Skip to main content
Erschienen in: Molecular Cancer 1/2018

Open Access 01.12.2018 | Review

Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities

Erschienen in: Molecular Cancer | Ausgabe 1/2018

Abstract

c-Met is a receptor tyrosine kinase belonging to the MET (MNNG HOS transforming gene) family, and is expressed on the surfaces of various cells. Hepatocyte growth factor (HGF) is the ligand for this receptor. The binding of HGF to c-Met initiates a series of intracellular signals that mediate embryogenesis and wound healing in normal cells. However, in cancer cells, aberrant HGF/c-Met axis activation, which is closely related to c-Met gene mutations, overexpression, and amplification, promotes tumor development and progression by stimulating the PI3K/AKT, Ras/MAPK, JAK/STAT, SRC, Wnt/β-catenin, and other signaling pathways. Thus, c-Met and its associated signaling pathways are clinically important therapeutic targets. In this review, we elaborate on the molecular structure of c-Met and HGF and the mechanism through which their interaction activates the PI3K/AKT, Ras/MAPK, and Wnt signaling pathways. We also summarize the connection between c-Met and RON and EGFR, which are also receptor tyrosine kinases. Finally, we introduce the current therapeutic drugs that target c-Met in primary tumors, and their use in clinical research.
Abkürzungen
HCC
Hepatocellular carcinoma
APC
Adenomatosis polyposis coli
CAF
Cancer associated fibroblast
CCC
Cholangiocarcinoma
CK1α
Casein kinase 1α
c-Met
Mesenchymal-epithelial transition factor
DSH/Dvl
Dishevelled protein
EGFR
Epidermal growth factor receptor
ErbB
Erb-b2 receptor tyrosine kinase 4
Gab1
Grb2-associated binder
GEFs
Guanine nucleotide exchange factors
GRB2
Growth factor receptor-bound protein 2
GSK3
Glycogen synthase kinase 3
HCC
Hepatocellular carcinoma
HDM2
Human double minute 2
HGF/SF
Hepatocyte growth factor/scatter factor
HIF-1
Hypoxia inducible factor-1
IPT domains
Immunoglobulin-like regions in plexins and transcription factors
JNK
Jun N-terminal kinase
LRP5/LRP6/Frizzled
Low-density lipoprotein receptor-related protein 5/6/Frizzled
MDS
Multisubstrate docking site
MET
MNNG HOS transforming gene
MIBC
Muscle invasive bladder cancer
MMC
Mitomycin C
MSP
Macrophage stimulating protein
MTOR
Mammalian target of rapamycin
NSCLC
Non-small cell lung carcinoma
PCa
Prostate cancer
PCAF
Acetyltransferase p300/CBP-associated factor
PFS
Progression free survival
PHA-66752
3-benzyloxy-2-amino
PI3K
Phosphatidylinositol-3-kinase
PIP2
Phosphatidylinositol-4, 5-diphosphate
PIP3
Phosphatidylinositol-3,4,5-triphosphate
PP2A
Protein phosphatase 2A
PSI domain
Plexin-semaphorin-integrin
PTEN
Phosphatase and tension homology deleted on chromosome 10
PTK
Protein tyrosine kinase
RCC
Renal cell carcinoma
RON
Receptor originated from nantes
SEMA domain
Sema homology region
TCF/LEF
T-cell factor/lymphoid enhancer factor
UM
Uveal melanoma
VEGF
Vascular endothelial growth factor

Background

c-Met (mesenchymal-epithelial transition factor), which belongs to the MET family, along with RON, is a type of receptor tyrosine kinase that is expressed on the surfaces of various epithelial cells; its ligand is HGF/SF(ligand hepatocyte growth factor/scatter factor) [1, 2]. HGF belongs to the soluble cytokine family and is also a member of the plasminogen-related growth factor family. It is synthesized by mesenchymal cells, fibroblasts, and smooth muscle cells, and acts through a paracrine mechanism to activate HGF/c-Met signaling to exert its biological functions [3]. Under normal conditions, HGF/c-Met can mediate embryogenesis, tissue regeneration, wound healing, and the formation of nerve and muscle, which is controlled by the tumor suppressor p53. Thus, this axis plays an important role in normal biological functions in humans [46].
However, as a type of proto-oncogene, abnormal activation of c-Met can promote the development and progression of multiple cancers such as liver, lung, colon, breast, pancreatic, ovarian, prostate, and gastric carcinomas, in addition to cancers of the nervous system such as glioblastoma [79]. The HGF/c-Met axis, which can interact and cooperate with other types of tyrosine kinases, can stimulate various downstream signaling pathways in tumor cells, such as PI3K/AKT, JAK/STAT, Ras/MAPK, SRC, and Wnt/β-catenin, among others [1013]. These aforementioned phenomena regulate multiple biological processes such as tumor proliferation, invasion, metastasis, anti-apoptosis, EMT, and angiogenesis [1417]. It has been determined that c-Met gene mutations, overexpression, and amplification also occur in a variety of human tumor types, and these events are closely related to the aberrant activation of the HGF/c-Met signaling pathway [18, 19]. Meanwhile, high c-Met expression is closely associated with poor prognosis in cancer patients. Studies have shown that abnormal activation of c-Met is critical for resistance to targeted therapies such as tyrosine kinase inhibitors and drugs that act against associated signaling pathways. Therefore, as abnormal c-Met function can increase the difficulty associated with tumor treatment, understanding its role in cancer is extremely important [4, 20].

Structures of c-met and HGF

The MET (c-Met encoding) gene is located on human chromosome 7 (7q21-q31), includes 21 exons and 20 introns, and encodes a protein that is approximately 120 kDa in size [21]. The translated product is processed to form a heterodimer that is linked by the extracellular α chain and the transmembrane β chain. The transmembrane chain consists of a SEMA domain (sema homology region; SEMA), a PSI domain (plexin-semaphorin-integrin; PSI), four IPT domains (immunoglobulin-like regions in plexins and transcription factors), a transmembrane domain, a juxtamembrane domain, a tyrosine kinase domain (TK domain), and a c-terminal docking site (carboxyl terminal; CT). SEMA is the site where HGF binds directly to c-Met, and PSI can stabilize this interaction. Ser-975 and Tyr-1003 sites at the juxtamembrane domain play an important role in the negative regulation of c-Met [14, 22, 23]. When HGF binds c-Met, Tyr-1234 and Tyr-1235 in the intracellular tyrosine kinase domain undergo autophosphorylation, which results in autophosphorylation of Tyr-1349 and Tyr-1356 in the C-terminal docking site. This facilitates the recruitment of intracellular effector molecules such as growth factor receptor-bound protein 2(GRB2), SRC, PI3K, and GAB1, and consequently the activation of downstream signaling pathways (Fig. 1) [24, 25].
The HGF gene encoding a 728-amino-acid protein is located on human chromosome 7 and consists of 18 exons and 17 introns [21]. Mature HGF is a heterodimer consisting of an α chain (69 kDa) and a β chain (34 kDa), which are linked by a disulfide bond. This protein consists of six domains. An N-terminal hairpin domain and four Kringle domains comprise the α chain, and the hairpin domain and first two Kringle domains are necessary for HGF to exert its biological function. The β chain forms a serine protease analog domain lacking catalytic activity, and this is the binding site for c-Met.

HGF/c-met cascades in carcinoma

The binding of HGF to c-Met can initiate several downstream signaling pathways; we selected three significant pathways, based on their functions in carcinoma for futher review.

HGF/c-met and the Ras pathway

The binding of c-Met by its selective ligand HGF can induce structural changes in c-Met [26]; specifically, its intracellular protein tyrosine kinase (PTK) domain becomes activated, resulting in exposure of the multisubstrate docking site (MDS). Grb2 is then recruited to this site [27]. After autophosphorylation of the PTK domain, it can bind the SH2/SH3 domain of Grb2 [28], which subsequently recruits downstream guanine nucleotide exchange factors (GEFs) such as SOS. Downstream SOS can recruit Ras-GTP from the cell matrix to the membrane and convert it to activated Ras-GTP. Ras successively activates Raf, MEK, MAPKs, ERK, JNK (Jun N-terminal kinase), and p38 (HOG), among others, and the activated MAPKs then enter the cell nuclei to activate transcription factors (e.g. Elk1, Etsl, c-Myc) through phosphorylation. This, in turn, can interfere with the cell cycle and induce cell transformation, consequently promoting carcinogenesis. MAPKs also induce the degradation of proteins and matrix, promote cell migration, and sustain tumor proliferation (Fig. 2) [29, 30].
In tumor cells, the mutation rate of the Ras gene is approximately 25%, whereas in pancreatic cancer and colon cancer, the mutation rates could be 85 and 40%, respectively. Such mutations are predominantly point mutations and gene amplifications [27]. Mutations occur in codons 11, 12, 13,18, 59, and 69, which affect the interaction between Ras and GAP. Upon mutation, its intrinsic GTPase activity is inhibited, which can lead to malignant cell transformation through sustained activation of Ras2GTP (Fig. 2).

HGF/c-met and PI3K pathway

When HGF binds c-Met and induces autophosphorylation, the phosphorylated residue acts as a docking site for the heterodimeric PI3K-p85 subunit. Here, the p85 subunit of phosphatidylinositol-3-kinase (PI3K) binds to the adaptor protein at the SH2/SH3 domain, using the same phosphorylated site. When PI3K recruits enough activated receptors, it initiates the phosphorylation of many phosphatidylinositol intermediates. Especially, in many tumor-associated signaling cascades, PI3K can convert phosphatidylinositol-4, 5-diphosphate (PIP2) to phosphatidylinositol-3,4,5-trisphosphate (PIP3). Phosphorylated RTKs can bind the SH2 domain of p85, and subsequently recruit the p85-p110 complex to cell membranes to activate the complex. Activated PI3K accelerates the conversion of PIP2 to PIP3. The association between PIP3 and signaling proteins containing a PH domain, namely, AKT and PDK1, facilitates the phosphorylation of AKT at Thr-308 and at Ser-473 by PDK1 [27]. Activated AKT, which later translocates to cell nuclei, modulates downstream transcription factors like FKHRL1, NF-κB, and Bcl-2, and inhibits the expression of tumor suppressor genes. AKT also phosphorylates GSK-3 and mammalian target of rapamycin (mTOR) or a series of inhibitory proteins such as p21CIP1 and p27KIP1; these, in turn, separately upregulate the expression of Cyclin D, shorten the cell cycle, and ultimately contribute to tumorigenesis [31]. In addition to this, RTKs might also activate the PI3K/AKT pathway through Ras (Fig. 2).
One study found that mTOR can regulate the degradation of extracellular matrix in cancer cells and influence the synthesis and secretion of matrix metalloproteinase; through this mechanism, this protein can also promote the invasion and metastasis of tumor cells [32]. Activated AKT might also phosphorylate nitric oxide synthase to produce NO, which positively regulates angiogenesis (Fig. 2).
The PI3K/AKT/mTOR pathway can modulate the expression of vascular endothelial growth factor (VEGF) and hypoxia inducible factor-1 (HIF-1) through the activation of human double minute 2 (HDM2) (Fig. 2) [33].
In addition, PTEN (phosphatase and tension homology deleted on chromosome 10) negatively regulates phosphorylation in the PI3K pathway. Specifically, this protein facilitates the dephosphorylation of PIP3, converting PIP3 to PIP2. Hence, it relieves the negative regulation of the downstream PI3K components AKT and mTOR. In tumor cells, mutations or deletions in PTEN are common, and enable the increased activation of the PI3K/AKT/mTOR pathway; this leads to aberrant activation of this pathway (Fig. 2).

Association between the HGF/c-met and Wnt/β-catenin signaling pathways

HGF/c-Met is closely related to Wnt/β-catenin signaling, and promotes tumor proliferation, invasion, and metastasis by modulating this signaling pathway [34]. Studies have shown that in colon cancer and glioblastoma, c-Met expression can enhance Wnt/β-catenin signal transduction, and prevent GSK3β from phosphorylating β-catenin; this, in turn, promotes the translocation of β-catenin to the nucleus, facilitating tumorigenesis. Accordingly, it has been shown that c-Met inhibitors can inhibit Wnt pathway activity in tumor cells [35, 36]. Meanwhile, it has been found that in breast cancer cells undergoing osteolytic bone metastasis, the activation of HGF/c-Met signaling can promote β-catenin translocation to the nucleus and enhance its transcriptional activity. Therefore, HGF/c-Met can exert its biological function through the Wnt signaling pathway (Fig. 2) [37].
In normal cells lacking Wnt pathway activation, β-catenin is cytoplasmic and is phosphorylated at Ser-31, Ser-37, Thr-4, and Ser-45 by GSK3β and CK1 proteins, which are part of the destruction complex. At the same time, it can be acetylated by acetyltransferase p300/CBP-associated factor (PCAF) at Lys-49. Subsequently, these modified sites are recognized by and associate with the β-TrCP E3 ubiquitin ligase, resulting in its degradation by the proteasome, thereby preventing translocation to the nucleus [38, 39]. However, in tumor cells, aberrant activation of the HGF/c-Met pathway and stimulation of Wnt pathway block phosphorylation and acetylation of β-catenin through different signals. This results in the accumulation of β-catenin in the cytoplasm; it then enters the nucleus to displace Groucho, which has a transcriptional inhibitory effect on T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors. β-catenin exerts its functions along with BCL9/LGS and Pygo to promote expression of Myc, Cyclin D1, and MMP-7, which facilitates proliferation, invasion, and metastasis (Fig. 2) [38, 40, 41].

Crosstalk between c-met and other receptors tyrosine kinases

C-met and RON

Studies have shown that c-Met and RON (receptor originated from nantes) are overexpressed [42] or aberrantly activated in many epithelial-derived malignant cancers [4349]. These proteins can be involved in tumorigenesis by promoting cell proliferation, inhibiting apoptosis, enhancing angiogenesis, and promoting metastasis, among other functions, by acting upstream of these processes [4649]. c-Met and RON can be activated by HGF and macrophage stimulating protein (MSP), respectively. Activated signaling depends on the tissue availability of adaptor proteins and signaling intermediates or the tendency of the adaptor proteins and signaling intermediates to undergo homodimerization or heterodimerization [50, 51]. MSP and HGF are highly homologous in sequence and structure [52], and are secreted as inactive single chains by multiple tissues and cells including smooth muscle, fibroblasts, adipose tissue, epithelial-derived tumors, liver, lungs, adrenal glands, placenta, and kidney. They are subsequently activated by proteasomal cleavage and form dimeric peptides consisting of α and β chains. In contrast to HGF, the high-affinity RON-binding site (for MSP) is located in the β chain [51].
The dimerization of these two monomers represents a major regulatory mechanism for the activation of tyrosine kinase receptors [53]. In some cases, the formation of a heterodimeric complex permits interaction and crosstalk between different receptors of the same subfamily. The epidermal growth factor receptor (EGFR) family is the best example of a tyrosine kinase receptor that undergoes homo and heterodimerization [54, 55]. Therefore, it is important to study the dimerization mechanism of PTKs. RON and c-Met are co-expressed in many types of tumors and crosstalk between c-Met and RON has been demonstrated [52]. Analysis of their structural homology suggested that they might interact, and in fact, studies have indicated that c-Met and RON can form heterodimers and phosphorylate each other [56]. One study showed that oncogenic addiction to c-Met requires co-expression of RON in four different tumor cell lines [50]. In these cases, RON was constitutively activated, and this was dependent on transphosphorylation by c-Met [50]. Experimentally, it has been shown that c-Met has stronger kinase activity than RON [57], and thus it is possible that heterodimers might be more efficiently activated than RON-RON homodimers. The fact that oncogenic addiction to c-Met requires RON implies that c-Met-RON heterodimers can promote the activation of diverse signaling cascades through different platforms. However, c-Met and RON possess remarkably similar tyrosine-binding sites that serve as docking sites for signaling molecules, and thus these signaling platforms might also be redundant. However, one study found that these two receptors have different kinase activities. Specifically, c-Met can be activated directly through Grb2 binding, but requires modulation for activation by other platforms [58]; in contrast, RON relies mainly on Grb2-associated binder (Gab1), based on the fact that the binding of Gab2 by RON attenuates the recruitment of Gab1 and represses signal transduction.
Grb2 has a unique role with respect to c-MET-RON heterodimers. Although Grb2 inhibits RON autophosphorylation, it enhances this process with c-MET [59]. Considering heterodimers of the EGFR family, the signaling diversity through heterodimers could depend on the relative abundance of each receptor [54].
RON expression might partially modulate c-Met activity, which can be applied when modeling this receptor. With respect to this, we found that knockdown of RON enhances the level and duration of HGF-mediated activation of MAPK and AKT [53]. Although the functional relevance of c-Met-RON heterodimers has not been fully explored, some studies suggest that general knockdown of RON leads to changes in c-Met signaling. For example, it was found that silencing RON in pancreatic cancer cell lines leads to upregulation of c-Met expression and activity [56]. This suggests that inhibitors that co-target or simultaneously block the kinase activities of both c-Met and RON might be clinically useful. However, most studies have not considered the possibility that separately inhibiting either c-Met or RON might lead to compensation by [60] the other.

C-met and EGFR

It has been confirmed that signal transduction between the c-Met and EGFR pathways is closely linked in breast cancer, lung cancer, brain cancer, and other tumors; however, the associated mechanism is still not fully understood [6164]. Studies have shown that 70% of EGFR-activating mutations in non-small cell lung carcinoma (NSCLC) are associated with an initial positive response to the EGFR inhibitors gefitinib or erlotinib [65]. However, the vast majority of tumors that respond to EGFR inhibitors achieve acquired resistance [66]. Interestingly, the expression and activation of c-Met are associated with initial resistance and acquired resistance to EGFR inhibitors in patients with NSCLC [6668]. Initial resistance might occur through the simultaneous activation of c-Met and EGFR pathways in lung cancer, whereas inhibiting both maximizes the inhibitory effect on the tumor [61]. As such, studies have shown that c-Met might be an effective therapeutic target for overcoming EGFR inhibitor resistance in lung cancer [62].
Possible explanations regarding this mechanism are as follows. One study has already shown that the second mutation in EGFR, T790 M, and the amplification of the MET proto-oncogene will lead to the activation of its downstream ERBB3-initiated PI3K/AKT pathway, resulting in EGFR-TKI acquired resistance [67, 69, 70]. When the c-MET gene is amplified, the two downstream pathways (Grb2/MAPK and PI3K/AKT) are activated by the increase in the number of ERBB3 receptors [69, 70].
In addition, continuous interaction with HGF facilitates c-Met amplification-mediated reversible resistance to EGFR-TKI treatment [66, 70]. When HGF activates Met, it activates MAPK and PI3K/AKT signaling pathways through Gab1, leading to the occurrence of irreversible EGFR-TKI resistance [66].
If EGFR and Met mutations exist simultaneously, drug resistance will be further exacerbated [70]. Therefore, we speculate that c-Met activation of downstream PI3K/AKT and MAPK pathways bypasses EGFR activation because they can both act as tyrosine kinase receptors and activate this pathway (Fig. 3). In addition, c-Met can either directly or indirectly transactivate the PI3K pathway; the fact that c-Met is not activated by this RTK also supports this hypothesis [71].
Another study found that EGFR mutation and Met activation were observed in tumor cells. At the same time, whereas the activation of c-Met was not the result of gene mutation, it resulted in poor prognosis for NSCLC metastasis [68]. In addition, after reversible resistance to EGFR-TKIs in lung cancer cells, HGF can induce an irreversible second mutation (Fig. 3) [66].
HGF/c-Met is activated in approximately 50% of hepatocellular carcinomas (HCC), and expression levels of these proteins are associated with poor clinical prognosis for this disease [7275]. Cells with constitutive c-Met activity respond to c-Met inhibition [76]; however, one study found that monotherapy does not completely eliminate tumor growth, suggesting that tumor survival mechanisms that bypass the inhibition of this pathway might be involved in the maintenance of tumor growth in response to these treatments [77].
In previous studies, inhibition of the EGFR pathway was shown to lead to either activation or inhibition of the c-Met pathway, whereas another study showed that c-Met inhibition leads to the activation of the EGFR pathway in a c-Met-positive HCC model [76]. In addition, EGFR inhibitor monotherapies are not significantly effective with respect to in vitro cell viability [76]. c-Met inhibitor monotherapy triggers several survival mechanisms that bypass cell death induced by these agents, including increased expression of the EGFR ligand TGF-α and ErbB3. It has been determined that members of the EGFR family can form homodimers or heterodimers and that different dimers have different signal transduction capabilities; specifically, ErbB3 can heterodimerize with ErbB1 to form one of the most potent dimers [78]. Experiments have shown that c-Met inhibition enhances EGFR signaling by increasing ErbB3 expression [76]. In addition, the increase in TGF-α expression that results from c-Met inhibition, whether this occurs through an autocrine or paracrine mechanism, and its effect on HCC cell survival requires further study.

Current clinical trials targeting c-met

Currently, drugs targeting c-Met that are in clinical trials can be classified as monoclonal antibodies (e.g. onartuzumab) and small molecule inhibitors. Small molecule inhibitors bind to the tyrosine kinase domain of c-Met and can be further classified into ATP competitive inhibitors (e.g. crizotinib) and non-ATP competitive inhibitors (e.g. tivantinib). Inhibitors belonging to the same group inhibit c-Met downstream signaling in a similar manner. Therefore, in this review, we will introduce current clinical trials targeting c-Met, and have chosen onartuzumab, crizotinib, and tivantinib as examples to elaborate on their c-Met binding sites, as shown in Fig. 1.

Anti-c-met monoclonal antibody Metmab (onartuzumab)

Onartuzumab is a humanized single-armed specific monoclonal antibody targeting c-Met. The binding of onartuzumab to c-Met is highly specific and this antibody can block c-Met-HGF binding specifically by blocking the HGF α-chain and by forming a complex with the Sema-PSI domain of c-Met [79]; this process occurs without exerting an agonistic activity or triggering c-Met dimerization.
Onartuzumab has been applied as a c-Met inhibitor for the treatment of NSCLC and breast cancer in clinical trials (Table 1) [80], and it proved to be considerably effective. Other studies also found that onartuzumab in combination with erlotinib and placebo is effective for NSCLC. Therefore, this drug might have potential to treat c-Met-overexpressing cancer.
Table 1
Ongoing studies with Metmb (onartuzumab)
NCT Number
Combination Drugs
PHASES
URL
NCT01897038
Drug: Onartuzumab
Phase1
Drug: Sorafenib
NCT01519804
Drug: Placebo
Phase2
Drug: cisplatin/carboplatin
Drug: onartuzumab
Drug: paclitaxel
NCT00854308
Drug: Erlotinib HCl
Phase2
Drug: MetMAb
Drug: placebo (0.9% saline)
NCT01496742
Drug: Placebo
Phase2
Drug: RO5490258
Drug: bevacizumab [Avastin]
Drug: cisplatin/carboplatin
Drug: paclitaxel
Drug: pemetrexed
NCT02031744
Drug: erlotinib [Tarceva]
Phase3
Drug: Placebo
Drug: Onartuzumab [MetMAb]
NCT01887886
Drug: erlotinib
Phase3
Drug: onartuzumab
Drug: placebo
NCT01456325
Drug: Erlotinib
Phase 3
Drug: Onartuzumab (MetMab)
Drug: Placebo

Small molecule inhibitors

Crizotinib

Crizotinib (PF-02341066, trade name: Xalkori), an effective small molecule inhibitor of c-Met, was derived from the first-generation series c-Met inhibitor, PHA-66752 (3-benzyloxy-2-amino). PF-22341066 targets the TK domain of c-Met, and after a series of reactions, some residues cause a conformational change, which interferes with the ATP binding site. One clinical trial (phase I) for the treatment of NSCLC with enhanced Met amplification, performed in 2014 [81], showed that crizotinib has increased potential for the treatment of c-Met-associated cancer. Crizotinib is one of five drugs approved by the FDA for the treatment of advanced NSCLC, to date [82], and it is used for the clinical treatment of ROS1-positive lung cancer [83]. Moreover, the number of studies focusing on the combination of crizotinib and other drugs is increasing. Huang et al. [84] showed that crizotinib with cisplatin induces G2/M cell cycle arrest and apoptosis in ovarian cancer cells. Stanley et al. [85] elaborated on the different growth inhibitory effects caused by the combination of c-Met inhibitors with cytotoxic drugs using breast cancer cell lines (BT474, MCF7, MDA-MB-468, and SKBr3). Results suggested that crizotinib and EGFR-TKIs might have a synergistic effect on MCF7 and MDA-MB-468 cells and an antagonistic effect on BT474 and SKBr3 cells. The combination of EGFR-TKIs and crizotinib was shown to have a more pronounced effect than a single drug regimen on breast cancer. In addition, sensitivity to mitomycin C (MMC), when combined with crizotinib, was studied using a colorectal cancer cell line. The results also showed that a combination of the two drugs resulted in increased tumor cell apoptosis and a synergistic effect. Currently, several clinical trials are in progress. A summary of these trials is provided in Table 2.
Table 2
Ongoing studies with crizotinib
NCT Number
Combination Drugs
PHASES
URL
NCT00932451
Drug: PF-02341066
Phase2
NCT00932893
Drug: PF-02341066
Phase3
Drug: Pemetrexed
Drug: Docetaxel
NCT01154140
Drug: treatment
Phase3
Drug: treatment
NCT01685060
Drug: LDK378
Phase2
NCT01121575
Drug: PF-02341066
Phase1
Drug: PF-00299804
Drug: PF-02341066
Drug: PF-00299804
NCT00965731
Drug: Erlotinib
Phase1
Drug: PF-02341066
NCT02435108
Drug: crizotinib
Phase2

Cabozantinib

Cabozantinib (XL184) is a small molecule inhibitor of Met and AXL [86, 87], and has been approved by the FDA for the treatment in progressive metastatic thyroid medullary carcinoma [60, 88], and also for advanced renal cell carcinoma after the implementation of antiangiogenic therapy regimens [89]. Wakelee divided NSCLC patients into three groups as follows: the first two groups were administered erlotinib alone (150 mg poqd) and cabozantinib alone (60 mg poqd), whereas the third group was administered combination therapy (150 mg erlotinib/40 mg cabozantinib). Results showed that progression-free survival and overall survival were significantly improved with cabozantinib treatment. Shotani et al. [90] showed that cabozantinib is effective in inhibiting growth and invasion in BCa cell lines driven by HGF (5637 and T24), and blocked HGF-Met signaling to inhibit MMP1 expression. Thus, cabozantinib has potential for the treatment of muscle invasive bladder cancer (MIBC). At present, this drug is at the clinical stage for prostate cancer treatment, and has been tested in phase II trials.

Foretinib

Foretinib (GSK1363089) is an ATP-competitive c-Met inhibitor, and its therapeutic potential has been assessed for different tumors including head and neck cancer, gastric cancer, and liver cancer [56, 57, 91, 92]. Chia et al. [93] conducted a phase I study to determine the effect of combining foretinib with lapatinib on HER-2-positive metastatic breast cancer. The study suggested that the combined use of foretinib and lapatinib at doses of 45 mg and 1000 mg PO, respectively, could be tolerated relatively well. The most common grade 3 and higher toxic adverse reactions were mainly high blood pressure, diarrhea, nausea, and fatigue. Yin et al. [22] also demonstrated that foretinib inhibits prostate cancer (PCa) metastasis by targeting c-Met.

LY280163

LY280163 is an ATP competitive Met tyrosine kinase inhibitor developed by Lilly. Cheng et al. [94] showed that this drug can improve the response of MEK inhibitors such as trametinib in metastatic uveal melanoma (UM) patients and promote the expression of PARP. In addition, studies [95] have investigated the effect of LY2801653 on human cholangiocarcinoma (CCC) cell lines. Using a xenograft mouse model, it was determined that LY2801653 blocks c-Met phosphorylation, down-regulates downstream target expression, and inhibits CCC cell proliferation and xenograft tumor growth.

MK2461

MK2461 is an ATP competitive small molecule multi-target inhibitor developed by Merck Sharp & Dohme Corp. It is effective in inhibiting the proto-oncogene c-Met mutants N1100Y, Y1230C, Y1230H, Y1235D, and M1250Tn1100y. Currently, this drug is in experimental stage I clinical trials for advanced cancer.

Capmatinib

Capmatinib (INC280) blocks c-Met phosphorylation and the activation of key downstream molecules in c-Met-dependent tumor cell lines, causing mitochondrial membrane depolarization and DNA repair [96, 97]. The drug has been utilized in phase I trials for advanced solid cancer. Wei et al. [96] found that the addition of capmatinib could effectively block cell proliferation induced by cancer associated fibroblast (CAF) matrix with overexpression of HGF, and could eliminate CAF-induced ovarian cancer cell resistance. The latest study by Lara et al. [97] utilized a series of NSCLC cell lines (including three EGFR-mutant cell lines, HCC827, PC9 and H1975, one Kirsten rat sarcoma virus oncogene mutant cell line, H358, and one EGFR and KRAS wild type cell line, H1666) to determine whether capmatinib in combination with erlotinib could attenuate erlotinib resistance. The Massachusetts General Hospital in the United States has also launched a clinical trial for the use of capmatinib in stage IV patients with malignant NSCLC. In addition, Novartis Pharmaceuticals is performing clinical phase II trials using oral capmatinib combined with gefitinib for NSCLC patients with c-Met amplification.

Tivantinib

Tivantinib (ARQ197), developed by American ArQule Corporation and Japan’s Daiichi Sankyo and Kyowa Hakko Kogyo, is a non-ATP competitive inhibitor that blocks receptor activation and downstream signaling by binding to unactivated receptors [49, 98]. ARQ197 directly binds the A-loop and P-loop phenylalanines by inducing “hydrophobic collapse”, resulting in disruption of the ionic interaction in the catalytic residue with the help of Arg1227, Tyr1230, and other residues. In recent years, its pharmacokinetic mechanism has become increasingly controversial. It was previously believed that tivantinib can exert its biological effects by directly inhibiting c-Met receptor tyrosine kinases. However, several subsequent studies showed that the biological effect of tivantinib does not depend on the c-met receptor; in contrast, it inhibits tumor cells through microtubule depolymerization. Tivantinib inhibits tubulin polymerization, disrupting tubulin metabolism, prolonging cell G2/M phase, and promoting apoptosis [99, 100]. Studies have shown that combining tivantinib with erlotinib for treatment improves progression free survival (PFS) and is well tolerated [101103]. In patients with advanced solid tumors, tivantinib combined with sorafenib treatment was shown to be safe, especially for renal cell carcinoma (RCC), hepatocellular carcinoma (HCC), and melanoma patients harboring tumors with high levels of c-Met; an enhanced therapeutic effect was also observed for these cases. The combination of the two drugs was shown to enhance the antitumor activity of sorafenib, thus reducing associated resistance without promoting off-target effects [104]. As is described in Table 3, we summarized ongoing studies involving tivantinib.
Table 3
Ongoing studies with tivantinib
NCT Number
Combination Drugs
PHASES
URL
NCT01755767
Drug:Tivantinib
Phase 3
Drug: Placebo
NCT00988741
Drug: ARQ 197
Phase 2
Drug: Placebo
NCT01656265
Drug: ARQ 197
Phase 1
NCT00802555
Drug: ARQ 197
Phase 1
NCT00557609
Drug: ARQ 197
Phase 2
NCT01575522
Other: Laboratory Biomarker Analysis
Phase 2
Drug: Tivantinib
NCT01395758
Drug: ARQ 197 plus erlotinib
Phase 2
Drug: Pemetrexed, docetaxel or gemcitabine
NCT01244191
Drug: Tivantinib
Phase 3
Drug: Placebo
Drug: Erlotinib
NCT01069757
Drug: ARQ 197 and Erlotinib
Phase 1
NCT01251796
Drug: ARQ 197 and Erlotinib
Phase 1
NCT00777309
Drug: ARQ 197
Phase 2
Drug: Erlotinib
Drug: Placebo

Conclusions

Despite research on c-Met over the past 30 years, the structure and function of this tyrosine kinase has not been well established. HGF/c-Met mediates cascades that play a key role in tumorigenesis; extensive research on those pathways is not only beneficial for enhancing our understanding of the mechanisms associated with carcinoma, but also suggest promising targets for the development of novel cancer treatments. Considering the complexity of the HGF/c-Met axis, further exploration of the mechanism through which blocking c-Met activation modulates downstream pathways is required.
Recently, many clinical trials have found that drug resistance is more easily acquired with single drug therapy; therefore, research on combining c-Met inhibitors with other drugs (e.g. EGFR-TKI) will lead to the rapid discovery of effective treatment options. One study showed that in a nude mouse model of treatment-sensitive NSCLC, erlotinib resistance could be effectively reversed by the administration of SU1274. Meanwhile, Klempner et al. [105] found that cabozantinib could reverse resistance to crizotinib. Thus, the use of combinations of drugs to avoid resistance induced by the utilization of a single drug might become a major priority for researchers developing novel c-Met inhibitors.
Recently, phase II/III clinical trials for c-Met inhibitors have been initiated, and many of these drugs are regarded as second-line drugs. The main problem is that in most c-Met-overexpressing cancer cells, this receptor is not always the only driver of carcinoma, as it often interacts with other tyrosine kinase receptors. For example, the cytotoxic effect of tivantinib is not due solely to c-Met inhibition. Meanwhile, non-specific reactions are also a major problem when using c-Met inhibitors. For example, the c-Met monoclonal antibody Metmab is associated with several adverse events including rash, diarrhea, fatigue, and nausea/vomiting.
Under these circumstances, more precise information regarding how the drug functions and its relationship with c-Met and other tyrosine kinase receptors is required. In fact, as mentioned previously, the relationship between c-Met and its family member RON is being extensively studied, whereas the mechanism underlying the crosstalk between c-Met and RON is still not fully understood. One study showed that in pancreatic cancer, silencing RON might modulate the c-Met signaling pathway, resulting in a compensatory reaction during the downregulation of either tyrosine kinase receptor [5659]. As such, we might consider targeting c-Met and RON simultaneously. It turns out that c-Met and RON also interact with other tyrosine kinase receptors. Nevertheless, compared to RON, these other receptors might not be as indispensable for the activation of c-Met, and the significance of this crosstalk is still not well understood.
Meanwhile, the initiation of carcinoma can be induced by multiple factors including genome backgrounds, environmental factors [106], microenvironment [29, 107], even the non-coding RNAs [108110]. Considering that, further efforts on the interactions between c-Met and other cancer related risk factors will be necessary in promoting the process of precise medical treatment on c-Met which thus, demand a further comprehensive understanding of this tyrosine kinase receptor.
However, based on the fact that c-Met has an intimate association with cancer, targeting this receptor for the treatment of tumorigenesis is still thought to be associated with vast clinical significance.

Acknowledgements

Not applicable.

Funding

This work was supported in part by grants from the National Natural Science Foundation of China (81472531, 81572787, 81672683, 81672993, 81772928 and 81702907), the Natural Science Foundation of Hunan Province (2015JJ1022, 2016JC2035 and 2017SK2105) and the Overseas Expertise Introduction Project for Discipline Innovation (111 Project, 111-2-12).

Availability of data and materials

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Salgia R. MET in lung cancer: biomarker selection based on scientific rationale. Mol Cancer Ther. 2017;16:555–65.PubMedCrossRef Salgia R. MET in lung cancer: biomarker selection based on scientific rationale. Mol Cancer Ther. 2017;16:555–65.PubMedCrossRef
2.
Zurück zum Zitat Fu YT, Zheng HB, Zhou L, Zhang DQ, Liu XL, Sun H. Valproic acid, targets papillary thyroid cancer through inhibition of c-Met signalling pathway. Am J Transl Res. 2017;9:3138–47.PubMedPubMedCentral Fu YT, Zheng HB, Zhou L, Zhang DQ, Liu XL, Sun H. Valproic acid, targets papillary thyroid cancer through inhibition of c-Met signalling pathway. Am J Transl Res. 2017;9:3138–47.PubMedPubMedCentral
3.
Zurück zum Zitat Rucki AA, Xiao Q, Muth S, Chen J, Che X, Kleponis J, Sharma R, Anders RA, Jaffee EM, Zheng L. Dual Inhibition of Hedgehog and c-Met Pathways for Pancreatic Cancer Treatment. Mol Cancer Ther. 2017;16:2399-409. Rucki AA, Xiao Q, Muth S, Chen J, Che X, Kleponis J, Sharma R, Anders RA, Jaffee EM, Zheng L. Dual Inhibition of Hedgehog and c-Met Pathways for Pancreatic Cancer Treatment. Mol Cancer Ther. 2017;16:2399-409.
4.
Zurück zum Zitat Zhu L, Xiong X, Kim Y, Okada N, Lu F, Zhang H, Sun H. Acid sphingomyelinase is required for cell surface presentation of met receptor tyrosine kinase in cancer cells. J Cell Sci. 2016;129:4238–51.PubMedPubMedCentralCrossRef Zhu L, Xiong X, Kim Y, Okada N, Lu F, Zhang H, Sun H. Acid sphingomyelinase is required for cell surface presentation of met receptor tyrosine kinase in cancer cells. J Cell Sci. 2016;129:4238–51.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat 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. 2016;15:886–94.PubMedPubMedCentralCrossRef 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. 2016;15:886–94.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Szturz P, Raymond E, Abitbol C, Albert S, de Gramont A, Faivre S. Understanding c-MET signalling in squamous cell carcinoma of the head & neck. Crit Rev Oncol Hematol. 2017;111:39–51.PubMedCrossRef Szturz P, Raymond E, Abitbol C, Albert S, de Gramont A, Faivre S. Understanding c-MET signalling in squamous cell carcinoma of the head & neck. Crit Rev Oncol Hematol. 2017;111:39–51.PubMedCrossRef
8.
Zurück zum Zitat Kim B, Jung N, Lee S, Sohng JK, Jung HJ. Apigenin inhibits cancer stem cell-like phenotypes in human Glioblastoma cells via suppression of c-Met Signaling. Phytother Res. 2016;30:1833–40.PubMedCrossRef Kim B, Jung N, Lee S, Sohng JK, Jung HJ. Apigenin inhibits cancer stem cell-like phenotypes in human Glioblastoma cells via suppression of c-Met Signaling. Phytother Res. 2016;30:1833–40.PubMedCrossRef
9.
Zurück zum Zitat International Cancer Genome Consortium PedBrain Tumor P. Recurrent MET fusion genes represent a drug target in pediatric glioblastoma. Nat Med. 2016;22:1314–20.CrossRef International Cancer Genome Consortium PedBrain Tumor P. Recurrent MET fusion genes represent a drug target in pediatric glioblastoma. Nat Med. 2016;22:1314–20.CrossRef
10.
Zurück zum Zitat Imura Y, Nakai T, Yamada S, Outani H, Takenaka S, Hamada K, Araki N, Itoh K, Yoshikawa H, Naka N. Functional and therapeutic relevance of hepatocyte growth factor/c-MET signaling in synovial sarcoma. Cancer Sci. 2016;107:1867–76.PubMedPubMedCentralCrossRef Imura Y, Nakai T, Yamada S, Outani H, Takenaka S, Hamada K, Araki N, Itoh K, Yoshikawa H, Naka N. Functional and therapeutic relevance of hepatocyte growth factor/c-MET signaling in synovial sarcoma. Cancer Sci. 2016;107:1867–76.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Liang Y, Liu J, Liu T, Yang X. Anti-c-Met antibody bioconjugated with hollow gold nanospheres as a novel nanomaterial for targeted radiation ablation of human cervical cancer cell. Oncol Lett. 2017;14:2254–60.PubMedPubMedCentralCrossRef Liang Y, Liu J, Liu T, Yang X. Anti-c-Met antibody bioconjugated with hollow gold nanospheres as a novel nanomaterial for targeted radiation ablation of human cervical cancer cell. Oncol Lett. 2017;14:2254–60.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Pilotto S, Carbognin L, Karachaliou N, Ma PC, Rosell R, Tortora G, Bria E. Tracking MET de-addiction in lung cancer: a road towards the oncogenic target. Cancer Treat Rev. 2017;60:1–11.PubMedCrossRef Pilotto S, Carbognin L, Karachaliou N, Ma PC, Rosell R, Tortora G, Bria E. Tracking MET de-addiction in lung cancer: a road towards the oncogenic target. Cancer Treat Rev. 2017;60:1–11.PubMedCrossRef
13.
Zurück zum Zitat Wu JC, Wang CT, Hung HC, Wu WJ, Wu DC, Chang MC, Sung PJ, Chou YW, Wen ZH, Tai MH. Heteronemin is a novel c-Met /STAT3 inhibitor against advanced prostate cancer cells. Prostate. 2016;76:1469–83.PubMedCrossRef Wu JC, Wang CT, Hung HC, Wu WJ, Wu DC, Chang MC, Sung PJ, Chou YW, Wen ZH, Tai MH. Heteronemin is a novel c-Met /STAT3 inhibitor against advanced prostate cancer cells. Prostate. 2016;76:1469–83.PubMedCrossRef
14.
Zurück zum Zitat Safaie Qamsari E, Safaei Ghaderi S, Zarei B, Dorostkar R, Bagheri S, Jadidi-Niaragh F, Somi MH, Yousefi M. The c-Met receptor: implication for targeted therapies in colorectal cancer. Tumour Biol. 2017;39:1010428317699118.PubMedCrossRef Safaie Qamsari E, Safaei Ghaderi S, Zarei B, Dorostkar R, Bagheri S, Jadidi-Niaragh F, Somi MH, Yousefi M. The c-Met receptor: implication for targeted therapies in colorectal cancer. Tumour Biol. 2017;39:1010428317699118.PubMedCrossRef
15.
Zurück zum Zitat 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. 2016;15:1568–79.PubMedCrossRef 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. 2016;15:1568–79.PubMedCrossRef
16.
Zurück zum Zitat Kuang W, Deng Q, Deng C, Li W, Shu S, Zhou M. Hepatocyte growth factor induces breast cancer cell invasion via the PI3K/Akt and p38 MAPK signaling pathways to up-regulate the expression of COX2. Am J Transl Res. 2017;9:3816–26.PubMedPubMedCentral Kuang W, Deng Q, Deng C, Li W, Shu S, Zhou M. Hepatocyte growth factor induces breast cancer cell invasion via the PI3K/Akt and p38 MAPK signaling pathways to up-regulate the expression of COX2. Am J Transl Res. 2017;9:3816–26.PubMedPubMedCentral
17.
Zurück zum Zitat Leung E, Xue A, Wang Y, Rougerie P, Sharma VP, Eddy R, Cox D, Condeelis J. Blood vessel endothelium-directed tumor cell streaming in breast tumors requires the HGF/C-Met signaling pathway. Oncogene. 2017;36:2680–92.PubMedCrossRef Leung E, Xue A, Wang Y, Rougerie P, Sharma VP, Eddy R, Cox D, Condeelis J. Blood vessel endothelium-directed tumor cell streaming in breast tumors requires the HGF/C-Met signaling pathway. Oncogene. 2017;36:2680–92.PubMedCrossRef
18.
Zurück zum Zitat Bahrami A, Shahidsales S, Khazaei M, Ghayour-Mobarhan M, Maftouh M, Hassanian SM, Avan A. c-Met as a potential target for the treatment of gastrointestinal cancer: current status and future perspectives. J Cell Physiol. 2017;232:2657–73.PubMedCrossRef Bahrami A, Shahidsales S, Khazaei M, Ghayour-Mobarhan M, Maftouh M, Hassanian SM, Avan A. c-Met as a potential target for the treatment of gastrointestinal cancer: current status and future perspectives. J Cell Physiol. 2017;232:2657–73.PubMedCrossRef
19.
Zurück zum Zitat Barrow-McGee R, Kishi N, Joffre C, Menard L, Hervieu A, Bakhouche BA, Noval AJ, Mai A, Guzman C, Robert-Masson L, et al. Beta 1-integrin-c-Met cooperation reveals an inside-in survival signalling on autophagy-related endomembranes. Nat Commun. 2016;7:11942.PubMedPubMedCentralCrossRef Barrow-McGee R, Kishi N, Joffre C, Menard L, Hervieu A, Bakhouche BA, Noval AJ, Mai A, Guzman C, Robert-Masson L, et al. Beta 1-integrin-c-Met cooperation reveals an inside-in survival signalling on autophagy-related endomembranes. Nat Commun. 2016;7:11942.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Caenepeel S, Cooke K, Wadsworth S, Huang G, Robert L, Moreno BH, Parisi G, Cajulis E, Kendall R, Beltran P, et al. MAPK pathway inhibition induces MET and GAB1 levels, priming BRAF mutant melanoma for rescue by hepatocyte growth factor. Oncotarget. 2017;8:17795–809.PubMedPubMedCentralCrossRef Caenepeel S, Cooke K, Wadsworth S, Huang G, Robert L, Moreno BH, Parisi G, Cajulis E, Kendall R, Beltran P, et al. MAPK pathway inhibition induces MET and GAB1 levels, priming BRAF mutant melanoma for rescue by hepatocyte growth factor. Oncotarget. 2017;8:17795–809.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Li Y, Liu H, Chen J. Dysregulation of HGF/ c-Met signal pathway and their targeting drugs in lung cancer. Zhongguo Fei Ai Za Zhi. 2014;17:625–34.PubMed Li Y, Liu H, Chen J. Dysregulation of HGF/ c-Met signal pathway and their targeting drugs in lung cancer. Zhongguo Fei Ai Za Zhi. 2014;17:625–34.PubMed
22.
Zurück zum Zitat 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. 2017;37:3209–18.PubMedPubMedCentralCrossRef 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. 2017;37:3209–18.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Hass R, Jennek S, Yang Y, Friedrich K. c-Met expression and activity in urogenital cancers - novel aspects of signal transduction and medical implications. Cell Commun Signal. 2017;15:10.PubMedPubMedCentralCrossRef Hass R, Jennek S, Yang Y, Friedrich K. c-Met expression and activity in urogenital cancers - novel aspects of signal transduction and medical implications. Cell Commun Signal. 2017;15:10.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Al-U'datt DGF, Al-Husein BAA, Qasaimeh GR. A mini-review of c-Met as a potential therapeutic target in melanoma. Biomed Pharmacother. 2017;88:194–202.PubMedCrossRef Al-U'datt DGF, Al-Husein BAA, Qasaimeh GR. A mini-review of c-Met as a potential therapeutic target in melanoma. Biomed Pharmacother. 2017;88:194–202.PubMedCrossRef
26.
27.
Zurück zum Zitat Rosário M. How to make tubes: signaling by the met receptor tyrosine kinase. Trends Cell Biol. 2003;13:328–35.PubMedCrossRef Rosário M. How to make tubes: signaling by the met receptor tyrosine kinase. Trends Cell Biol. 2003;13:328–35.PubMedCrossRef
28.
Zurück zum Zitat Vogel W, Ullrich A. Multiple in vivo phosphorylated tyrosine phosphatase SHP-2 engages binding to Grb2 via tyrosine 584. Cell Growth Differ. 1996;7:1589–97.PubMed Vogel W, Ullrich A. Multiple in vivo phosphorylated tyrosine phosphatase SHP-2 engages binding to Grb2 via tyrosine 584. Cell Growth Differ. 1996;7:1589–97.PubMed
29.
Zurück zum Zitat Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, Gong Z, Zhang S, Zhou J, Cao K, et al. Role of tumor microenvironment in tumorigenesis. J Cancer. 2017;8:761–73.PubMedPubMedCentralCrossRef Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, Gong Z, Zhang S, Zhou J, Cao K, et al. Role of tumor microenvironment in tumorigenesis. J Cancer. 2017;8:761–73.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Liebmann C. Regulation of MAP kinase activity by peptide receptor signalling pathway: paradigms of multiplicity. Cell Signal. 2001;13:777–85.PubMedCrossRef Liebmann C. Regulation of MAP kinase activity by peptide receptor signalling pathway: paradigms of multiplicity. Cell Signal. 2001;13:777–85.PubMedCrossRef
31.
Zurück zum Zitat Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 2006;441:424–30.PubMedCrossRef Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 2006;441:424–30.PubMedCrossRef
32.
Zurück zum Zitat Mottet D, Dumont V, Deccache Y, Demazy C, Ninane N, Raes M, Michiels C. Regulation of hypoxia-inducible factor-1alpha protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. J Biol Chem. 2003;278:31277–85.PubMedCrossRef Mottet D, Dumont V, Deccache Y, Demazy C, Ninane N, Raes M, Michiels C. Regulation of hypoxia-inducible factor-1alpha protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. J Biol Chem. 2003;278:31277–85.PubMedCrossRef
33.
Zurück zum Zitat Jiang BH, Liu LZ. AKT signaling in regulating angiogenesis. Curr Cancer Drug Targets. 2008;8:19–26.PubMedCrossRef Jiang BH, Liu LZ. AKT signaling in regulating angiogenesis. Curr Cancer Drug Targets. 2008;8:19–26.PubMedCrossRef
34.
Zurück zum Zitat Holland JD, Gyorffy B, Vogel R, Eckert K, Valenti G, Fang L, Lohneis P, Elezkurtaj S, Ziebold U, Birchmeier W. Combined Wnt/beta-catenin, met, and CXCL12/CXCR4 signals characterize basal breast cancer and predict disease outcome. Cell Rep. 2013;5:1214–27.PubMedCrossRef Holland JD, Gyorffy B, Vogel R, Eckert K, Valenti G, Fang L, Lohneis P, Elezkurtaj S, Ziebold U, Birchmeier W. Combined Wnt/beta-catenin, met, and CXCL12/CXCR4 signals characterize basal breast cancer and predict disease outcome. Cell Rep. 2013;5:1214–27.PubMedCrossRef
35.
Zurück zum Zitat Tuynman JB, Vermeulen L, Boon EM, Kemper K, Zwinderman AH, Peppelenbosch MP, Richel DJ. Cyclooxygenase-2 inhibition inhibits c-Met kinase activity and Wnt activity in colon cancer. Cancer Res. 2008;68:1213–20.PubMedCrossRef Tuynman JB, Vermeulen L, Boon EM, Kemper K, Zwinderman AH, Peppelenbosch MP, Richel DJ. Cyclooxygenase-2 inhibition inhibits c-Met kinase activity and Wnt activity in colon cancer. Cancer Res. 2008;68:1213–20.PubMedCrossRef
36.
Zurück zum Zitat Kim KH, Seol HJ, Kim EH, Rheey J, Jin HJ, Lee Y, Joo KM, Lee J, Nam DH. Wnt/beta-catenin signaling is a key downstream mediator of MET signaling in glioblastoma stem cells. Neuro-Oncology. 2013;15:161–71.PubMedCrossRef Kim KH, Seol HJ, Kim EH, Rheey J, Jin HJ, Lee Y, Joo KM, Lee J, Nam DH. Wnt/beta-catenin signaling is a key downstream mediator of MET signaling in glioblastoma stem cells. Neuro-Oncology. 2013;15:161–71.PubMedCrossRef
37.
Zurück zum Zitat Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/met and beta-catenin/Wnt pathways. Eur J Cancer. 2010;46:1679–91.PubMedCrossRef Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/met and beta-catenin/Wnt pathways. Eur J Cancer. 2010;46:1679–91.PubMedCrossRef
38.
Zurück zum Zitat Tortelote GG, Reis RR, de Almeida MF, Abreu JG. Complexity of the Wnt/betacatenin pathway: searching for an activation model. Cell Signal. 2017;40:30–43.PubMedCrossRef Tortelote GG, Reis RR, de Almeida MF, Abreu JG. Complexity of the Wnt/betacatenin pathway: searching for an activation model. Cell Signal. 2017;40:30–43.PubMedCrossRef
39.
Zurück zum Zitat Wu C, Zhuang Y, Jiang S, Liu S, Zhou J, Wu J, Teng Y, Xia B, Wang R, Zou X. Interaction between Wnt/beta-catenin pathway and microRNAs regulates epithelial-mesenchymal transition in gastric cancer (review). Int J Oncol. 2016;48:2236–46.PubMedCrossRef Wu C, Zhuang Y, Jiang S, Liu S, Zhou J, Wu J, Teng Y, Xia B, Wang R, Zou X. Interaction between Wnt/beta-catenin pathway and microRNAs regulates epithelial-mesenchymal transition in gastric cancer (review). Int J Oncol. 2016;48:2236–46.PubMedCrossRef
40.
Zurück zum Zitat Yan Q, Zeng Z, Gong Z, Zhang W, Li X, He B, Song Y, Li Q, Zeng Y, Liao Q, et al. EBV-miR-BART10-3p facilitates epithelial-mesenchymal transition and promotes metastasis of nasopharyngeal carcinoma by targeting BTRC. Oncotarget. 2015;6:41766–82.PubMedPubMedCentral Yan Q, Zeng Z, Gong Z, Zhang W, Li X, He B, Song Y, Li Q, Zeng Y, Liao Q, et al. EBV-miR-BART10-3p facilitates epithelial-mesenchymal transition and promotes metastasis of nasopharyngeal carcinoma by targeting BTRC. Oncotarget. 2015;6:41766–82.PubMedPubMedCentral
41.
Zurück zum Zitat Arend RC, Londono-Joshi AI, Straughn JM Jr, Buchsbaum DJ. The Wnt/beta-catenin pathway in ovarian cancer: a review. Gynecol Oncol. 2013;131:772–9.PubMedCrossRef Arend RC, Londono-Joshi AI, Straughn JM Jr, Buchsbaum DJ. The Wnt/beta-catenin pathway in ovarian cancer: a review. Gynecol Oncol. 2013;131:772–9.PubMedCrossRef
42.
Zurück zum Zitat Mariani M, McHugh M, Petrillo M, Sieber S, He S, Andreoli M, Wu Z, Fiedler P, Scambia G, Shahabi S, Ferlini C. HGF/c-Met axis drives cancer aggressiveness in the neo-adjuvant setting of ovarian cancer. Oncotarget. 2014;5:4855–67.PubMedPubMedCentralCrossRef Mariani M, McHugh M, Petrillo M, Sieber S, He S, Andreoli M, Wu Z, Fiedler P, Scambia G, Shahabi S, Ferlini C. HGF/c-Met axis drives cancer aggressiveness in the neo-adjuvant setting of ovarian cancer. Oncotarget. 2014;5:4855–67.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Raman JD. Re: prognostic value of MET, RON and histoprognostic factors for urothelial carcinoma in the upper urinary tract. E. Comperat, M. Roupret, E. Chartier-Kastler, M. O. Bitker, F. Richard, P. Camparo, F. Capron and O. Cussenot. J Urol 2008; 179: 868-872. J Urol. 2008;180:1183. author reply 1183-1184PubMedCrossRef Raman JD. Re: prognostic value of MET, RON and histoprognostic factors for urothelial carcinoma in the upper urinary tract. E. Comperat, M. Roupret, E. Chartier-Kastler, M. O. Bitker, F. Richard, P. Camparo, F. Capron and O. Cussenot. J Urol 2008; 179: 868-872. J Urol. 2008;180:1183. author reply 1183-1184PubMedCrossRef
44.
Zurück zum Zitat Cheng HL, Liu HS, Lin YJ, Chen HH, Hsu PY, Chang TY, Ho CL, Tzai TS, Chow NH. Co-expression of RON and MET is a prognostic indicator for patients with transitional-cell carcinoma of the bladder. Br J Cancer. 2005;92:1906–14.PubMedPubMedCentralCrossRef Cheng HL, Liu HS, Lin YJ, Chen HH, Hsu PY, Chang TY, Ho CL, Tzai TS, Chow NH. Co-expression of RON and MET is a prognostic indicator for patients with transitional-cell carcinoma of the bladder. Br J Cancer. 2005;92:1906–14.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Maggiora P. The RON and MET oncogenes are co-expressed in human ovarian carcinomas and cooperate in activating invasiveness. Exp Cell Res. 2003;288:382–9.PubMedCrossRef Maggiora P. The RON and MET oncogenes are co-expressed in human ovarian carcinomas and cooperate in activating invasiveness. Exp Cell Res. 2003;288:382–9.PubMedCrossRef
46.
Zurück zum Zitat McClaine RJ, Marshal AM, Wagh PK, Waltz SE. Ron receptor tyrosine Kinase activation confers resistance to Tamoxifen in breast cancer cell lines. Neoplasia. 2010;12:650–8.PubMedPubMedCentralCrossRef McClaine RJ, Marshal AM, Wagh PK, Waltz SE. Ron receptor tyrosine Kinase activation confers resistance to Tamoxifen in breast cancer cell lines. Neoplasia. 2010;12:650–8.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Previdi S, Abbadessa G, Dalo F, France DS, Broggini M. Breast cancer-derived bone metastasis can be effectively reduced through specific c-MET inhibitor tivantinib (ARQ 197) and shRNA c-MET knockdown. Mol Cancer Ther. 2012;11:214–23.PubMedCrossRef Previdi S, Abbadessa G, Dalo F, France DS, Broggini M. Breast cancer-derived bone metastasis can be effectively reduced through specific c-MET inhibitor tivantinib (ARQ 197) and shRNA c-MET knockdown. Mol Cancer Ther. 2012;11:214–23.PubMedCrossRef
48.
Zurück zum Zitat Takeuchi H, Bilchik A, Saha S, Turner R, Wiese D, Tanaka M, Kuo C, Wang HJ, Hoon DS. c-MET expression level in primary colon cancer: a predictor of tumor invasion and lymph node metastases. Clin Cancer Res. 2003;9:1480–8.PubMed Takeuchi H, Bilchik A, Saha S, Turner R, Wiese D, Tanaka M, Kuo C, Wang HJ, Hoon DS. c-MET expression level in primary colon cancer: a predictor of tumor invasion and lymph node metastases. Clin Cancer Res. 2003;9:1480–8.PubMed
49.
Zurück zum Zitat Zhao S, Ammanamanchi S, Brattain M, Cao L, Thangasamy A, Wang J, Freeman JW. Smad4-dependent TGF-beta signaling suppresses RON receptor tyrosine kinase-dependent motility and invasion of pancreatic cancer cells. J Biol Chem. 2008;283:11293–301.PubMedPubMedCentralCrossRef Zhao S, Ammanamanchi S, Brattain M, Cao L, Thangasamy A, Wang J, Freeman JW. Smad4-dependent TGF-beta signaling suppresses RON receptor tyrosine kinase-dependent motility and invasion of pancreatic cancer cells. J Biol Chem. 2008;283:11293–301.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Benvenuti S, Lazzari L, Arnesano A, Li Chiavi G, Gentile A, Comoglio PM. Ron kinase transphosphorylation sustains MET oncogene addiction. Cancer Res. 2011;71:1945–55.PubMedCrossRef Benvenuti S, Lazzari L, Arnesano A, Li Chiavi G, Gentile A, Comoglio PM. Ron kinase transphosphorylation sustains MET oncogene addiction. Cancer Res. 2011;71:1945–55.PubMedCrossRef
51.
Zurück zum Zitat Gaudino G, Follenzi A, Naldini L, Collesi C, Santoro M, Gallo KA, Godowski PJ, Comoglio PM. RON is a heterodimeric tyrosine kinase receptor activated by the HGF homologue MSP. EMBO J. 1994;13:3524–32.PubMedPubMedCentral Gaudino G, Follenzi A, Naldini L, Collesi C, Santoro M, Gallo KA, Godowski PJ, Comoglio PM. RON is a heterodimeric tyrosine kinase receptor activated by the HGF homologue MSP. EMBO J. 1994;13:3524–32.PubMedPubMedCentral
52.
Zurück zum Zitat Follenzi A, Bakovic S, Gual P, Stella MC, Longati P, Comoglio PM. Cross-talk between the proto-oncogenes met and Ron. Oncogene. 2000;19:3041–9.PubMedCrossRef Follenzi A, Bakovic S, Gual P, Stella MC, Longati P, Comoglio PM. Cross-talk between the proto-oncogenes met and Ron. Oncogene. 2000;19:3041–9.PubMedCrossRef
53.
Zurück zum Zitat Heldin CH. Dimerization of cell surface receptors in signal transduction. Cell. 1995;80:213–23.PubMedCrossRef Heldin CH. Dimerization of cell surface receptors in signal transduction. Cell. 1995;80:213–23.PubMedCrossRef
54.
Zurück zum Zitat Peace BE, Hughes MJ, Degen SJ, Waltz SE. Point mutations and overexpression of Ron induce transformation, tumor formation, and metastasis. Oncogene. 2001;20:6142–51.PubMedCrossRef Peace BE, Hughes MJ, Degen SJ, Waltz SE. Point mutations and overexpression of Ron induce transformation, tumor formation, and metastasis. Oncogene. 2001;20:6142–51.PubMedCrossRef
55.
Zurück zum Zitat Pinkas-Kramarski R, Shelly M, Glathe S, Ratzkin BJ, Yarden Y. Neu differentiation factor/neuregulin isoforms activate distinct receptor combinations. J Biol Chem. 1996;271:19029–32.PubMedCrossRef Pinkas-Kramarski R, Shelly M, Glathe S, Ratzkin BJ, Yarden Y. Neu differentiation factor/neuregulin isoforms activate distinct receptor combinations. J Biol Chem. 1996;271:19029–32.PubMedCrossRef
56.
Zurück zum Zitat Zhao S, Cao L, Freeman JW. Knockdown of RON receptor kinase delays but does not prevent tumor progression while enhancing HGF/MET signaling in pancreatic cancer cell lines. Oncogene. 2013;2:e76.CrossRef Zhao S, Cao L, Freeman JW. Knockdown of RON receptor kinase delays but does not prevent tumor progression while enhancing HGF/MET signaling in pancreatic cancer cell lines. Oncogene. 2013;2:e76.CrossRef
57.
Zurück zum Zitat Wang MH, Wang D, Chen YQ. Oncogenic and invasive potentials of human macrophage-stimulating protein receptor, the RON receptor tyrosine kinase. Carcinogenesis. 2003;24:1291–300.PubMedCrossRef Wang MH, Wang D, Chen YQ. Oncogenic and invasive potentials of human macrophage-stimulating protein receptor, the RON receptor tyrosine kinase. Carcinogenesis. 2003;24:1291–300.PubMedCrossRef
58.
Zurück zum Zitat Chaudhuri A, Xie MH, Yang B, Mahapatra K, Liu J, Marsters S, Bodepudi S, Ashkenazi A. Distinct involvement of the Gab1 and Grb2 adaptor proteins in signal transduction by the related receptor tyrosine kinases RON and MET. J Biol Chem. 2011;286:32762–74.PubMedPubMedCentralCrossRef Chaudhuri A, Xie MH, Yang B, Mahapatra K, Liu J, Marsters S, Bodepudi S, Ashkenazi A. Distinct involvement of the Gab1 and Grb2 adaptor proteins in signal transduction by the related receptor tyrosine kinases RON and MET. J Biol Chem. 2011;286:32762–74.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, Stuart W, McClaine R, Aronow B, Hoffman RM, et al. Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res. 2010;70:1130–40.PubMedPubMedCentralCrossRef Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, Stuart W, McClaine R, Aronow B, Hoffman RM, et al. Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res. 2010;70:1130–40.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Velpula KK, Dasari VR, Asuthkar S, Gorantla B, Tsung AJ. EGFR and c-Met cross talk in Glioblastoma and its regulation by human cord blood stem cells. Transl Oncol. 2012;5:379–IN318.PubMedPubMedCentralCrossRef Velpula KK, Dasari VR, Asuthkar S, Gorantla B, Tsung AJ. EGFR and c-Met cross talk in Glioblastoma and its regulation by human cord blood stem cells. Transl Oncol. 2012;5:379–IN318.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat McDermott U, Pusapati RV, Christensen JG, Gray NS, Settleman J. Acquired resistance of non-small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependency. Cancer Res. 2010;70:1625–34.PubMedPubMedCentralCrossRef McDermott U, Pusapati RV, Christensen JG, Gray NS, Settleman J. Acquired resistance of non-small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependency. Cancer Res. 2010;70:1625–34.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Agarwal S, Zerillo C, Kolmakova J, Christensen JG, Harris LN, Rimm DL, Digiovanna MP, Stern DF. Association of constitutively activated hepatocyte growth factor receptor (met) with resistance to a dual EGFR/Her2 inhibitor in non-small-cell lung cancer cells. Br J Cancer. 2009;100:941–9.PubMedPubMedCentralCrossRef Agarwal S, Zerillo C, Kolmakova J, Christensen JG, Harris LN, Rimm DL, Digiovanna MP, Stern DF. Association of constitutively activated hepatocyte growth factor receptor (met) with resistance to a dual EGFR/Her2 inhibitor in non-small-cell lung cancer cells. Br J Cancer. 2009;100:941–9.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Breindel JL, Haskins JW, Cowell EP, Zhao M, Nguyen DX, Stern DF. EGF receptor activates MET through MAPK to enhance non-small cell lung carcinoma invasion and brain metastasis. Cancer Res. 2013;73:5053–65.PubMedPubMedCentralCrossRef Breindel JL, Haskins JW, Cowell EP, Zhao M, Nguyen DX, Stern DF. EGF receptor activates MET through MAPK to enhance non-small cell lung carcinoma invasion and brain metastasis. Cancer Res. 2013;73:5053–65.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Mueller KL, Hunter LA, Ethier SP, Boerner JL. Met and c-Src cooperate to compensate for loss of epidermal growth factor receptor kinase activity in breast cancer cells. Cancer Res. 2008;68:3314–22.PubMedCrossRef Mueller KL, Hunter LA, Ethier SP, Boerner JL. Met and c-Src cooperate to compensate for loss of epidermal growth factor receptor kinase activity in breast cancer cells. Cancer Res. 2008;68:3314–22.PubMedCrossRef
65.
Zurück zum Zitat Ramalingam SS, Owonikoko TK, Khuri FR. Lung cancer: new biological insights and recent therapeutic advances. CA Cancer J Clin. 2011;61:91–112.PubMedCrossRef Ramalingam SS, Owonikoko TK, Khuri FR. Lung cancer: new biological insights and recent therapeutic advances. CA Cancer J Clin. 2011;61:91–112.PubMedCrossRef
66.
Zurück zum Zitat Wang W, Li Q, Takeuchi S, Yamada T, Koizumi H, Nakamura T, Matsumoto K, Mukaida N, Nishioka Y, Sone S, et al. Met kinase inhibitor E7050 reverses three different mechanisms of hepatocyte growth factor-induced tyrosine kinase inhibitor resistance in EGFR mutant lung cancer. Clin Cancer Res. 2012;18:1663–71.PubMedCrossRef Wang W, Li Q, Takeuchi S, Yamada T, Koizumi H, Nakamura T, Matsumoto K, Mukaida N, Nishioka Y, Sone S, et al. Met kinase inhibitor E7050 reverses three different mechanisms of hepatocyte growth factor-induced tyrosine kinase inhibitor resistance in EGFR mutant lung cancer. Clin Cancer Res. 2012;18:1663–71.PubMedCrossRef
67.
Zurück zum Zitat Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43.PubMedCrossRef Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43.PubMedCrossRef
68.
Zurück zum Zitat Benedettini E, Sholl LM, Peyton M, Reilly J, Ware C, Davis L, Vena N, Bailey D, Yeap BY, Fiorentino M, et al. Met activation in non-small cell lung cancer is associated with de novo resistance to EGFR inhibitors and the development of brain metastasis. Am J Pathol. 2010;177:415–23.PubMedPubMedCentralCrossRef Benedettini E, Sholl LM, Peyton M, Reilly J, Ware C, Davis L, Vena N, Bailey D, Yeap BY, Fiorentino M, et al. Met activation in non-small cell lung cancer is associated with de novo resistance to EGFR inhibitors and the development of brain metastasis. Am J Pathol. 2010;177:415–23.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Sequist LV, Martins RG, Spigel D, Grunberg SM, Spira A, Janne PA, Joshi VA, McCollum D, Evans TL, Muzikansky A, et al. First-line gefitinib in patients with advanced non-small-cell lung cancer harboring somatic EGFR mutations. J Clin Oncol. 2008;26:2442–9.PubMedCrossRef Sequist LV, Martins RG, Spigel D, Grunberg SM, Spira A, Janne PA, Joshi VA, McCollum D, Evans TL, Muzikansky A, et al. First-line gefitinib in patients with advanced non-small-cell lung cancer harboring somatic EGFR mutations. J Clin Oncol. 2008;26:2442–9.PubMedCrossRef
70.
Zurück zum Zitat Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D, Lifshits E, Toschi L, Rogers A, Mok T, Sequist L, et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell. 2010;17:77–88.PubMedPubMedCentralCrossRef Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D, Lifshits E, Toschi L, Rogers A, Mok T, Sequist L, et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell. 2010;17:77–88.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Raghav KP, Gonzalez-Angulo AM, Blumenschein GR Jr. Role of HGF/MET axis in resistance of lung cancer to contemporary management. Transl Lung Cancer Res. 2012;1:179–93.PubMedPubMedCentral Raghav KP, Gonzalez-Angulo AM, Blumenschein GR Jr. Role of HGF/MET axis in resistance of lung cancer to contemporary management. Transl Lung Cancer Res. 2012;1:179–93.PubMedPubMedCentral
72.
Zurück zum Zitat Kaposi-Novak P, Lee JS, Gomez-Quiroz L, Coulouarn C, Factor VM, Thorgeirsson SS. Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype. J Clin Invest. 2006;116:1582–95.PubMedPubMedCentralCrossRef Kaposi-Novak P, Lee JS, Gomez-Quiroz L, Coulouarn C, Factor VM, Thorgeirsson SS. Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype. J Clin Invest. 2006;116:1582–95.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat 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. 2009;49:491–503.PubMedCrossRef 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. 2009;49:491–503.PubMedCrossRef
74.
Zurück zum Zitat Wang ZL, Liang P, Dong BW, Yu XL, Yu DJ. Prognostic factors and recurrence of small hepatocellular carcinoma after hepatic resection or microwave ablation: a retrospective study. J Gastrointest Surg. 2008;12:327–37.PubMedCrossRef Wang ZL, Liang P, Dong BW, Yu XL, Yu DJ. Prognostic factors and recurrence of small hepatocellular carcinoma after hepatic resection or microwave ablation: a retrospective study. J Gastrointest Surg. 2008;12:327–37.PubMedCrossRef
75.
Zurück zum Zitat Ueki T, Fujimoto J, Suzuki T, Yamamoto H, Okamoto E. Expression of hepatocyte growth factor and its receptor c-met proto-oncogene in hepatocellular carcinoma. Hepatology. 1997;25:862–6.PubMedCrossRef Ueki T, Fujimoto J, Suzuki T, Yamamoto H, Okamoto E. Expression of hepatocyte growth factor and its receptor c-met proto-oncogene in hepatocellular carcinoma. Hepatology. 1997;25:862–6.PubMedCrossRef
76.
Zurück zum Zitat Viticchie G, PAJ M. c-Met and other cell surface molecules: interaction, activation and functional consequences. Biomedicine. 2015;3:46–70.CrossRef Viticchie G, PAJ M. c-Met and other cell surface molecules: interaction, activation and functional consequences. Biomedicine. 2015;3:46–70.CrossRef
77.
Zurück zum Zitat You H, Ding W, Dang H, Jiang Y, Rountree CB. c-Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinoma. Hepatology. 2011;54:879–89.PubMedPubMedCentralCrossRef You H, Ding W, Dang H, Jiang Y, Rountree CB. c-Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinoma. Hepatology. 2011;54:879–89.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Yarden Y. The EGFR family and its ligands in human cancer. Signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001;37(Suppl 4):S3–8.PubMedCrossRef Yarden Y. The EGFR family and its ligands in human cancer. Signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001;37(Suppl 4):S3–8.PubMedCrossRef
79.
Zurück zum Zitat Merchant M, Ma X, Maun HR, Zheng Z, Peng J, Romero M, Huang A, Yang NY, Nishimura M, Greve J, et al. Monovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent. Proc Natl Acad Sci U S A. 2013;110:E2987–96.PubMedPubMedCentralCrossRef Merchant M, Ma X, Maun HR, Zheng Z, Peng J, Romero M, Huang A, Yang NY, Nishimura M, Greve J, et al. Monovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent. Proc Natl Acad Sci U S A. 2013;110:E2987–96.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Gelsomino F, Facchinetti F, Haspinger ER, Garassino MC, Trusolino L, De Braud F, Tiseo M. Targeting the MET gene for the treatment of non-small-cell lung cancer. Crit Rev Oncol Hematol. 2014;89:284–99.PubMedCrossRef Gelsomino F, Facchinetti F, Haspinger ER, Garassino MC, Trusolino L, De Braud F, Tiseo M. Targeting the MET gene for the treatment of non-small-cell lung cancer. Crit Rev Oncol Hematol. 2014;89:284–99.PubMedCrossRef
81.
Zurück zum Zitat Camidge DR, Ou SHI, Shapiro G, Otterson GA, Villaruz LC, Villalona-Calero MA. Efficacy and safety of crizotinib in patients with advanced c-MET-amplified non-small cell lung cancer (NSCLC). 2014. Camidge DR, Ou SHI, Shapiro G, Otterson GA, Villaruz LC, Villalona-Calero MA. Efficacy and safety of crizotinib in patients with advanced c-MET-amplified non-small cell lung cancer (NSCLC). 2014.
82.
Zurück zum Zitat Shea M, Costa DB, Rangachari D. Management of advanced non-small cell lung cancers with known mutations or rearrangements: latest evidence and treatment approaches. Ther Adv Respir Dis. 2016;10:113–29.PubMedCrossRef Shea M, Costa DB, Rangachari D. Management of advanced non-small cell lung cancers with known mutations or rearrangements: latest evidence and treatment approaches. Ther Adv Respir Dis. 2016;10:113–29.PubMedCrossRef
83.
Zurück zum Zitat Facchinetti F, Rossi G, Bria E, Soria JC, Besse B, Minari R, Friboulet L, Tiseo M. Oncogene addiction in non-small cell lung cancer: focus on ROS1 inhibition. Cancer Treat Rev. 2017;55:83–95.PubMedCrossRef Facchinetti F, Rossi G, Bria E, Soria JC, Besse B, Minari R, Friboulet L, Tiseo M. Oncogene addiction in non-small cell lung cancer: focus on ROS1 inhibition. Cancer Treat Rev. 2017;55:83–95.PubMedCrossRef
84.
Zurück zum Zitat Huang XX, Xie FF, Hou LJ, Chen XX, Ou RY, Yu JT, Qiu JG, Zhang WJ, Jiang QW, Yang Y, et al. Crizotinib synergizes with cisplatin in preclinical models of ovarian cancer. Am J Transl Res. 2017;9:1667–79.PubMedPubMedCentral Huang XX, Xie FF, Hou LJ, Chen XX, Ou RY, Yu JT, Qiu JG, Zhang WJ, Jiang QW, Yang Y, et al. Crizotinib synergizes with cisplatin in preclinical models of ovarian cancer. Am J Transl Res. 2017;9:1667–79.PubMedPubMedCentral
85.
Zurück zum Zitat Stanley A, Ashrafi GH, Seddon AM, Modjtahedi H. Synergistic effects of various her inhibitors in combination with IGF-1R, c-Met and Src targeting agents in breast cancer cell lines. Sci Rep. 2017;7:3964.PubMedPubMedCentralCrossRef Stanley A, Ashrafi GH, Seddon AM, Modjtahedi H. Synergistic effects of various her inhibitors in combination with IGF-1R, c-Met and Src targeting agents in breast cancer cell lines. Sci Rep. 2017;7:3964.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Vergote IB, Smith DC, Berger R, Kurzrock R, Vogelzang NJ, Sella A, Wheler J, Lee Y, Foster PG, Weitzman R, Buckanovich RJ. A phase 2 randomised discontinuation trial of cabozantinib in patients with ovarian carcinoma. Eur J Cancer. 2017;83:229–36.PubMedCrossRef Vergote IB, Smith DC, Berger R, Kurzrock R, Vogelzang NJ, Sella A, Wheler J, Lee Y, Foster PG, Weitzman R, Buckanovich RJ. A phase 2 randomised discontinuation trial of cabozantinib in patients with ovarian carcinoma. Eur J Cancer. 2017;83:229–36.PubMedCrossRef
87.
Zurück zum Zitat Escudier B, Lougheed JC, Albiges L. Cabozantinib for the treatment of renal cell carcinoma. Expert Opin Pharmacother. 2016;17:2499–504.PubMedCrossRef Escudier B, Lougheed JC, Albiges L. Cabozantinib for the treatment of renal cell carcinoma. Expert Opin Pharmacother. 2016;17:2499–504.PubMedCrossRef
88.
Zurück zum Zitat Bentzien F, Zuzow M, Heald N, Gibson A, Shi Y, Goon L, Yu P, Engst S, Zhang W, Huang D, et al. In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid. 2013;23:1569–77.PubMedPubMedCentralCrossRef Bentzien F, Zuzow M, Heald N, Gibson A, Shi Y, Goon L, Yu P, Engst S, Zhang W, Huang D, et al. In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid. 2013;23:1569–77.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, Hammers HJ, Donskov F, Roth BJ, Peltola K, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. The Lancet Oncology. 2016;17:917–27.PubMedCrossRef Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, Hammers HJ, Donskov F, Roth BJ, Peltola K, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. The Lancet Oncology. 2016;17:917–27.PubMedCrossRef
90.
Zurück zum Zitat Shintani T, Kusuhara Y, Daizumoto K, Dondoo TO, Yamamoto H, Mori H, Fukawa T, Nakatsuji H, Fukumori T, Takahashi M, Kanayama H. The involvement of Hepatocyte growth factor-MET-matrix metalloproteinase 1 Signaling in bladder cancer invasiveness and proliferation. Effect of the MET inhibitor, Cabozantinib (XL184), on bladder cancer cells. Urology. 2017;101:169 e167–13.CrossRef Shintani T, Kusuhara Y, Daizumoto K, Dondoo TO, Yamamoto H, Mori H, Fukawa T, Nakatsuji H, Fukumori T, Takahashi M, Kanayama H. The involvement of Hepatocyte growth factor-MET-matrix metalloproteinase 1 Signaling in bladder cancer invasiveness and proliferation. Effect of the MET inhibitor, Cabozantinib (XL184), on bladder cancer cells. Urology. 2017;101:169 e167–13.CrossRef
91.
Zurück zum Zitat Shah MA, Wainberg ZA, Catenacci DV, Hochster HS, Ford J, Kunz P, Lee FC, Kallender H, Cecchi F, Rabe DC, et al. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One. 2013;8:e54014.PubMedPubMedCentralCrossRef Shah MA, Wainberg ZA, Catenacci DV, Hochster HS, Ford J, Kunz P, Lee FC, Kallender H, Cecchi F, Rabe DC, et al. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One. 2013;8:e54014.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Choueiri TK, Vaishampayan U, Rosenberg JE, Logan TF, Harzstark AL, Bukowski RM, Rini BI, Srinivas S, Stein MN, Adams LM, et al. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol. 2013;31:181–6.PubMedCrossRef Choueiri TK, Vaishampayan U, Rosenberg JE, Logan TF, Harzstark AL, Bukowski RM, Rini BI, Srinivas S, Stein MN, Adams LM, et al. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol. 2013;31:181–6.PubMedCrossRef
93.
Zurück zum Zitat Chia SK, Ellard SL, Mates M, Welch S, Mihalcioiu C, Miller WH Jr, Gelmon K, Lohrisch C, Kumar V, Taylor S, et al. A phase-I study of lapatinib in combination with foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in human epidermal growth factor receptor 2 (HER-2)-positive metastatic breast cancer. Breast Cancer Res. 2017;19:54.PubMedPubMedCentralCrossRef Chia SK, Ellard SL, Mates M, Welch S, Mihalcioiu C, Miller WH Jr, Gelmon K, Lohrisch C, Kumar V, Taylor S, et al. A phase-I study of lapatinib in combination with foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in human epidermal growth factor receptor 2 (HER-2)-positive metastatic breast cancer. Breast Cancer Res. 2017;19:54.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Cheng H, Chua V, Liao C, Purwin TJ, Terai M, Kageyama K, Davies MA, Sato T, Aplin AE. Co-targeting HGF/cMET Signaling with MEK inhibitors in metastatic Uveal melanoma. Mol Cancer Ther. 2017;16:516–28.PubMedPubMedCentralCrossRef Cheng H, Chua V, Liao C, Purwin TJ, Terai M, Kageyama K, Davies MA, Sato T, Aplin AE. Co-targeting HGF/cMET Signaling with MEK inhibitors in metastatic Uveal melanoma. Mol Cancer Ther. 2017;16:516–28.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Barat S, Bozko P, Chen X, Scholta T, Hanert F, Gotze J, Malek NP, Wilkens L, Plentz RR. Targeting c-MET by LY2801653 for treatment of cholangiocarcinoma. Mol Carcinog. 2016;55:2037–50.PubMedCrossRef Barat S, Bozko P, Chen X, Scholta T, Hanert F, Gotze J, Malek NP, Wilkens L, Plentz RR. Targeting c-MET by LY2801653 for treatment of cholangiocarcinoma. Mol Carcinog. 2016;55:2037–50.PubMedCrossRef
96.
Zurück zum Zitat Wang J, Cheng JX. c-Met inhibition enhances chemosensitivity of human ovarian cancer cells. Clin Exp Pharmacol Physiol. 2017;44:79–87.PubMedCrossRef Wang J, Cheng JX. c-Met inhibition enhances chemosensitivity of human ovarian cancer cells. Clin Exp Pharmacol Physiol. 2017;44:79–87.PubMedCrossRef
97.
Zurück zum Zitat Liu X, Wang Q, Yang G, Marando C, Koblish HK, Hall LM, Fridman JS, Behshad E, Wynn R, Li Y, et al. A novel kinase inhibitor, INCB28060, blocks c-MET-dependent signaling, neoplastic activities, and cross-talk with EGFR and HER-3. Clin Cancer Res. 2011;17:7127–38.PubMedCrossRef Liu X, Wang Q, Yang G, Marando C, Koblish HK, Hall LM, Fridman JS, Behshad E, Wynn R, Li Y, et al. A novel kinase inhibitor, INCB28060, blocks c-MET-dependent signaling, neoplastic activities, and cross-talk with EGFR and HER-3. Clin Cancer Res. 2011;17:7127–38.PubMedCrossRef
98.
Zurück zum Zitat Okusaka T, Aramaki T, Inaba Y, Nakamura S, Morimoto M, Moriguchi M, Sato T, Ikawa Y, Ikeda M, Furuse J. Phase I study of tivantinib in Japanese patients with advanced hepatocellular carcinoma: distinctive pharmacokinetic profiles from other solid tumors. Cancer Sci. 2015;106:611–7.PubMedPubMedCentralCrossRef Okusaka T, Aramaki T, Inaba Y, Nakamura S, Morimoto M, Moriguchi M, Sato T, Ikawa Y, Ikeda M, Furuse J. Phase I study of tivantinib in Japanese patients with advanced hepatocellular carcinoma: distinctive pharmacokinetic profiles from other solid tumors. Cancer Sci. 2015;106:611–7.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Xiang Q, Zhen Z, Deng DY, Wang J, Chen Y, Li J, Zhang Y, Wang F, Chen N, Chen H, Chen Y. Tivantinib induces G2/M arrest and apoptosis by disrupting tubulin polymerization in hepatocellular carcinoma. J Exp Clin Cancer Res. 2015;34:118.PubMedPubMedCentralCrossRef Xiang Q, Zhen Z, Deng DY, Wang J, Chen Y, Li J, Zhang Y, Wang F, Chen N, Chen H, Chen Y. Tivantinib induces G2/M arrest and apoptosis by disrupting tubulin polymerization in hepatocellular carcinoma. J Exp Clin Cancer Res. 2015;34:118.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Best J, Schotten C, Lohmann G, Gerken G, Dechene A. Tivantinib for the treatment of hepatocellular carcinoma. Expert Opin Pharmacother. 2017;18:727–33.PubMedCrossRef Best J, Schotten C, Lohmann G, Gerken G, Dechene A. Tivantinib for the treatment of hepatocellular carcinoma. Expert Opin Pharmacother. 2017;18:727–33.PubMedCrossRef
101.
Zurück zum Zitat Sequist LV, von Pawel J, Garmey EG, Akerley WL, Brugger W, Ferrari D, Chen Y, Costa DB, Gerber DE, Orlov S, et al. Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol. 2011;29:3307–15.PubMedCrossRef Sequist LV, von Pawel J, Garmey EG, Akerley WL, Brugger W, Ferrari D, Chen Y, Costa DB, Gerber DE, Orlov S, et al. Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol. 2011;29:3307–15.PubMedCrossRef
102.
Zurück zum Zitat 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. 2015;26:2066–72.PubMedCrossRef 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. 2015;26:2066–72.PubMedCrossRef
103.
Zurück zum Zitat Scagliotti G, von Pawel J, Novello S, Ramlau R, Favaretto A, Barlesi F, Akerley W, Orlov S, Santoro A, Spigel D, et al. Phase III multinational, randomized, double-blind, placebo-controlled study of Tivantinib (ARQ 197) plus Erlotinib versus Erlotinib alone in previously treated patients with locally advanced or metastatic Nonsquamous non-small-cell lung cancer. J Clin Oncol. 2015;33:2667–74.PubMedCrossRef Scagliotti G, von Pawel J, Novello S, Ramlau R, Favaretto A, Barlesi F, Akerley W, Orlov S, Santoro A, Spigel D, et al. Phase III multinational, randomized, double-blind, placebo-controlled study of Tivantinib (ARQ 197) plus Erlotinib versus Erlotinib alone in previously treated patients with locally advanced or metastatic Nonsquamous non-small-cell lung cancer. J Clin Oncol. 2015;33:2667–74.PubMedCrossRef
104.
Zurück zum Zitat Puzanov I, Sosman J, Santoro A, Saif MW, Goff L, Dy GK, Zucali P, Means-Powell JA, Ma WW, Simonelli M, et al. Phase 1 trial of tivantinib in combination with sorafenib in adult patients with advanced solid tumors. Investig New Drugs. 2015;33:159–68.CrossRef Puzanov I, Sosman J, Santoro A, Saif MW, Goff L, Dy GK, Zucali P, Means-Powell JA, Ma WW, Simonelli M, et al. Phase 1 trial of tivantinib in combination with sorafenib in adult patients with advanced solid tumors. Investig New Drugs. 2015;33:159–68.CrossRef
105.
Zurück zum Zitat Klempner SJ, Borghei A, Hakimian B, Ali SM, Ou SI. Intracranial activity of Cabozantinib in MET exon 14-positive NSCLC with brain metastases. J Thorac Oncol. 2017;12:152–6.PubMedCrossRef Klempner SJ, Borghei A, Hakimian B, Ali SM, Ou SI. Intracranial activity of Cabozantinib in MET exon 14-positive NSCLC with brain metastases. J Thorac Oncol. 2017;12:152–6.PubMedCrossRef
106.
Zurück zum Zitat Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer J. 2015;21:111–6.PubMedCrossRef Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer J. 2015;21:111–6.PubMedCrossRef
107.
Zurück zum Zitat Corbet C, Feron O. Tumour acidosis: from the passenger to the driver's seat. Nat Rev Cancer. 2017;17:577–93.PubMedCrossRef Corbet C, Feron O. Tumour acidosis: from the passenger to the driver's seat. Nat Rev Cancer. 2017;17:577–93.PubMedCrossRef
108.
Zurück zum Zitat Fan C, Tang Y, Wang J, Xiong F, Guo C, Wang Y, Zhang S, Gong Z, Wei F, Yang L, et al. Role of long non-coding RNAs in glucose metabolism in cancer. Mol Cancer. 2017;16:130.PubMedPubMedCentralCrossRef Fan C, Tang Y, Wang J, Xiong F, Guo C, Wang Y, Zhang S, Gong Z, Wei F, Yang L, et al. Role of long non-coding RNAs in glucose metabolism in cancer. Mol Cancer. 2017;16:130.PubMedPubMedCentralCrossRef
109.
110.
Zurück zum Zitat Song Y, Li X, Zeng Z, Li Q, Gong Z, Liao Q, Li X, Chen P, Xiang B, Zhang W, et al. Epstein-Barr virus encoded miR-BART11 promotes inflammation-induced carcinogenesis by targeting FOXP1. Oncotarget. 2016;7:36783–99.PubMedPubMedCentral Song Y, Li X, Zeng Z, Li Q, Gong Z, Liao Q, Li X, Chen P, Xiang B, Zhang W, et al. Epstein-Barr virus encoded miR-BART11 promotes inflammation-induced carcinogenesis by targeting FOXP1. Oncotarget. 2016;7:36783–99.PubMedPubMedCentral
Metadaten
Titel
Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities
Publikationsdatum
01.12.2018
Erschienen in
Molecular Cancer / Ausgabe 1/2018
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0796-y

Weitere Artikel der Ausgabe 1/2018

Molecular Cancer 1/2018 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.