, M. Prostate cancer and the met hepatocyte growth factor receptor. Adv. Cancer Res. 2004, 91, 31–67. [Google Scholar] [PubMed]
  26. Tu, W.H.; Zhu, C.; Clark, C.; Christensen, J.G.; Sun, Z. Efficacy of c-Met inhibitor for advanced prostate cancer. BMC Cancer 2010, 10. [Google Scholar] [CrossRef] [PubMed]
  27. Guzmán-Ramírez, N.; Völler, M.; Wetterwald, A.; Germann, M.; Cross, N.A.; Rentsch, C.A.; Schalken, J.; Thalmann, G.N.; Cecchini, M.G. In vitro propagation and characterization of neoplastic stem/progenitor-like cells from human prostate cancer tissue. Prostate 2009, 69, 1683–1693. [Google Scholar] [CrossRef] [PubMed]
  28. Van Leenders, G.J.L.H.; Sookhlall, R.; Teubel, W.J.; de Ridder, C.M.A.; Reneman, S.; Sacchetti, A.; Vissers, K.J.; van Weerden, W.; Jenster, G. Activation of c-MET induces a stem-like phenotype in human prostate cancer. PLoS One 2011, 6, e26753. [Google Scholar] [CrossRef] [PubMed]
  29. Pisters, L.L.; Troncoso, P.; Zhau, H.E.; Li, W.; von Eschenbach, A.C.; Chung, L.W. c-Met proto-oncogene expression in benign and malignant human prostate tissues. J. Urol. 1995, 154, 293–298. [Google Scholar] [CrossRef] [PubMed]
  30. Humphrey, P.A.; Zhu, X.; Zarnegar, R.; Swanson, P.E.; Ratliff, T.L.; Vollmer, R.T.; Day, M.L. Hepatocyte growth factor and its receptor (c-MET) in prostatic carcinoma. Am. J. Pathol. 1995, 147, 386–396. [Google Scholar] [PubMed]
  31. Watanabe, M.; Fukutome, K.; Kato, H.; Murata, M.; Kawamura, J.; Shiraishi, T.; Yatani, R. Progression-linked overexpression of c-Met in prostatic intraepithelial neoplasia and latent as well as clinical prostate cancers. Cancer Lett. 1999, 141, 173–178. [Google Scholar] [CrossRef] [PubMed]
  32. Knudsen, B.S.; Gmyrek, G.A.; Inra, J.; Scherr, D.S.; Vaughan, E.D.; Nanus, D.M.; Kattan, M.W.; Gerald, WL.; Vande Woude, G.F. High expression of the Met receptor in prostate cancer metastasis to bone. Urology 2002, 60, 1113–1117. [Google Scholar] [CrossRef] [PubMed]
  33. Strohmeyer, D.; Strauss, F.; Rössing, C.; Roberts, C.; Kaufmann, O.; Bartsch, G.; Effert, P. Expression of bFGF, VEGF and c-met and their correlation with microvessel density and progression in prostate carcinoma. Anticancer Res. 2004, 24, 1797–1804. [Google Scholar] [PubMed]
  34. Jacobsen, F.; Ashtiani, S.N.; Tennstedt, P.; Heinzer, H.; Simon, R.; Sauter, G.; Sirma, H.; Tsourlakis, M.C.; Minner, S.; Schlomm, T.; et al. High c-MET expression is frequent but not associated with early PSA recurrence in prostate cancer. Exp. Ther. Med. 2013, 5, 102–106. [Google Scholar] [PubMed]
  35. Pfeiffer, M.J.; Smit, F.P.; Sedelaar, J.P.; Schalken, J.A. Steroidogenic enzymes and stem cell markers are upregulated during androgen deprivation in prostate cancer. Mol. Med. 2011, 17, 657–664. [Google Scholar] [CrossRef] [PubMed]
  36. Schimmoller, F.; Zayzafoon, M.; Chung, L.W.K.; Zhau, H.E.; Fagerlund, K.M.; Aftab, D.T. Cabozantinib (XL184), a dual MET-VEGFR2 inhibitor, blocks osteoblastic and osteolytic progression of human prostate cancer xenografts in mouse bone. Mol. Cancer Ther. 2011, 10, A233. [Google Scholar] [CrossRef]
  37. Smith, D.C.; Smith, M.R.; Sweeney, C.; Elfiky, A.A.; Logothetis, C.; Corn, P.G.; Vogelzang, N.J.; Small, E.J.; Harzstark, A.L.; Gordon, M.S.; et al. Cabozantinib in patients with advanced prostate cancer: Results of a phase II randomized discontinuation trial. J. Clin. Oncol. 2013, 31, 412–419. [Google Scholar]
  38. Yap, T.A.; Olmos, D.; Brunetto, A.T.; 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. 2011, 29, 1271–1279. [Google Scholar]
  39. Dahut, W.L.; Scripture, C.; Posadas, E.; Jain, L.; Gulley, J.L.; Arlen, P.M.; Wright, J.J.; Yu, Y.; Cao, L.; Steinberg, S.M.; et al. A phase II clinical trial of sorafenib in androgen-independent prostate cancer. Clin. Cancer Res. 2008, 14, 209–214. [Google Scholar]
  40. Michaelson, M.D.; Oudard, S.; Ou, Y.C.; Sengeløv, L.; Saad, F.; Houede, N.; Ostler, P.; Stenzl, A.; Daugaard, G; Jones, R.; et al. Randomized, placebo-controlled, phase III trial of sunitinib plus prednisone versus prednisone alone in progressive, metastatic, castration-resistant prostate cancer. J. Clin. Oncol. 2013, 32, 76–82. [Google Scholar]
  41. Beck, S.D.; Patel, M.I.; Snyder, M.E.; Kattan, M.W.; Motzer, R.J.; Reuter, V.E.; Russo, P. Effect of papillary and chromophobe cell type on disease-free survival after nephrectomy for renal cell carcinoma. Ann. Surg. Oncol. 2004, 11, 71–77. [Google Scholar] [CrossRef] [PubMed]
  42. Giubellino, A.; Linehan, W.M.; Bottaro, D.P. Targeting the Met signaling pathway in renal cancer. Expert Rev. Anticancer Ther. 2009, 9, 785–793. [Google Scholar] [CrossRef] [PubMed]
  43. Schmidt, L.; Duh, F.M.; Chen, F.; Kishida, T.; Glenn, G.; Choyke, P.; Scherer, S.W.; Zhuang, Z.; Lubensky, I.; Dean, M.; et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat. Genet. 1997, 16, 68–73. [Google Scholar]
  44. Schmidt, L.; Junker, K.; Weirich, G.; Glenn, G.; Choyke, P.; Lubensky, I.; Zhuang, Z.; Jeffers, M.; Vande, W.G.; Neumann, H.; et al. Two North American families with hereditary papillary renal carcinoma and identical novel mutations in the MET protooncogene. Cancer Res. 1998, 58, 1719–1722. [Google Scholar]
  45. Zbar, B.; Tory, K.; Merino, M.; Schmidt, L.; Glenn, G.; Choyke, P.; Walther, M.M.; Lerman, M.; Linehan, W.M. Hereditary papillary renal cell carcinoma. J. Urol. 1994, 151, 561–566. [Google Scholar] [PubMed]
  46. Schmidt, L.; Junker, K.; Nakaigawa, N.; Kinjerski, T.; Weirich, G.; Miller, M.; Lubensky, I.; Neumann, H.P.; Brauch, H.; Decker, J.; et al. Novel mutations of the MET proto-oncogene in papillary renal carcinomas. Oncogene 1999, 18, 2343–2350. [Google Scholar]
  47. Betsunoh, H.; Mukai, S.; Akiyama, Y.; Fukushima, T.; Minamiguchi, N.; Hasui, Y.; Osada, Y.; Kataoka, H. Clinical relevance of hepsin and hepatocyte growth factor activator inhibitor type 2 expression in renal cell carcinoma. Cancer Sci. 2007, 98, 491–498. [Google Scholar] [CrossRef] [PubMed]
  48. Choi, J.S.; Kim, M.K.; Seo, J.W.; Choi, Y.L.; Kim, D.H.; Chun, Y.K.; Ko, Y.H. MET expression in sporadic renal cell carcinomas. J. Korea Med. Sci. 2006, 21, 672–677. [Google Scholar] [CrossRef]
  49. Oh, R.R.; Park, J.Y.; Lee, J.H.; Shin, M.S.; Kim, H.S.; Lee, S.K.; Kim, Y.S.; Lee, S.H.; Lee, S.N.; Yang, Y.M.; et al. Expression of HGF/SF and Met protein is associated with genetic alterations of VHL gene in primary renal cell carcinomas. APMIS 2002, 110, 229–238. [Google Scholar]
  50. Peruzzi, B.; Athauda, G.; Bottaro, D.P. The von Hippel-Lindau tumor suppressor gene product represses oncogenic beta-catenin signaling in renal carcinoma cells. Proc. Natl. Acad. Sci. USA 2006, 103, 14531–14536. [Google Scholar] [CrossRef] [PubMed]
  51. Gilney, G.T.; Aziz, S.A.; Camp, R.L.; Conrad, P.; Schwartz, B.E.; Chen, C.R.; Kelly, W.K.; Kluger, H.M. c-Met is a prognostic marker and potential therapeutic target in clear cell renal cell carcinoma. Ann. Oncol. 2013, 24, 343–349. [Google Scholar] [CrossRef] [PubMed]
  52. Miyata, Y.; Sagara, Y.; Kanda, S.; Hayashi, T.; Kanetake, H. Phosphorylated hepatocyte growth factor receptor/c-Met is associated with tumor growth and prognosis in patients with bladder cancer: Correlation with matrix metalloproteinase-2 and -7 and E-cadherin. Hum. Pathol. 2009, 40, 496–504. [Google Scholar] [CrossRef] [PubMed]
  53. Eder, J.P.; Shapiro, G.I.; Appleman, L.J.; Zhu, A.X.; Miles, D.; Keer, H.; Cancilla, B.; Chu, F.; Hitchcock-Bryan, S.; Sherman, L.; et al. A phase I study of foretinib, a multi-targeted inhibitor of c-Met and vascular endothelial growth factor receptor 2. Clin. Cancer Res. 2010, 16, 3507–3516. [Google Scholar]
  54. Choueiri, T.K.; Vaishampayan, U.; Rosenberg, J.E.; Logan, T.F.; Harzstark, A.L.; Bukowski, R.M.; Rini, B.I.; Srinivas, S.; Stein, M.N.; Adams, L.M.; et al. A phase II and biomarker study of the dual MET/VEGFR-2 inhibitor foretinib in patients with papillary renal cell cancer. J. Clin. Oncol. 2013, 31, 181–186. [Google Scholar]
  55. Garcia, A.; Rosen, L.; Cunningham, C.C.; Nemunaitis, J.; Li, C.; Rulewski, N.; Dovholuk, A.; Savage, R.; Chan, T.; Bukowksi, R.; et al. Phase 1 study of ARQ 197, a selective inhibitor of the c-Met RTK in patients with metastatic solid tumors reaches recommended phase 2 dose. J. Clin. Oncol. 2007, 25. Abstract No 3525. [Google Scholar]
  56. Choueiri, T.K.; Pal, S.K.; McDermott, D.F.; Morrissey, S.; Ferguson, K.C.; Holland, J.; Kaelin, W.G., Jr.; Dutcher, J.P. A phase I study of cabozantinib (XL184) in patients with RCC. Ann. Oncol. 2014, in press. [Google Scholar]
  57. Stein, J.P.; Grossfeld, G.D.; Ginberg, D.A.; Esrig, D.; Freeman, J.A.; Figueroa, A.J.; Skinner, D.; Cote, R. Prognostic markers in bladder cancer: A contemporary review of the literature. J. Urol. 1998, 160, 645–659. [Google Scholar] [CrossRef] [PubMed]
  58. Allard, P.; Bernard, P.; Frader, Y.; Tetu, B. The early clinical course of primary Ta and T1 bladder cancer: A proposed prognostic index. Br. J. Urol. 1998, 81, 692–698. [Google Scholar] [CrossRef] [PubMed]
  59. Cheng, H.L.; Trink, B.; Tzai, T.S.; Liu, H.S.; Chan, S.H.; Ho, C.L.; Sidransky, D.; Chow, N.H. Overexpression of c-met as a prognostic indicator for transitional cell carcinoma of the urinary bladder: A comparison with p53 nuclear accumulation. J. Clin. Oncol. 2002, 20, 1544–1550. [Google Scholar] [CrossRef] [PubMed]
  60. Millán-Rodríguez, F.; Chéchile-Toniolo, G.; Salvador-Bayarri, J.; Palou, J.; Vicente-Rodríguez, J. Multivariate analysis of the prognostic factors of primary superficial bladder cancer. J. Urol. 2000, 163, 73–78. [Google Scholar] [CrossRef] [PubMed]
  61. Holmang, S.; Hedelin, H.; Anderstrom, C.; Johansson, S.L. The relationship among multiple recurrences, progression and prognosis of patients with stages Ta and T1 transitional cell cancer of the bladder followed for at least 20 years. J. Urol. 1995, 153, 1823–1827. [Google Scholar] [CrossRef] [PubMed]
  62. Crew, J.P.; O’Brien, T.; Bradburn, M.; Fuggle, S.; Bicknell, R.; Cranston, D.; Harris, A.L. Vascular endothelial growth factor is predictor of relapse and stage progression in superficial bladder cancer. Cancer Res. 1997, 57, 5281–5285. [Google Scholar] [PubMed]
  63. Chow, N.H.; Liu, H.S.; Chan, S.H. The role of nm23-H1 in the progression of transitional cell bladder cancer. Clin. Cancer Res. 2000, 6, 3595–3599. [Google Scholar] [PubMed]
  64. Chow, N.H.; Liu, H.S.; Chan, S.H.; Cheng, H.L.; Tzai, T.S. Expression of vascular endothelial growth factor in primary superficial bladder cancer. Anticancer Res. 1999, 19, 4593–4597. [Google Scholar] [PubMed]
  65. Bakkar, A.A.; Wallerand, H.; Radvanyi, F.; Lahaye, J.B.; Pissard, S.; Lecerf, L.; Kouyoumdjian, J.C.; Abbou, C.C.; Pairon, J.C.; Jaurand, M.C.; et al. FGFR3 and TP53 gene mutations define two distinct pathways in urothelial cell carcinoma of the bladder. Cancer Res. 2003, 63, 8108–8112. [Google Scholar]
  66. Neuzillet, Y.; Paoletti, X.; Ouerhani, S.; Mongiat-Artus, P.; Soliman, H.; de The, H.; Sibony, M.; Denoux, Y.; Molinie, V.; Herault, A.; et al. A meta-analysis of the relationship between FGFR3 and TP53 mutations in bladder cancer. PLoS One 2012, 7, e48993. [Google Scholar]
  67. Miyata, Y.; Sakai, H. Thrombospondin-1 in urological cancer: Pathological role, clinical significance, and therapeutic prospects. Int. J. Mol. Sci. 2013, 14, 12249–12272. [Google Scholar] [CrossRef] [PubMed]
  68. Yeh, C.Y.; Shin, S.M.; Yeh, H.H.; Wu, T.J.; Shin, J.W.; Chang, T.Y.; Raghavaraju, G.; Lee, C.T.; Chiang, J.H.; Tseng, V.S.; et al. Transcriptional activation of the Axl and PDGFR-α by c-Met through a ras- and Src-independent mechanism in human bladder cancer. BMC Cancer 2011. [Google Scholar] [CrossRef]
  69. Wang, M.H.; Ronsin, C.; Gesnel, M.C.; Coupey, L.; Skeel, A.; Leonard, E.J.; Breathnach, R. Identification of the ron gene product as the receptor for the human macrophage stimulating protein. Science 1994, 266, 117–119. [Google Scholar] [CrossRef] [PubMed]
  70. Compérat, E.; Roupret, M.; Chartier-Kastler, E.; Bitker, M.O.; Richard, F.; Camparo, P.; Capron, F.; Cussenot, O. Prognostic value of MET, RON and histoprognostic factors for urothelial carcinoma in the upper urinary tract. J. Urol. 2008, 179, 868–872. [Google Scholar] [CrossRef] [PubMed]
  71. Chen, Q.; Seol, D.W.; Carr, B.; Zarnegar, R. Co-expression and regulation of Met and Ron proto-oncogenes in human hepatocellular carcinoma tissues and cell lines. Hepatology 1997, 26, 59–66. [Google Scholar] [PubMed]
  72. Maggiora, P.; Lorenzato, A.; Fracchioli, S.; Costa, B.; Castagnaro, M.; Arisio, R.; Katsaros, D.; Massobrio, M.; Comoglio, P.M.; Flavia di Renzo, M. The RON and MET oncogenes are co-expressed in human ovarian carcinomas and cooperate in activating invasiveness. Exp. Cell. Res. 2003, 288, 382–389. [Google Scholar] [CrossRef] [PubMed]
  73. Tamatani, T.; Hattori, K.; Iyer, A.; Tamatani, K.; Oyasu, R. Hepatocyte growth factor is an invasion/migration factor of rat urothelial carcinoma cells in vitro. Carcinogenesis 1999, 20, 957–962. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, P.; Nishitani, M.A.; Tanimoto, S.; Kishimoto, T.; Fukumori, T.; Takahashi, M.; Kanayama, H.O. Bladder cancer cell invasion is enhanced by cross-talk with fibroblasts through hepatocyte growth factor. Urology 2007, 69, 780–784. [Google Scholar] [CrossRef] [PubMed]
  75. Natali, P.G.; Prat, M.; Nicotra, M.R.; Bigotti, A.; Olivero, M.; Comoglio, P.M.; di Renzo, M.F. Overexpression of the met/HGF receptor in renal cell carcinomas. Int. J. Cancer 1996, 69, 212–217. [Google Scholar] [PubMed]
  76. Joseph, A.; Weiss, G.H.; Jin, L.; Fuchs, A.; Chowdhury, S.; O’Shaugnessy, P.; Goldberg, I.D.; Rosen, E.M. Expression of scatter factor in human bladder carcinoma. J. Natl. Cancer Inst. 1995, 87, 372–377. [Google Scholar] [CrossRef] [PubMed]
  77. Cheng, H.L.; Liu, H.S.; Lin, Y.J.; Chen, H.H.; Hsu, P.Y.; Chang, T.Y.; Ho, C.L.; Tzai, T.S.; Chow, N.H. Co-expression of RON and MET is a prognostic indicator for patients with transitional-cell carcinoma of the bladder. Br. J. Cancer 2005, 92, 1906–1914. [Google Scholar] [CrossRef] [PubMed]
  78. Sanchez-Carbayo, M.; Socci, N.D.; Lozano, J.J.; Haab, B.B.; Cordon-Cardo, C. Profiling bladder cancer using targeted antibody arrays. Am. J. Pathol. 2006, 168, 93–103. [Google Scholar] [CrossRef] [PubMed]
  79. Li, B.; Kanamaru, H.; Noriki, S.; Fukuda, M.; Okada, K. Differential expression of hepatocyte growth factor in papillary and nodular tumors of the bladder. Int. J. Urol. 1998, 5, 436–440. [Google Scholar] [CrossRef] [PubMed]
  80. Millis, S.Z.; Bryant, D.; Basu, G.; Bender, R.; Vranic, S.; Gatalica, Z.; Vogelzang, N.J. Molecular profiling of infiltrating urothelial carcinoma of bladder and nonbladder origin. Clin. Genitourin. Cancer 2014. [Google Scholar] [CrossRef]
  81. Arena, S.; Pisacane, A.; Mazzone, M.; Comoglio, P.M.; Bardelli, A. Genetic targeting of the kinase activity of the Met receptor in cancer cells. Proc. Natl. Acad. Sci. USA 2007, 104, 11412–11417. [Google Scholar] [CrossRef] [PubMed]
  82. Parr, C.; Jiang, W.G. Expression of hepatocyte growth factor/scatter factor, its activator, inhibitors and the c-Met receptor in human cancer cells. Int. J. Oncol. 2001, 19, 857–863. [Google Scholar] [PubMed]
  83. Koga, F.; Tsutsumi, S.; Neckers, L.M. Low dose geldanamycin inhibits hepatocyte growth factor and hypoxia-stimulated invasion of cancer cells. Cell. Cycle 2007, 6, 1393–1402. [Google Scholar] [CrossRef] [PubMed]
  84. Olaussen, K.A.; Commo, F.; Tailler, M.; Lacroix, L.; Vitale, I.; Raza, S.Q.; Richon, C.; Dessen, P.; Lazar, V.; Soria, J.C.; Kroemer, G. Synergistic proapoptotic effects of the two tyrosine kinase inhibitors pazopanib and lapatinib on multiple carcinoma cell lines. Oncogene 2009, 28, 4249–4260. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Miyata, Y.; Asai, A.; Mitsunari, K.; Matsuo, T.; Ohba, K.; Mochizuki, Y.; Sakai, H. Met in Urological Cancers. Cancers 2014, 6, 2387-2403. https://doi.org/10.3390/cancers6042387

AMA Style

Miyata Y, Asai A, Mitsunari K, Matsuo T, Ohba K, Mochizuki Y, Sakai H. Met in Urological Cancers. Cancers. 2014; 6(4):2387-2403. https://doi.org/10.3390/cancers6042387

Chicago/Turabian Style

Miyata, Yasuyoshi, Akihiro Asai, Kensuke Mitsunari, Tomohiro Matsuo, Kojiro Ohba, Yasushi Mochizuki, and Hideki Sakai. 2014. "Met in Urological Cancers" Cancers 6, no. 4: 2387-2403. https://doi.org/10.3390/cancers6042387

Article Metrics

Back to TopTop