A novel c-Met inhibitor, MK8033, synergizes with carboplatin plus paclitaxel to inhibit ovarian cancer cell growth

  • Authors:
    • Douglas C. Marchion
    • Elona Bicaku
    • Yin Xiong
    • Nadim Bou Zgheib
    • Entidhar Al Sawah
    • Xiaomang Ba Stickles
    • Patricia L. Judson
    • Alex S. Lopez
    • Christopher L. Cubitt
    • Jesus Gonzalez-Bosquet
    • Robert M. Wenham
    • Sachin M. Apte
    • Anders Berglund
    • Johnathan M. Lancaster
  • View Affiliations

  • Published online on: March 5, 2013     https://doi.org/10.3892/or.2013.2329
  • Pages: 2011-2018
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Elevated serum levels of hepatocyte growth factor (HGF) and high tumor expression of c-Met are both indicators of poor overall survival from ovarian cancer (OVCA). In the present study, we evaluated the role of the HGF signaling pathway in OVCA cell line chemoresistance and OVCA patient overall survival as well as the influence of HGF/c-Met signaling inhibition on the sensitivity of OVCA cells to combinational carboplatin plus paclitaxel therapy. The prevalence of the HGF receptor, c-Met, was determined by immunohistochemistry in primary OVCA samples (n=79) and OVCA cell lines (n=41). The influence of the c-Met-specific inhibitor MK8033 on OVCA cell sensitivity to combinations of carboplatin plus paclitaxel was examined in a subset of OVCA cells (n=8) by CellTiter-Blue cell viability assays. Correlation tests were used to identify genes associated with response to MK8033 and carboplatin plus paclitaxel. Identified genes were evaluated for influence on overall survival from OVCA using principal component analysis (PCA) modeling in an independent clinical OVCA dataset (n=218). Immunohistochemistry analysis indicated that 83% of OVCA cells and 92% of primary OVCA expressed the HGF receptor, c-Met. MK8033 exhibited significant anti-proliferative effects against a panel of human OVCA cell lines. Combination index values determined by the Chou-Talalay isobologram equation indicated synergistic activity in combinations of MK8033 and carboplatin plus paclitaxel. Pearson's correlation identified a 47-gene signature to be associated with MK8033-carboplatin plus paclitaxel response. PCA modeling indicated an association of this 47-gene response signature with overall survival from OVCA (P=0.013). These data indicate that HGF/c-Met pathway signaling may influence OVCA chemosensitivity and overall patient survival. Furthermore, HGF/c-Met inhibition by MK8033 represents a promising new therapeutic avenue to increase OVCA sensitivity to carboplatin plus paclitaxel.

Introduction

Ovarian cancer (OVCA) is the fifth leading cause of cancer-related mortality among women in the United States and Western Europe, with the highest mortality rate of all gynecologic malignancies. Approximately 75% of patients with OVCA are diagnosed at an advanced stage (III/IV) with disseminated intraperitoneal metastases (1). Although the majority of patients initially experience a complete clinical response to primary surgery followed by platinum/taxane-based chemotherapy, most patients eventually develop platinum/taxane-resistant persistent or recurrent disease. These patients have a poor prognosis. Despite cytoreductive surgery and aggressive chemotherapy, the majority of patients with OVCA succumb to the disease within 5 years (2). These dismal statistics highlight the need for research into the cellular basis of platinum response, as well as the design of targeted therapeutic strategies.

The hepatocyte growth factor (HGF) signaling pathway may be a viable target for the development of directed therapeutic regimens for the treatment of OVCA. HGF is the only known ligand of the HGF receptor (c-Met), which is expressed in approximately 70% of OVCAs (3,4). Overexpression of HGF and/or c-Met has been associated with poor clinical outcome in OVCA (57), whereas c-Met overexpression has further been associated with a chemo-resistant subset of ovarian clear-cell adenocarcinomas (8,9). The targeted inhibition of c-Met has been shown to reduce c-Met expression, block OVCA cell proliferation, and reduce tumor burden in pre-clinical mouse models (1012).

In this study, we analyzed the expression of the HGF receptor, c-Met, in primary OVCA and OVCA cell lines, and tested the activity of the HGF/c-Met signaling inhibitor, MK8033, alone and in combination with standard of care therapy (carboplatin plus paclitaxel) in a panel of OVCA cells. We then identified the genes associated with sensitivity to MK8033 plus carboplatin-paclitaxel and used the principal component analysis (PCA) to define a gene expression signature of OVCA cell response. Finally, this PCA response signature was tested in an independent survival dataset of primary OVCA for which response to chemotherapy and overall survival were known.

Materials and methods

Cell culture

OVCA cell lines were obtained from the American Type Culture Collection (Manassas, VA, USA) (CAOV3, OV90, OVCAR3 and SKOV3); from the European Collection of Cell Cultures (Salisbury, England) (A2780CP and A2780S); from Kyoto University (Japan) (CHI, CHIcisR, M41, M41CSR, Tyknu and TyknuCisR); or were kind gifts from Dr Patricia Kruk, Department of Pathology, College of Medicine, University of South Florida, Tampa, FL, and Professor Susan Murphy, Department of OBGYN/Division of Gynecologic Oncology, Duke University, Durham, NC, USA (A2008, C13, CAOV2, HeyA8, IGR-OV1, IMCC3, IMCC5, MCAS, OV2008, OVCA420, OVCA429, OVCA432, OVCA433, FUOV1, PEO1, PEO4, SK-OV-6, T8, TOV-112D, TOV-21-G, Dov13, BG1, Ovary1847, OVCAR10, OVCAR8, OVCAR5, OVCAR4, OVCAR2 and SK-OV-4). Cell lines were maintained in RPMI-1640 (Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (Fisher Scientific, Pittsburg, PA, USA), 1% sodium pyruvate, 1% penicillin/streptomycin (Cellgro, Manassas, VA, USA), and 1% nonessential amino acids (HyClone, Hudson, NH, USA). Mycoplasma testing was performed every 6 months in accordance with the manufacturer's protocol (Lonza, Rockland, ME, USA).

Staining and scoring of c-Met expression in OVCA

Slides were stained using a Ventana Discovery XT automated system as per the manufacturer's protocol with proprietary reagents (Ventana Medical Systems, Tucson, AZ, USA). Briefly, slides were deparaffinized on the automated system with EZ Prep solution (Ventana). The heat-induced antigen retrieval method was used in Cell Conditioning 1 (Ventana). The mouse monoclonal antibody that reacts to c-Met (#18-7366; Invitrogen) was used at a 1:2,000 concentration in PSS antibody diluent (Ventana) and incubated for 60 min. The Ventana OmniMap anti-mouse secondary antibody was used for 16 min. The detection system used was the Ventana ChromoMap kit, and slides were counterstained with hematoxylin. Slides were then dehydrated and coverslipped as per normal laboratory protocol. The c-Met expression score was defined as the product of intensity and cellularity, where an intensity of 1 was weak; 2, moderate; 3, strong, and a cellularity of 1 ≤33%, 2 =34–65% and 3 ≥66%.

RNA extraction and microarray expression analysis

RNA extraction and microarray expression analyses were performed as previously described (13). Briefly, RNA was extracted using the RNeasy kit following the manufacturer's recommendations (Qiagen, Valencia, CA, USA). Quality of the RNA was measured using an Agilent 2100 Bioanalyzer. The targets for Affymetrix DNA microarray analysis were prepared according to the manufacturer's instructions. For OVCA cell lines (n=41), targets were hybridized to customized Human Affymetrix HuRSTA gene chips (HuRSTA-2a520709) [GEO accession number GSE34615 (14)].

Statistical analyses

Expression data were subjected to background correction and normalization using the Robust Multichip Average algorithm in the Affymetrix Expression Console (http://www.affymetrix.com/products/software/specific/expression_console_software.affx). Pearson's correlation test was performed on individual gene expression and IC50 values. Probe sets with P<0.001 were considered to have significant correlations with IC50 values.

Cell viability assays

For cells cultured in the presence of cisplatin, resistance was quantified using MTT proliferation assay, in accordance with manufacturer's instructions (Promega, Madison, WI, USA). Cells (5,000/well) were seeded in 96-well plates, allowed to adhere, and incubated with increasing doses of cisplatin for 48 h at 37°C. Following drug incubation, plates were read at 570 nm using a SpectraMax 190 microplate reader (Molecular Devices, USA). For drug combination experiments, CellTiter-Blue cell viability assays were performed using 384-well plates and an automated pipetting station. Briefly, for these assays, 1.2×103 cells (24-μl volume) were plated in each well and allowed to adhere overnight at 37°C and 5% CO2. Drug dilutions were then added using the same pipetting station, and the plate was incubated for 72 h. Following drug incubation, fluorescence was measured using a Synergy 4 microplate reader (Bio-Tek Instruments, Inc.). The fluorescence data were transferred to a spreadsheet program to calculate the percent viability relative to untreated cells. Experiments were repeated a minimum of three times.

Synergism analyses

Drug combination experiments were analyzed for synergistic, additive, or antagonistic effects using the combination index method developed by Chou and Talalay. For the application of this method, the drug concentration dilutions of the drugs were used at fixed dose ratios based on the IC50 values of each drug obtained from preliminary experiments (e.g., 50:1, 2:5, 1:250). Briefly, the dose-effect curve for each drug alone is determined based on experimental observations using the median-effect principle and is compared to the effect achieved with a combination of the two drugs to derive a combination index value. This method involves plotting dose-effect curves, for each agent and their combination, using the following median-effect equation: fa/fu = (D/Dm)m, where D is the dose of the drug, Dm is the dose required for a 50% effect (equivalent to IC50), fa and fu are the affected and unaffected fractions, respectively (fa=1-fu), and m is the exponent signifying the sigmoidicity of the dose-effect curve. The computer software program XLfit was used to calculate the values of Dm and m. The combination index values used for the analysis of the drug combinations were determined by the isobologram equation for mutually nonexclusive drugs that have different modes of action: combination index = (D)1/(Dx)1 + (D)2/(Dx)2 + (D)1(D)2/(Dx)1(Dx)2, where (Dx)1 and (Dx)2 in the denominators are the doses (or concentrations) for D1 (drug 1) and D2 (drug 2) alone that gives x% inhibition, whereas (D)1 and (D)2 in the numerators are the doses of drugs 1 and 2 in combination that also inhibited x% (i.e., isoeffective). Combination indices <1, equal to 1, and >1 indicate synergism, additive effects and antagonism, respectively.

Building signatures

The PCA methodology was used to derive a gene expression signature with a corresponding score. First, the data were reduced into a small set of uncorrelated principal components. This set of principal components was generated based on its ability to account for variation. The first principal component (1st PCA) is used to represent the overall variability in the data. The gene expression score is equal to ∑wixi, a weighted average expression among the differentially expressed genes, where xi represents gene i expression level, wi is the corresponding weight (loading coefficient) with ∑w2i = 1, and the wi values maximize the variance of ∑wixi. Directional signs of PCA scores are recognized to be arbitrary and can vary between software and the algorithm used to calculate the PCA model (15); however, this does not affect the interpretation of the PCA model and can be easily solved by multiplying both scores and loadings by −1, a 180° rotation. Details of this methodology have been reported by our group previously (13).

Associations with overall survival from OVCA

PCA models were then explored for associations with overall survival from OVCA (using median PC1 score as the threshold to define high vs. low pathway score) using a dataset for which both gene expression and overall survival data were available, the publicly available Australian (Aus) Dataset (Affymetrix U133Plus GeneChips, n=218).

Results

c-Met is significantly expressed in OVCA

Since overexpression of HGF has been associated with poor clinical outcome in OVCA (57), we evaluated the expression of c-Met, the only known HGF receptor, in OVCA cells and primary OVCA tumor samples. Pathological scoring of c-Met immunostains (cellularity × intensity) in 79 primary OVCA samples and 41 OVCA cell lines indicated the presence of c-Met expression in 92% (73/79) (Table I) and 83% (34/41) (Table II) of samples, respectively. c-Met expression in primary OVCA did not appear to be associated with stage, response to primary platinum therapy, or CA125 levels (Table I).

Table I

c-Met expression in ovarian cancer samples and available clinical data.

Table I

c-Met expression in ovarian cancer samples and available clinical data.

SampleDiagnosisScoreCR vs. IRDebulkingAge (years)StageCA125
1Focal adenocarcinoma6CRO57IIIC541
2Papillary serous adenocarcinoma1CRO84IIIC7350
3Adenocarcinoma2CRO53IIIC
4Papillary serous adenocarcinoma4CRO61IIIC
5Papillary serous adenocarcinoma6CRO57IIIC
6Papillary serous adenocarcinoma1CRO55IV
7Papillary serous adenocarcinoma2CRO49IIIC305
8Papillary serous adenocarcinoma1CRO65IIIC
9Papillary serous adenocarcinoma4CRO60IIIC925
10Papillary serous adenocarcinoma2CRO51IIIB
11Papillary serous adenocarcinoma4CRO56IIIC316
12Papillary serous adenocarcinoma2CRO66IIIC224
13Papillary serous adenocarcinoma2CRS56IIIC
14Papillary serous adenocarcinoma4CRO73IIIC
15Papillary serous adenocarcinoma1CRO62IIIC1717
16Papillary serous adenocarcinoma2CRO78IIIC
17Papillary serous adenocarcinoma6CRO43IIIC64
18Papillary serous adenocarcinoma1CRO45IIIC
19Papillary serous adenocarcinoma1CRS74IIIC1800
20Papillary serous adenocarcinoma2CRO76IIIC
21Papillary serous adenocarcinoma2IRS79IV
22Papillary serous adenocarcinoma2IRS71IV1636
23Papillary serous adenocarcinoma2CRO56IIIC
24Papillary serous adenocarcinoma1CRO81IIIC
25Papillary serous adenocarcinoma2CRO56IIIC260
26papillary serous adenocarcinoma3CRO35IV47
27Papillary serous adenocarcinoma2CRO53IIIA
28Papillary serous adenocarcinoma1CRS77IV>600
29Papillary serous adenocarcinoma2CRO65IIIC1118
30Papillary serous adenocarcinoma1CRS47IIIC712
31Papillary serous adenocarcinoma2CRS76IIIC1848
32Papillary serous adenocarcinoma2CRO70IIIC
33Papillary serous adenocarcinoma3CRS57IIIC266
34Adenocarcinoma metastatic2CRO57IIIC175
35Papillary serous adenocarcinoma1CRO65IV404
36Papillary serous adenocarcinoma2CRO76IV
37Papillary serous adenocarcinoma1CRO66IIIC
38Papillary serous adenocarcinoma1CRO68IIIC
39Papillary serous adenocarcinoma3IRO73IIIC
40Papillary serous adenocarcinoma2CRO63IV
41Papillary serous adenocarcinoma2IRS63IIIC
42Papillary serous adenocarcinoma2IRO47IIIC
43Papillary serous adenocarcinoma2CRO42IIIC110
44Papillary serous adenocarcinoma1CRO74IIIC4557
45Papillary serous adenocarcinoma2CRS64IIIC
46Papillary serous adenocarcinoma2IRS64IIIC456
47Papillary serous adenocarcinoma1IRO71IIIC
48Papillary serous adenocarcinoma6IRO69IIIC
49Papillary serous adenocarcinoma3CRO49IIIC
50Papillary serous adenocarcinoma2CRO62IV
51Papillary serous adenocarcinoma2IR
52Focal adenocarcinoma3CRS88IIIC
53Serous adenocarcinoma1IRO74IIIC101
54Papillary serous adenocarcinoma2IRO71IIIC
55Papillary serous adenocarcinoma4IRO69IIIC1606
56Papillary serous adenocarcinoma1IRO52IIIC
57Papillary serous adenocarcinoma2CRO67IIIC
58Papillary serous adenocarcinoma1CRO66IIIC824
59Papillary Serous adenocarcinoma2IRO52IIIC
60Papillary serous adenocarcinoma1CRS73IIIC2354
61Papillary serous adenocarcinoma0CRO75IIIC
62Papillary serous adenocarcinoma2IRO65IIIC
63Focal cellular atypian/aCRO74IIIC
64Papillary serous adenocarcinoma2IRO79IIIC417
65Papillary serous adenocarcinoma1CRO73IIIC180
66Papillary serous adenocarcinoma2IRO53IV96
67Papillary serous adenocarcinoma1CRO60IIIC
68Papillary serous adenocarcinoma2CR
69Papillary serous adenocarcinoma2IRS53IIIC
70Papillary serous adenocarcinoma1IRO41IIIC2800
71Papillary serous adenocarcinoma1CRO80IIIC
72Papillary serous adenocarcinoma2CRO42IIIA
73Papillary serous adenocarcinoma2IRS66IIIC90
74Papillary serous adenocarcinoma1CRS60IIIC750
75Papillary serous adenocarcinoma4CRO77IIIC9814
76Papillary serous adenocarcinoma0CRO72III
77Papillary serous adenocarcinoma1IRO66IIIC
78Papillary serous adenocarcinoma0CRO54III
79Papillary serous adenocarcinoma3CRO38IIIC

[i] The c-Met expression score was determined by intensity × cellularity, where intensity was graded as 1, weak; 2, moderate; or 3, strong, and cellularity was graded as 1 when ≤33%, 2 when 34–65%, or 3 when ≥66%. CR, complete response; IR, incomplete response to primary therapy.

Table II

c-Met expression in ovarian cancer cell lines.

Table II

c-Met expression in ovarian cancer cell lines.

Cell lineCellularityIntensityScore
A2008224
A2780CP000
A2780S111
BGI000
C13313
CAOV3313
CHI000
CHI cisR111
CAOV2326
Dov 13326
HeyA8313
IGR-OV1326
IMCC3212
IMCC5111
M41111
M41CSR212
MCAS313
OV2008111
OV90111
Ovary1847111
OVCA 429212
OVCA 432Acellularn/an/a
OVCA 433313
OVCA420122
OVCAR10000
OVCAR2326
OVCAR3212
OVCAR4326
OVCAR5326
OVCAR8313
PEO1326
PEO4224
SKOV8212
SKOV3326
SKOV4000
SKOV6212
T8326
Tov-112D212
Tov-21-G111
Tyknu000
Tyknu CisR000

[i] The c-Met expression score was determined by intensity × cellularity, where intensity was graded as 1, weak; 2, moderate; or 3, strong, and cellularity was graded as 1 when ≤33%, 2 when 34–65% or 3 when ≥66%.

Inhibition of HGF/c-Met signaling has in vitro anti-proliferative effects on OVCA cells and synergizes with combination carboplatin plus paclitaxel

To determine whether HGF signaling influenced OVCA cell line chemosensitivity, we evaluated the activity of MK8033, an inhibitor of HGF/c-Met signaling. MK8033 exhibited strong anti-proliferative effects against a panel of OVCA cell lines (Fig. 1). Furthermore, when combined in constant molar ratio of 1 with carboplatin plus paclitaxel (molar ratio of 20,000:1), MK8033 significantly lowered the carboplatin-paclitaxel IC50 in the majority of cell lines tested (Fig. 1). Combination index values calculated using the Chou-Talalay isobologram equation indicated synergistic activity between MK8033 and carboplatin-paclitaxel for almost all OVCA cell lines tested, as indicated by combination index values of <0.7 (Fig. 1).

Molecular determinants of MK8033-carboplatin-paclitaxel sensitivity influence overall survival from OVCA

Pearson's correlation test of IC50 values and Affymetrix HuRSTA genechip expression data identified 47 genes to be correlated with MK8033 plus carboplatin-paclitaxel sensitivity [false discovery rate (FDR) of <0.2] (Table III). PCA modeling of these genes indicated that the expression signature was associated with overall survival from OVCA (AUS dataset, n=218, P=0.013) (Fig. 2).

Table III

Genes associated with ovarian cancer cell sensitivity to MK8033 plus carboplatin-paclitaxel.

Table III

Genes associated with ovarian cancer cell sensitivity to MK8033 plus carboplatin-paclitaxel.

Signature gene symbols
SETD6UBQLN1HDAC3LOC653441
RNF207NDOR1CLTCL1PHC1
ABCC8IGHMPPIL4MATR3
DNASE1L1FBXO4RESTCLYBL
ACDPRKAA1LPHN3MLLT4
NUP155RIPK4DKFZp667EFCBP2
FAM91A2TASP1ACTN2KIAA0528
FAM91A1ADRBK2OSBPL6PPM1J
MGC5566FAM46CSTARTMEFF1
MRPL30ITIH4FUNDC2SGK269
MAGIXC1orf164RAB15LOC389634
HFEIL17RDEDA

Discussion

HGF is a paracrine growth factor originally found to be associated with liver regeneration (1618). HGF signaling orchestrates a multitude of biological functions during embryogenesis, including cellular migration, invasion, proliferation, angiogenesis, wound healing, and tissue repair (1929), and has been further associated with the development or progression of several different types of cancer, including OVCA (6,7,12,3045). In OVCA, increased levels of HGF or overexpression of the HGF receptor c-Met has been associated with poor clinical outcome (57,1618).

We found a near ubiquitous expression of c-Met in both primary OVCA and OVCA cell lines. To better understand the importance of HGF signaling in OVCA, we evaluated the activity of the novel inhibitor of HGF/c-Met signaling, MK8033, alone and in combination with carboplatin plus paclitaxel in a panel of OVCA cell lines (n=8). Although MK8033 showed anti-proliferative effects as a single agent, when used in a constant ratio to carboplatin-paclitaxel, the isobologram analysis indicated synergistic activity in the inhibition of OVCA cell proliferation. Pearson's correlation of cell line gene expression data and IC50 values identified the expression of 47 genes to be correlated with sensitivity to MK8033 plus carboplatin-paclitaxel. As shown by PCA modeling, genes correlated with MK8033 plus carboplatin-paclitaxel response were associated with overall survival from OVCA, suggesting that these genes may have a biological influence on OVCA response to therapy. Further analysis of the MK8033 plus carboplatin-paclitaxel response signature identified 6 of 47 genes to be significantly correlated with OVCA survival, including i) HFE (+ correlation, P=0.02); ii) UBQLN1 (+ correlation, P=0.04); iii) CLTCL1 (- correlation, P=0.04); iv) LPHN3 (- correlation, P=0.04); v) OSBPL6 (- correlation, P=0.03); and vi) SGK269 (- correlation, P=0.04).

The products of several of these genes have previously been implicated in the development and/or progression of cancer. For example, the product of the HFE gene, hemochromatosis protein, is involved in the regulation of iron absorption (46). Mutations in HFE and deregulation of iron absorption have been linked to liver cirrhosis and colon cancer risk (47,48). Ubiquilin 1, encoded by the UBQLN1 gene, has been shown to be associated with lung adenocarcinoma (49) and may affect OVCA response to metallodrugs (50). CLTCL1 encodes the heavy chain of clathrin, and the expression and/or activity of clathrin has been found to be associated with bladder cancer (51), TRAIL-resistant breast cancer (52), and uptake of some chemotherapeutic agents (53). Furthermore, the oxysterol binding protein (OSBP)-related protein 6, encoded by the OSBPL6 gene, belongs to a family of proteins recently identified as receptors for several natural products, including cephalostatin-1, ritterazine B, schweinfurthin A and OSW-1 (54).

In summary, we showed that targeted inhibition of HGF/c-Met signaling using the c-Met specific inhibitor, MK8033, worked synergistically with combinations of carboplatin plus paclitaxel to induce OVCA cell growth arrest. Furthermore, we demonstrated that genes associated with response to MK8033 plus carboplatin-paclitaxel may influence overall survival from OVCA, based on PCA modeling. The results of this study suggest that the inhibition of HGF/c-Met signaling may be a beneficial addition to the OVCA standard of care regimen of carboplatin plus paclitaxel therapy.

Acknowledgements

The authors thank Rasa Hamilton (Moffitt Cancer Center) for editorial assistance. We would also like to thank Merck Pharmaceuticals for their contributions. This study was supported in part by the Hearing the Ovarian Cancer Whisper, Jacquie Liggett Foundation, the Ocala Royal Dames for Cancer Research Inc., and the Phi Beta Psi Sorority.

References

1 

Baker VV: Salvage therapy for recurrent epithelial ovarian cancer. Hematol Oncol Clin North Am. 17:977–988. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Omura GA, Brady MF, Homesley HD, et al: Long-term follow-up and prognostic factor analysis in advanced ovarian carcinoma: the Gynecologic Oncology Group experience. J Clin Oncol. 9:1138–1150. 1991.PubMed/NCBI

3 

Di Renzo MF, Narsimhan RP, Olivero M, et al: Expression of the Met/HGF receptor in normal and neoplastic human tissues. Oncogene. 6:1997–2003. 1991.

4 

Huntsman D, Resau JH, Klineberg E and Auersperg N: Comparison of c-met expression in ovarian epithelial tumors and normal epithelia of the female reproductive tract by quantitative laser scan microscopy. Am J Pathol. 155:343–348. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Aune G, Lian AM, Tingulstad S, et al: Increased circulating hepatocyte growth factor (HGF): a marker of epithelial ovarian cancer and an indicator of poor prognosis. Gynecol Oncol. 121:402–406. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Goode EL, Chenevix-Trench G, Hartmann LC, et al: Assessment of hepatocyte growth factor in ovarian cancer mortality. Cancer Epidemiol Biomarkers Prev. 20:1638–1648. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Sawada K, Radjabi AR, Shinomiya N, et al: c-Met overexpression is a prognostic factor in ovarian cancer and an effective target for inhibition of peritoneal dissemination and invasion. Cancer Res. 67:1670–1679. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Yamamoto S, Tsuda H, Miyai K, Takano M, Tamai S and Matsubara O: Gene amplification and protein overexpression of MET are common events in ovarian clear-cell adenocarcinoma: their roles in tumor progression and prognostication of the patient. Mod Pathol. 24:1146–1155. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Yamamoto S, Tsuda H, Miyai K, Takano M, Tamai S and Matsubara O: Accumulative copy number increase of MET drives tumor development and histological progression in a subset of ovarian clear-cell adenocarcinomas. Mod Pathol. 25:122–130. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Basha R, Ingersoll SB, Sankpal UT, et al: Tolfenamic acid inhibits ovarian cancer cell growth and decreases the expression of c-Met and survivin through suppressing specificity protein transcription factors. Gynecol Oncol. 122:163–170. 2011. View Article : Google Scholar

11 

Zillhardt M, Christensen JG and Lengyel E: An orally available small-molecule inhibitor of c-Met, PF-2341066, reduces tumor burden and metastasis in a preclinical model of ovarian cancer metastasis. Neoplasia. 12:1–10. 2010.

12 

Zillhardt M, Park SM, Romero IL, et al: Foretinib (GSK1363089), an orally available multikinase inhibitor of c-Met and VEGFR-2, blocks proliferation, induces anoikis, and impairs ovarian cancer metastasis. Clin Cancer Res. 17:4042–4051. 2011. View Article : Google Scholar

13 

Marchion DC, Cottrill HM, Xiong Y, et al: BAD phosphorylation determines ovarian cancer chemosensitivity and patient survival. Clin Cancer Res. 17:6356–6366. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Bicaku E, Xiong Y, Marchion DC, et al: In vitro analysis of ovarian cancer response to cisplatin, carboplatin, and paclitaxel identifies common pathways that are also associated with overall patient survival. Br J Cancer. 106:1967–1975. 2012. View Article : Google Scholar

15 

Jolliffe IT: Principal Component Analysis. 2nd edition. Springer-Verlag; New York: 2002

16 

Nakamura T, Nawa K and Ichihara A: Partial purification and characterization of hepatocyte growth factor from serum of hepatectomized rats. Biochem Biophys Res Commun. 122:1450–1459. 1984. View Article : Google Scholar : PubMed/NCBI

17 

Nakamura T, Teramoto H and Ichihara A: Purification and characterization of a growth factor from rat platelets for mature parenchymal hepatocytes in primary cultures. Proc Natl Acad Sci USA. 83:6489–6493. 1986. View Article : Google Scholar : PubMed/NCBI

18 

Nakamura T, Nawa K, Ichihara A, Kaise N and Nishino T: Purification and subunit structure of hepatocyte growth factor from rat platelets. FEBS Lett. 224:311–316. 1987. View Article : Google Scholar : PubMed/NCBI

19 

Jeffers M, Rao MS, Rulong S, et al: Hepatocyte growth factor/scatter factor-Met signaling induces proliferation, migration, and morphogenesis of pancreatic oval cells. Cell Growth Differ. 7:1805–1813. 1996.PubMed/NCBI

20 

Montesano R, Matsumoto K, Nakamura T and Orci L: Identification of a fibroblast-derived epithelial morphogen as hepatocyte growth factor. Cell. 67:901–908. 1991. View Article : Google Scholar : PubMed/NCBI

21 

Weidner KM, Di Cesare S, Sachs M, Brinkmann V, Behrens J and Birchmeier W: Interaction between Gab1 and the c-Met receptor tyrosine kinase is responsible for epithelial morphogenesis. Nature. 384:173–176. 1996. View Article : Google Scholar : PubMed/NCBI

22 

Bladt F, Riethmacher D, Isenmann S, Aguzzi A and Birchmeier C: Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud. Nature. 376:768–771. 1995. View Article : Google Scholar : PubMed/NCBI

23 

Grant DS, Kleinman HK, Goldberg ID, et al: Scatter factor induces blood vessel formation in vivo. Proc Natl Acad Sci USA. 90:1937–1941. 1993. View Article : Google Scholar : PubMed/NCBI

24 

Trusolino L and Comoglio PM: Scatter-factor and semaphorin receptors: cell signalling for invasive growth. Nat Rev Cancer. 2:289–300. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Michalopoulos GK and DeFrances MC: Liver regeneration. Science. 276:60–66. 1997. View Article : Google Scholar

26 

Sonnenberg E, Meyer D, Weidner KM and Birchmeier C: Scatter factor/hepatocyte growth factor and its receptor, the c-met tyrosine kinase, can mediate a signal exchange between mesenchyme and epithelia during mouse development. J Cell Biol. 123:223–235. 1993. View Article : Google Scholar

27 

Mizuno S and Nakamura T: Hepatocyte growth factor: a regenerative drug for acute hepatitis and liver cirrhosis. Regen Med. 2:161–170. 2007. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

You WK and McDonald DM: The hepatocyte growth factor/c-Met signaling pathway as a therapeutic target to inhibit angiogenesis. BMB Rep. 41:833–839. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Sattler M, Reddy MM, Hasina R, Gangadhar T and Salgia R: The role of the c-Met pathway in lung cancer and the potential for targeted therapy. Ther Adv Med Oncol. 3:171–184. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Gumustekin M, Kargi A, Bulut G, et al: HGF/c-Met overexpressions, but not met mutation, correlates with progression of non-small cell lung cancer. Pathol Oncol Res. 18:209–218. 2012. View Article : Google Scholar : PubMed/NCBI

32 

El-Attar HA and Sheta MI: Hepatocyte growth factor profile with breast cancer. Indian J Pathol Microbiol. 54:509–513. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P and 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. 46:1679–1691. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Kim EJ, Eom SJ, Hong JE, Lee JY, Choi MS and Park JH: Benzyl isothiocyanate inhibits basal and hepatocyte growth factor-stimulated migration of breast cancer cells. Mol Cell Biochem. 369:431–440. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Derman MP, Chen JY, Spokes KC, Songyang Z and Cantley LG: An 11-amino acid sequence from c-met initiates epithelial chemotaxis via phosphatidylinositol 3-kinase and phospholipase C. J Biol Chem. 271:4251–4255. 1996. View Article : Google Scholar : PubMed/NCBI

36 

Li C, Wu JJ, Hynes M, et al: c-Met is a marker of pancreatic cancer stem cells and therapeutic target. Gastroenterology. 141:2218–2227. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Lee YJ, Kim DH, Lee SH, Kim DW, Nam HS and Cho MK: Expression of the c-Met proteins in malignant skin cancers. Ann Dermatol. 23:33–38. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Syed ZA, Yin W, Hughes K, Gill JN, Shi R and Clifford JL: HGF/c-met/Stat3 signaling during skin tumor cell invasion: indications for a positive feedback loop. BMC Cancer. 11:1802011. View Article : Google Scholar : PubMed/NCBI

39 

Yoshizawa Y, Yamada Y, Kanayama S, et al: Signaling pathway involved in cyclooxygenase-2 up-regulation by hepatocyte growth factor in endometrial cancer cells. Oncol Rep. 26:957–964. 2011.PubMed/NCBI

40 

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

41 

Osada S, Kanematsu M, Imai H and Goshima S: Clinical significance of serum HGF and c-Met expression in tumor tissue for evaluation of properties and treatment of hepatocellular carcinoma. Hepatogastroenterology. 55:544–549. 2008.PubMed/NCBI

42 

Eksioglu-Demiralp E, Akdeniz T and Bayik M: Aberrant expression of c-met and HGF/c-met pathway provides survival advantage in B-chronic lymphocytic leukemia. Cytometry B Clin Cytom. 80:1–7. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Que W and Chen J: Knockdown of c-Met inhibits cell proliferation and invasion and increases chemosensitivity to doxorubicin in human multiple myeloma U266 cells in vitro. Mol Med Rep. 4:343–349. 2011.PubMed/NCBI

44 

Inno A, Salvatore MD, Cenci T, et al: Is there a role for IGF1R and c-MET pathways in resistance to cetuximab in metastatic colorectal cancer? Clin Colorectal Cancer. 10:325–332. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Bu R, Uddin S, Bavi P, et al: HGF/c-Met pathway has a prominent role in mediating antiapoptotic signals through AKT in epithelial ovarian carcinoma. Lab Invest. 91:124–137. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Zhou XY, Tomatsu S, Fleming RE, et al: HFE gene knockout produces mouse model of hereditary hemochromatosis. Proc Natl Acad Sci USA. 95:2492–2497. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Xue X, Taylor M, Anderson E, et al: Hypoxia-inducible factor-2alpha activation promotes colorectal cancer progression by dysregulating iron homeostasis. Cancer Res. 72:2285–2293. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Ba Q, Hao M, Huang H, et al: Iron deprivation suppresses hepatocellular carcinoma growth in experimental studies. Clin Cancer Res. 17:7625–7633. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Zhang C and Saunders AJ: An emerging role for Ubiquilin 1 in regulating protein quality control system and in disease pathogenesis. Discov Med. 8:18–22. 2009.PubMed/NCBI

50 

Guidi F, Puglia M, Gabbiani C, et al: 2D-DIGE analysis of ovarian cancer cell responses to cytotoxic gold compounds. Mol Biosyst. 8:985–993. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Menashe I, Figueroa JD, Garcia-Closas M, et al: Large-scale pathway-based analysis of bladder cancer genome-wide association data from five studies of European background. PLoS One. 7:e293962012. View Article : Google Scholar : PubMed/NCBI

52 

Zhang Y and Zhang B: TRAIL resistance of breast cancer cells is associated with constitutive endocytosis of death receptors 4 and 5. Mol Cancer Res. 6:1861–1871. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Bolden SW, Hambley JW, Johnston GA and Rogers LJ: Neonatal stress and long-term modulation of GABA receptors in rat brain. Neurosci Lett. 111:258–262. 1990. View Article : Google Scholar : PubMed/NCBI

54 

Burgett AW, Poulsen TB, Wangkanont K, et al: Natural products reveal cancer cell dependence on oxysterol-binding proteins. Nat Chem Biol. 7:639–647. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May 2013
Volume 29 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Marchion DC, Bicaku E, Xiong Y, Bou Zgheib N, Al Sawah E, Stickles XB, Judson PL, Lopez AS, Cubitt CL, Gonzalez-Bosquet J, Gonzalez-Bosquet J, et al: A novel c-Met inhibitor, MK8033, synergizes with carboplatin plus paclitaxel to inhibit ovarian cancer cell growth. Oncol Rep 29: 2011-2018, 2013
APA
Marchion, D.C., Bicaku, E., Xiong, Y., Bou Zgheib, N., Al Sawah, E., Stickles, X.B. ... Lancaster, J.M. (2013). A novel c-Met inhibitor, MK8033, synergizes with carboplatin plus paclitaxel to inhibit ovarian cancer cell growth. Oncology Reports, 29, 2011-2018. https://doi.org/10.3892/or.2013.2329
MLA
Marchion, D. C., Bicaku, E., Xiong, Y., Bou Zgheib, N., Al Sawah, E., Stickles, X. B., Judson, P. L., Lopez, A. S., Cubitt, C. L., Gonzalez-Bosquet, J., Wenham, R. M., Apte, S. M., Berglund, A., Lancaster, J. M."A novel c-Met inhibitor, MK8033, synergizes with carboplatin plus paclitaxel to inhibit ovarian cancer cell growth". Oncology Reports 29.5 (2013): 2011-2018.
Chicago
Marchion, D. C., Bicaku, E., Xiong, Y., Bou Zgheib, N., Al Sawah, E., Stickles, X. B., Judson, P. L., Lopez, A. S., Cubitt, C. L., Gonzalez-Bosquet, J., Wenham, R. M., Apte, S. M., Berglund, A., Lancaster, J. M."A novel c-Met inhibitor, MK8033, synergizes with carboplatin plus paclitaxel to inhibit ovarian cancer cell growth". Oncology Reports 29, no. 5 (2013): 2011-2018. https://doi.org/10.3892/or.2013.2329