59 results on '"Proto-Oncogene Proteins c-met drug effects"'
Search Results
2. The novel role of circular RNA ST3GAL6 on blocking gastric cancer malignant behaviours through autophagy regulated by the FOXP2/MET/mTOR axis.
- Author
-
Xu P, Zhang X, Cao J, Yang J, Chen Z, Wang W, Wang S, Zhang L, Xie L, Fang L, Xia Y, Xuan Z, Lv J, Xu H, and Xu Z
- Subjects
- Animals, Cell Movement drug effects, Cell Movement genetics, Cell Proliferation drug effects, Cell Proliferation genetics, Disease Models, Animal, Forkhead Transcription Factors drug effects, Mice, Neoplasms drug therapy, Neoplasms prevention & control, Proto-Oncogene Proteins c-met drug effects, Sialyltransferases pharmacology, Signal Transduction drug effects, Signal Transduction genetics, Stomach Neoplasms prevention & control, TOR Serine-Threonine Kinases drug effects, beta-Galactoside alpha-2,3-Sialyltransferase, Autophagy drug effects, Sialyltransferases metabolism, Stomach Neoplasms drug therapy
- Abstract
Gastric cancer (GC) ranks third in mortality among all cancers worldwide. Circular RNAs (circRNAs) play an important role in the occurrence and development of gastric cancer. Forkhead box P2 (FOXP2), as a transcription factor, is closely associated with the development of many types of tumours. However, the regulatory network between FOXP2 and circRNAs remains to be explored. In our study, circST3GAL6 was significantly downregulated in GC and was associated with poor prognosis in GC patients. Overexpression of circST3GAL6 inhibited the malignant behaviours of GC cells, which was mediated by inducing apoptosis and autophagy. In addition, we demonstrated that circST3GAL6 regulated FOXP2 through the mir-300 sponge. We further found that FOXP2 inhibited MET Proto-Oncogene (MET), which was the initiating factor that regulated the classic AKT/mTOR pathway of autophagy. In conclusion, our results suggested that circST3GAL6 played a tumour suppressive role in gastric cancer through miR-300/FOXP2 axis and regulated apoptosis and autophagy through FOXP2-mediated transcriptional inhibition of the MET axis, which may become a potential target for GC therapy., (© 2022 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.)
- Published
- 2022
- Full Text
- View/download PDF
3. Phase I Study of 2- or 3-Week Dosing of Telisotuzumab Vedotin, an Antibody-Drug Conjugate Targeting c-Met, Monotherapy in Patients with Advanced Non-Small Cell Lung Carcinoma.
- Author
-
Camidge DR, Morgensztern D, Heist RS, Barve M, Vokes E, Goldman JW, Hong DS, Bauer TM, Strickler JH, Angevin E, Motwani M, Parikh A, Sun Z, Bach BA, Wu J, Komarnitsky PB, and Kelly K
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal pharmacology, Carcinoma, Non-Small-Cell Lung pathology, Female, Humans, Immunoconjugates pharmacology, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Staging, Proto-Oncogene Proteins c-met drug effects, Time Factors, Antibodies, Monoclonal administration & dosage, Carcinoma, Non-Small-Cell Lung drug therapy, Immunoconjugates administration & dosage, Lung Neoplasms drug therapy
- Abstract
Purpose: Telisotuzumab vedotin (Teliso-V) is an anti-c-Met-directed antibody-drug conjugate. Here, we present safety and efficacy data from a phase I/Ib study of Teliso-V monotherapy evaluated in once every 2 weeks/once every 3 weeks schedules in patients with non-small cell lung cancer (NSCLC)., Patients and Methods: During dose escalation, patients received Teliso-V monotherapy intravenously once every 3 weeks (0.15-3.3 mg/kg) or once every 2 weeks (1.6-2.2 mg/kg). The dose-expansion phase enrolled patients with NSCLC and c-Met H -score ≥150 (c-Met+) or MET amplification/exon 14 skipping mutations. Safety, pharmacokinetics, and efficacy were assessed. Herein, the analysis of patients receiving ≥1.6 mg/kg once every 2 weeks or ≥2.4 mg/kg once every 3 weeks Teliso-V is reported., Results: Fifty-two patients with NSCLC were enrolled and received ≥1.6 mg/kg Teliso-V once every 2 weeks ( n = 28) or ≥2.4 mg/kg Teliso-V once every 3 weeks ( n = 24). The most common adverse events were fatigue (54%), peripheral neuropathy (42%), and nausea (38%). No dose-limiting toxicities were observed for Teliso-V once every 2 weeks and once every 3 weeks up to 2.2 and 2.7 mg/kg, respectively. The recommended phase II dose was established at 1.9 mg/kg once every 2 weeks and 2.7 mg/kg once every 3 weeks on the basis of overall safety and pharmacokinetics. Forty of 52 patients were c-Met+ (33 nonsquamous, 6 squamous, 1 mixed histology) and were included in the efficacy-evaluable population. Of those, 9 (23%) had objective responses with median duration of response of 8.7 months; median progression-free survival was 5.2 months., Conclusions: Teliso-V monotherapy was tolerated and showed antitumor activity in c-Met+ NSCLC. On the basis of overall safety, pharmacokinetics, and efficacy outcomes, 1.9 mg/kg Teliso-V once every 2 weeks and 2.7 mg/kg once every 3 weeks schedules were selected for further clinical development., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
4. A comparison of sunitinib with cabozantinib, crizotinib, and savolitinib for treatment of advanced papillary renal cell carcinoma: a randomised, open-label, phase 2 trial.
- Author
-
Pal SK, Tangen C, Thompson IM Jr, Balzer-Haas N, George DJ, Heng DYC, Shuch B, Stein M, Tretiakova M, Humphrey P, Adeniran A, Narayan V, Bjarnason GA, Vaishampayan U, Alva A, Zhang T, Cole S, Plets M, Wright J, and Lara PN Jr
- Subjects
- Aged, Anilides adverse effects, Canada, Carcinoma, Renal Cell mortality, Crizotinib administration & dosage, Crizotinib adverse effects, Female, Humans, Kidney Neoplasms mortality, Male, Middle Aged, Progression-Free Survival, Protein Kinase Inhibitors adverse effects, Proto-Oncogene Proteins c-met drug effects, Pyrazines administration & dosage, Pyrazines adverse effects, Pyridines adverse effects, Sunitinib adverse effects, Triazines administration & dosage, Triazines adverse effects, United States, Anilides administration & dosage, Carcinoma, Renal Cell drug therapy, Kidney Neoplasms drug therapy, Protein Kinase Inhibitors administration & dosage, Pyridines administration & dosage, Sunitinib administration & dosage
- Abstract
Background: MET (also known as hepatocyte growth factor receptor) signalling is a key driver of papillary renal cell carcinoma (PRCC). Given that no optimal therapy for metastatic PRCC exists, we aimed to compare an existing standard of care, sunitinib, with the MET kinase inhibitors cabozantinib, crizotinib, and savolitinib for treatment of patients with PRCC., Methods: We did a randomised, open-label, phase 2 trial done in 65 centres in the USA and Canada. Eligible patients were aged 18 years or older with metastatic PRCC who had received up to one previous therapy (excluding vascular endothelial growth factor-directed and MET-directed agents). Patients were randomly assigned to receive sunitinib, cabozantinib, crizotinib, or savolitinib, with stratification by receipt of previous therapy and PRCC subtype. All drug doses were administered orally: sunitinib 50 mg, 4 weeks on and 2 weeks off (dose reductions to 37·5 mg and 25 mg allowed); cabozantinib 60 mg daily (reductions to 40 mg and 20 mg allowed); crizotinib 250 mg twice daily (reductions to 200 mg twice daily and 250 mg once daily allowed); and savolitinib 600 mg daily (reductions to 400 mg and 200 mg allowed). Progression-free survival (PFS) was the primary endpoint. Analyses were done in an intention-to-treat population, with patients who did not receive protocol therapy excluded from safety analyses. This trial is registered with ClinicalTrials.gov, NCT02761057., Findings: Between April 5, 2016, and Dec 15, 2019, 152 patients were randomly assigned to one of four study groups. Five patients were identified as ineligible post-randomisation and were excluded from these analyses, resulting in 147 eligible patients. Assignment to the savolitinib (29 patients) and crizotinib (28 patients) groups was halted after a prespecified futility analysis; planned accrual was completed for both sunitinib (46 patients) and cabozantinib (44 patients) groups. PFS was longer in patients in the cabozantinib group (median 9·0 months, 95% CI 6-12) than in the sunitinib group (5·6 months, 3-7; hazard ratio for progression or death 0·60, 0·37-0·97, one-sided p=0·019). Response rate for cabozantinib was 23% versus 4% for sunitinib (two-sided p=0·010). Savolitinib and crizotinib did not improve PFS compared with sunitinib. Grade 3 or 4 adverse events occurred in 31 (69%) of 45 patients receiving sunitinib, 32 (74%) of 43 receiving cabozantinib, ten (37%) of 27 receiving crizotinib, and 11 (39%) of 28 receiving savolitinib; one grade 5 thromboembolic event was recorded in the cabozantinib group., Interpretation: Cabozantinib treatment resulted in significantly longer PFS compared with sunitinib in patients with metastatic PRCC., Funding: National Institutes of Health and National Cancer Institute., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
5. Dual-function chimeric antigen receptor T cells targeting c-Met and PD-1 exhibit potent anti-tumor efficacy in solid tumors.
- Author
-
Yuan X, Sun Z, Yuan Q, Hou W, Liang Q, Wang Y, Mo W, Wang H, and Yu M
- Subjects
- Animals, B7-H1 Antigen metabolism, Cell Line, Tumor, Cell Survival drug effects, Cells, Cultured, Cytokines drug effects, Enzyme-Linked Immunosorbent Assay, Humans, Mice, Mice, Inbred NOD, Tumor Microenvironment drug effects, Xenograft Model Antitumor Assays, Immunotherapy, Adoptive methods, Neoplasms pathology, Programmed Cell Death 1 Receptor drug effects, Proto-Oncogene Proteins c-met drug effects, Receptors, Chimeric Antigen administration & dosage
- Abstract
Purpose Programmed cell death 1 (PD-1), which is upregulated under the continuous induction of the tumor microenvironment, causes chimeric antigen receptor (CAR)-T cell hypofunction via interaction with programmed death ligand 1 (PD-L1). This study aimed to construct CAR-T cells that are resistant to PD-1 inhibition to improve the effect of CAR-T cells in solid tumors. Methods We constructed a type of dual-function CAR-T cell that targets tumor-associated antigen c-Met and blocks the binding of PD-1 with PD-L1. The expression of c-Met, PD-L1, and inhibitory receptors was measured using flow cytometry. The cytotoxicity, cytokine release, and differentiation level of CAR-T cells were determined using lactate dehydrogenase release assay, enzyme-linked immunosorbent assay, and flow cytometry, respectively. The levels of p-Akt, p-MAPK, caspase-3, and Bcl2 were detected by western blot. The in vivo anti-tumor effect was evaluated using tumor xenograft models. Results Dual-function CAR-T cells could mediate enhanced active signals upon encountering target antigens and had targeted cytotoxicity to target cells. However, the cytotoxicity of c-Met-CAR-PD-1
+ T cells was impaired due to the interaction of PD-1 with PD-L1. By blocking the binding of PD-1 and PD-L1, the novel dual-function CAR-PD-1+ T cells could maintain cytotoxicity to PD-L1+ tumor cells. In tumor tissue, the dual-function CAR-T cells showed lower inhibitory receptor expression and lower differentiation characteristics, which resulted in potent anti-tumor effects and prolonged survival in PD-L1+ tumor xenograft models compared to single-target CAR-T cells. Conclusion These results confirm that the novel dual-function CAR-T cells exhibit stronger anti-tumor activity against solid tumors than traditional single-target CAR-T cells and present a new approach that enhance the activity of CAR-T cells in solid tumors.- Published
- 2021
- Full Text
- View/download PDF
6. Crizotinib induced antitumor activity and synergized with chemotherapy and hormonal drugs in breast cancer cells via downregulating MET and estrogen receptor levels.
- Author
-
Ayoub NM, Ibrahim DR, Alkhalifa AE, and Al-Husein BA
- Subjects
- Antineoplastic Agents administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Cell Line, Tumor, Cell Survival drug effects, Crizotinib administration & dosage, Dose-Response Relationship, Drug, Drug Synergism, Estrogen Antagonists pharmacology, Humans, Inhibitory Concentration 50, Lapatinib pharmacology, Receptor, ErbB-2 drug effects, Antineoplastic Agents pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Breast Neoplasms pathology, Crizotinib pharmacology, Proto-Oncogene Proteins c-met drug effects, Receptors, Estrogen drug effects
- Abstract
MET is a receptor tyrosine kinase known to drive neoplastic transformation and aggressive tumor phenotypes. Crizotinib is an oral multi-targeted tyrosine kinase inhibitor of MET, ALK, RON, and ROS1 kinases. In this study, the anticancer effects of crizotinib on breast cancer cells were investigated in vitro along with the molecular mechanisms associated with these effects. Besides, the antiproliferative effects of crizotinib in combination with chemotherapy, hormonal drugs, and targeted agents were examined. Results showed that crizotinib produced dose-dependent antiproliferative effects in BT-474 and SK-BR-3 breast cancer cells with IC
50 values of 1.7 μM and 5.2 μM, respectively. Crizotinib inhibited colony formation of BT-474 cells at low micromolar concentrations (1-5 μM). Immunofluorescence and Western blotting indicated that crizotinib reduced total levels of MET and estrogen receptor (ERα) in BT-474 cells. Also, crizotinib reduced the levels of phosphorylated (active) MET and HER2 in BT-474 cells. The combined treatment of crizotinib with doxorubicin and paclitaxel resulted in synergistic growth inhibition of BT-474 cells with combination index values of 0.46 and 0.35, respectively. Synergy was also observed with the combination of crizotinib with the hormonal drugs 4-hydroxytamoxifen and fulvestrant in BT-474 cells. Alternatively, the combination of crizotinib with lapatinib produced antagonistic antiproliferative effects in both BT-474 and SK-BR-3 cells. Collectively, these findings demonstrate the anticancer effects of crizotinib in breast cancer cells and reveal ERα as a potential therapeutic target of the drug apart from its classical kinase inhibitory activity. Crizotinib could be an appealing option in combination with chemotherapy or hormonal drugs for the management of breast cancer.- Published
- 2021
- Full Text
- View/download PDF
7. Diverse Receptor Tyrosine Kinase Phosphorylation in Urine-Derived Tubular Epithelial Cells from Autosomal Dominant Polycystic Kidney Disease Patients.
- Author
-
Ikeda K, Kusaba T, Tomita A, Watanabe-Uehara N, Ida T, Kitani T, Yamashita N, Uehara M, Matoba S, Yamada T, and Tamagaki K
- Subjects
- Adult, Aged, Aminopyridines pharmacology, Animals, Cell Line, Cell Proliferation drug effects, Cells, Cultured, Cysts, Dogs, Epithelial Cells enzymology, Epithelial Cells metabolism, Female, Humans, Kidney physiopathology, Madin Darby Canine Kidney Cells, Male, Middle Aged, Phosphorylation, Piperazines pharmacology, Polycystic Kidney, Autosomal Dominant metabolism, Proto-Oncogene Proteins c-met antagonists & inhibitors, Proto-Oncogene Proteins c-met drug effects, Kidney Tubules, Distal metabolism, Polycystic Kidney, Autosomal Dominant enzymology, Proto-Oncogene Proteins c-met metabolism, Receptor Protein-Tyrosine Kinases metabolism, Urine cytology
- Abstract
Backgrounds: The clinical features of autosomal dominant polycystic kidney disease (ADPKD) differ among patients even if they have the same gene mutation in PKD1 or PKD2. This suggests that there is diversity in the expression of other modifier genes or in the underlying molecular mechanisms of ADPKD, but these are not well understood., Methods: We primarily cultured solute carrier family 12 member 3 (SLC12A3)-positive urine-derived distal tubular epithelial cells from 6 ADPKD patients and 4 healthy volunteers and established immortalized cell lines. The diversity in receptor tyrosine kinase (RTK) phosphorylation by phospho-RTK array in immortalized tubular epithelial cells was analyzed., Results: We noted diversity in the activation of several molecules, including Met, a receptor of hepatocyte growth factor (HGF). Administration of golvatinib, a selective Met inhibitor, or transfection of small interfering RNA for Met suppressed cell proliferation and downstream signaling only in the cell lines in which hyperphosphorylation of Met was observed. In three-dimensional culture of Madin-Darby canine kidney (MDCK) cells as a cyst formation model of ADPKD, HGF activated Met, resulting in an increased total cyst number and total cyst volume. Administration of golvatinib inhibited these phenotypes in MDCK cells., Conclusion: Analysis of urine-derived tubular epithelial cells demonstrated diverse RTK phosphorylation in ADPKD, and Met phosphorylation was noted in some patients. Considering the difference in the effects of golvatinib on immortalized tubular epithelial cells among patients, this analysis may aid in selecting suitable drugs for individual ADPKD patients., (© 2020 S. Karger AG, Basel.)
- Published
- 2020
- Full Text
- View/download PDF
8. Crizotinib and erlotinib inhibits growth of c-Met + /EGFRvIII + primary human glioblastoma xenografts.
- Author
-
Goodwin CR, Rath P, Oyinlade O, Lopez H, Mughal S, Xia S, Li Y, Kaur H, Zhou X, Ahmed AK, Ho S, Olivi A, and Lal B
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Brain Neoplasms drug therapy, Brain Neoplasms pathology, ErbB Receptors drug effects, ErbB Receptors metabolism, Glioblastoma pathology, Heterografts drug effects, Humans, Mice, Nude, Neoplasm Recurrence, Local drug therapy, Crizotinib pharmacology, Erlotinib Hydrochloride pharmacology, Glioblastoma drug therapy, Proto-Oncogene Proteins c-met drug effects
- Abstract
Objectives: Receptor tyrosine kinases (RTK), such as c-Met and epidermal growth factor receptor (EGFR), are implicated in the malignant progression of glioblastoma. Studies show that RTK systems can co-modulate distinct and overlapping oncogenic downstream signaling pathways. EGFRvIII, a constitutively activated EGFR deletion mutant variant, leads to increased tumor growth and diminishes the tumor growth response to HGF: c-Met pathway inhibitor therapy. Conversely, activation of the c-Met pathway diminishes the tumor growth response to EGFR pathway inhibitors. Previously we reported that EGFRvIII and c-Met pathway inhibitors synergize to inhibit tumor growth in isogenic GBM cell lines engineered to express EGFRvIII. More recently, studies suggest that despite targeting RTK signaling in glioblastoma multiforme, a subpopulation of stem-like tumor-propagating cells can persist to replenish the tumor cell population leading to tumor recurrence., Patients and Methods: Mayo 39 and Mayo 59 xenograft lines were cultured and xenografts were maintained. Subcutaneous xenograft lines were serially passaged in nude mice to generate subcutaneous xenografts. Xenografts were implanted in 6-8 week old nude mice. Once tumors reached a substantial size (150 mm
3 ), mice were randomly divided into 4 groups: 1) control vehicle, 2) Crizotinib (crizo), 3) Erlotinib (erlot), or 4) Crizotinib + Erlotinib, (n = 5 per group)., Results: Crizotinib (c-Met pathway inhibitor) and Erlotinib (EGFR pathway inhibitor) in combination significantly inhibited tumor growth, phospho-EGFRvIII, phospho-Met, phospho-AKT, phospho-MAPK, and neurosphere growth in Mayo 39 and Mayo 59 primary GBM subcutaneous xenografts. The expression of the stem cell markers Nestin, Musashi, Olig 2 and Sox2 were also significantly down-regulated by c-Met inhibition, but no additive down-regulation was seen by co-treatment with Erlotinib., Conclusions: These results are consistent with and corroborate our previous findings demonstrating that targeting these two parallel pathways with c-Met and EGFR inhibitor therapy provides substantial anti-tumor activity in glioblastoma models., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
9. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): a randomised, double-blind, placebo-controlled, phase 3 trial.
- Author
-
Catenacci DVT, Tebbutt NC, Davidenko I, Murad AM, Al-Batran SE, Ilson DH, Tjulandin S, Gotovkin E, Karaszewska B, Bondarenko I, Tejani MA, Udrea AA, Tehfe M, De Vita F, Turkington C, Tang R, Ang A, Zhang Y, Hoang T, Sidhu R, and Cunningham D
- Subjects
- Adult, Aged, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Capecitabine administration & dosage, Capecitabine adverse effects, Cisplatin administration & dosage, Cisplatin adverse effects, Disease-Free Survival, Dose-Response Relationship, Drug, Double-Blind Method, Drug Administration Schedule, Epirubicin administration & dosage, Epirubicin adverse effects, Esophageal Neoplasms genetics, Esophageal Neoplasms pathology, Esophagogastric Junction pathology, Humans, Internationality, Kaplan-Meier Estimate, Middle Aged, Prognosis, Proportional Hazards Models, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Survival Analysis, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Esophageal Neoplasms drug therapy, Esophageal Neoplasms mortality, Stomach Neoplasms drug therapy, Stomach Neoplasms mortality
- Abstract
Background: Rilotumumab is a fully human monoclonal antibody that selectively targets the ligand of the MET receptor, hepatocyte growth factor (HGF). We aimed to assess the efficacy, safety, and pharmacokinetics of rilotumumab combined with epirubicin, cisplatin, and capecitabine, and to assess potential biomarkers, in patients with advanced MET-positive gastric or gastro-oesophageal junction adenocarcinoma., Methods: This multicentre, randomised, double-blind, placebo-controlled, phase 3 study was done at 152 centres in 27 countries. We recruited adults (aged ≥18 years) with unresectable locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, MET-positive tumours (≥25% of tumour cells with membrane staining of ≥1+ staining intensity), and evaluable disease, who had not received previous systemic therapy. Eligible patients were randomly assigned (1:1) via a computerised voice response system to receive rilotumumab 15 mg/kg intravenously or placebo in combination with open-label chemotherapy (epirubicin 50 mg/m
2 intravenously; cisplatin 60 mg/m2 intravenously; capecitabine 625 mg/m2 orally twice daily) in 21-day cycles for up to ten cycles. After completion of chemotherapy, patients continued to receive rilotumumab or placebo monotherapy until disease progression, intolerability, withdrawal of consent, or study termination. Randomisation was stratified by disease extent and ECOG performance status. Both patients and physicians were masked to study treatment assignment. The primary endpoint was overall survival, analysed by intention to treat. We report the final analysis. This study is registered with ClinicalTrials.gov, number NCT01697072., Findings: Between Nov 7, 2012, and Nov 21, 2014, 609 patients were randomly assigned to rilotumumab plus epirubicin, cisplatin, and capecitabine (rilotumumab group; n=304) or placebo plus epirubicin, cisplatin, and capecitabine (placebo group; n=305). Study treatment was stopped early after an independent data monitoring committee found a higher number of deaths in the rilotumumab group than in the placebo group; all patients in the rilotumumab group subsequently discontinued all study treatment. Median follow-up was 7·7 months (IQR 3·6-12·0) for patients in the rilotumumab group and 9·4 months (5·3-13·1) for patients in the placebo group. Median overall survival was 8·8 months (95% CI 7·7-10·2) in the rilotumumab group compared with 10·7 months (9·6-12·4) in the placebo group (stratified hazard ratio 1·34, 95% CI 1·10-1·63; p=0·003). The most common grade 3 or worse adverse events in the rilotumumab and placebo groups were neutropenia (86 [29%] of 298 patients vs 97 [32%] of 299 patients), anaemia (37 [12%] vs 43 [14%]), and fatigue (30 [10%] vs 35 [12%]). The frequency of serious adverse events was similar in the rilotumumab and placebo groups (142 [48%] vs 149 [50%]). More deaths due to adverse events occurred in the rilotumumab group than the placebo group (42 [14%] vs 31 [10%]). In the rilotumumab group, 33 (11%) of 298 patients had fatal adverse events due to disease progression, and nine (3%) had fatal events not due to disease progression. In the placebo group, 23 (8%) of 299 patients had fatal adverse events due to disease progression, and eight (3%) had fatal events not due to disease progression., Interpretation: Ligand-blocking inhibition of the MET pathway with rilotumumab is not effective in improving clinical outcomes in patients with MET-positive gastric or gastro-oesophageal adenocarcinoma., Funding: Amgen., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
10. A mini-review of c-Met as a potential therapeutic target in melanoma.
- Author
-
Al-U'datt DGF, Al-Husein BAA, and Qasaimeh GR
- Subjects
- Animals, Hepatocyte Growth Factor metabolism, Humans, Melanoma genetics, Proto-Oncogene Proteins c-met genetics, Signal Transduction drug effects, Antineoplastic Agents pharmacology, Melanoma drug therapy, Proto-Oncogene Proteins c-met drug effects
- Abstract
Melanoma is the third highest rated cancer in prevalence. Surgery, radiotherapy and targeted/biological therapies in addition to chemotherapy are available options for management of this cancer. Met is an appealing target for management of this type of cancer, since it targets many cancer vital processors, such as angiogenesis, cell growth, scattering and differentiation. In this review, we provide an overview about pathway abnormalities associated with melanoma. We also provide a summary about the events involved in Met signaling and related signaling molecules. We also show the evidence of the importance of Met signaling pathway as a target in cancer management. We also summarize clinical evidence about the use of Met signaling in management of cancer and summarize available trials related to targeting Met in other cancers., (Copyright © 2017 Elsevier Masson SAS. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
11. MET exon 14 skipping mutation in triple-negative pulmonary adenocarcinomas and pleomorphic carcinomas: An analysis of intratumoral MET status heterogeneity and clinicopathological characteristics.
- Author
-
Kwon D, Koh J, Kim S, Go H, Kim YA, Keam B, Kim TM, Kim DW, Jeon YK, and Chung DH
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma surgery, Adenocarcinoma of Lung, Adult, Aged, Aged, 80 and over, Anaplastic Lymphoma Kinase, Cell Line, Tumor, ErbB Receptors genetics, Exons, Female, Gene Amplification, Gene Dosage, Humans, Lung Neoplasms drug therapy, Lung Neoplasms surgery, Male, Middle Aged, Mutation, Neoadjuvant Therapy, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Proto-Oncogene Proteins p21(ras) genetics, Receptor Protein-Tyrosine Kinases genetics, Adenocarcinoma genetics, Adenocarcinoma pathology, Lung Neoplasms genetics, Lung Neoplasms pathology, Proto-Oncogene Proteins c-met genetics
- Abstract
Objectives: MET mutations leading to exon 14 skipping rarely occur in non-small cell lung cancer (NSCLC). Recently, small molecule inhibitors targeting MET mutations showed clinical benefit. However, the clinicopathological characteristics of NSCLC harboring MET mutations, and the correlation among mutations, protein expression, and gene copy number of MET in NSCLC remain unclear. Therefore, we address these issues., Materials and Methods: MET exon 14 skipping mutations were evaluated using real-time quantitative reverse-transcription-PCR (qRT-PCR) in 102 triple-negative (i.e., EGFR mutation (-)/ALK translocation (-)/KRAS mutation (-)) pulmonary adenocarcinomas, and 45 pleomorphic carcinomas. MET mutation and gene copy were also examined in microdissected tissues obtained from tumor areas with heterogeneous MET immunohistochemical expression., Results: MET mutations were detected in 8.8% (9/102) of triple-negative adenocarcinomas and 20% (9/45) of pleomorphic carcinomas of the lung. Patients with MET-mutated adenocarcinomas was significantly older than those without MET mutations (P=0.015). The male to female and ever-to never-smoker ratios were 3:6 and 2:7, respectively, among patients with MET-mutated adenocarcinomas. All (9/9) of the MET-mutated adenocarcinomas showed acinar predominant histology with associated lepidic patterns. In contrast, the male to female and ever- to never-smoker ratios were 8:1 and 7:1, respectively, among patients with MET-mutated pleomorphic carcinomas. The carcinoma component of MET-mutated pleomorphic carcinomas was mostly adenocarcinoma of acinar pattern (8/9). MET mutation was detected by qRT-PCR in all samples with heterogeneous MET expression microdissected from five cases with MET-mutated adenocarcinoma, while MET gene amplification was detected in tumor areas expressing high MET protein levels among MET-mutated adenocarcinomas., Conclusion: MET-mutated NSCLC is characterized by older age in patients with adenocarcinoma and by an acinar histology and variable MET expression in patients with adenocarcinoma and pleomorphic carcinomas. Moreover, MET gene amplification might occur in the tumor cells harboring the MET mutation., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
12. Anti-c-Met monoclonal antibody ABT-700 breaks oncogene addiction in tumors with MET amplification.
- Author
-
Wang J, Goetsch L, Tucker L, Zhang Q, Gonzalez A, Vaidya KS, Oleksijew A, Boghaert E, Song M, Sokolova I, Pestova E, Anderson M, Pappano WN, Ansell P, Bhathena A, Naumovski L, Corvaia N, and Reilly EB
- Subjects
- Animals, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents metabolism, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Gene Amplification, Humans, Male, Mice, Mice, SCID, Neoplasms, Experimental drug therapy, Neoplasms, Experimental genetics, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Protein Binding, Proto-Oncogene Mas, Proto-Oncogene Proteins c-met antagonists & inhibitors, Xenograft Model Antitumor Assays, Antibodies, Monoclonal pharmacology, Antineoplastic Agents pharmacology, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics
- Abstract
Background: c-Met is the receptor tyrosine kinase for hepatocyte growth factor (HGF) encoded by the MET proto-oncogene. Aberrant activation of c-Met resulting from MET amplification and c-Met overexpression is associated with poor clinical outcome in multiple malignancies underscoring the importance of c-Met signaling in cancer progression. Several c-Met inhibitors have advanced to the clinic; however, the development of inhibitory c-Met-directed therapeutic antibodies has been hampered by inherent agonistic activity., Method: We generated and tested a bivalent anti-c-Met monoclonal antibody ABT-700 in vitro for binding potency and antagonistic activity and in vivo for antitumor efficacy in human tumor xenografts. Human cancer cell lines and gastric cancer tissue microarrays were examined for MET amplification by fluorescence in situ hybridization (FISH)., Results: ABT-700 exhibits a distinctive ability to block both HGF-independent constitutive c-Met signaling and HGF-dependent activation of c-Met. Cancer cells addicted to the constitutively activated c-Met signaling driven by MET amplification undergo apoptosis upon exposure to ABT-700. ABT-700 induces tumor regression and tumor growth delay in preclinical tumor models of gastric and lung cancers harboring amplified MET. ABT-700 in combination with chemotherapeutics also shows additive antitumor effect. Amplification of MET in human cancer tissues can be identified by FISH., Conclusions: The preclinical attributes of ABT-700 in blocking c-Met signaling, inducing apoptosis and suppressing tumor growth in cancers with amplified MET provide rationale for examining its potential clinical utility for the treatment of cancers harboring MET amplification.
- Published
- 2016
- Full Text
- View/download PDF
13. miR-144-3p exerts anti-tumor effects in glioblastoma by targeting c-Met.
- Author
-
Lan F, Yu H, Hu M, Xia T, and Yue X
- Subjects
- 3' Untranslated Regions genetics, Animals, Antineoplastic Agents, Alkylating therapeutic use, Apoptosis drug effects, Cell Line, Tumor, Cell Survival drug effects, Dacarbazine analogs & derivatives, Dacarbazine therapeutic use, Humans, Mice, Mice, Nude, PTEN Phosphohydrolase biosynthesis, PTEN Phosphohydrolase genetics, Radiation-Sensitizing Agents pharmacology, Survival Analysis, Temozolomide, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Glioblastoma drug therapy, Glioblastoma genetics, MicroRNAs genetics, Proto-Oncogene Proteins c-met drug effects
- Abstract
The study aimed to explore the specific function and mechanism of miR-144-3p in glioblastoma (GBM) cells with different phosphatase and tensin homolog (PTEN) phenotypes. We demonstrated that the miR-144-3p level was significantly down-regulated in glioma compared with the non-neoplastic brain tissues, and decreased with ascending grades. The loss of miR-144-3p effectively predicted the decreased overall survival in glioma patients. Interestingly, the expression of MET was up-regulated and inversely associated with miR-144-3p level in glioma tissues. Next, we certified that miR-144-3p specifically bound to MET 3'-untranslated region (3' UTR) and inhibited its expression. miR-144-3p potently repressed GBM cell proliferation and invasion via suppressing MET in vitro and in vivo. In addition, our results showed no difference in malignancy inhibition induced by miR-144-3p in GBM cells with different PTEN phenotypes. miR-144-3p inhibited several survival signaling pathways by targeting MET independent of PTEN status in GBM cells. Over-expression of miR-144-3p inhibited survival capability and increased apoptosis, resulting in enhancement of radiation and temozolomide sensitivity. Our data provide new insights into the potential application of miR-144-3p in GBM therapy by targeting MET and then inhibiting the downstream signaling., (© 2015 International Society for Neurochemistry.)
- Published
- 2015
- Full Text
- View/download PDF
14. Novel role for endogenous hepatocyte growth factor in the pathogenesis of intracranial aneurysms.
- Author
-
Peña-Silva RA, Chalouhi N, Wegman-Points L, Ali M, Mitchell I, Pierce GL, Chu Y, Ballas ZK, Heistad D, and Hasan D
- Subjects
- Adult, Aged, Aneurysm, Ruptured metabolism, Animals, Cells, Cultured, Disease Models, Animal, E-Selectin metabolism, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Female, Hepatocyte Growth Factor blood, Hepatocyte Growth Factor pharmacology, Humans, Intracranial Aneurysm metabolism, Male, Mice, Mice, Inbred C57BL, Middle Aged, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular pathology, Proto-Oncogene Proteins c-met antagonists & inhibitors, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Pyrazines adverse effects, Pyrazines pharmacology, Signal Transduction physiology, Triazoles adverse effects, Triazoles pharmacology, Vascular Cell Adhesion Molecule-1 metabolism, Aneurysm, Ruptured etiology, Aneurysm, Ruptured physiopathology, Hepatocyte Growth Factor physiology, Intracranial Aneurysm etiology, Intracranial Aneurysm physiopathology
- Abstract
Inflammation plays a key role in formation and rupture of intracranial aneurysms. Because hepatocyte growth factor (HGF) protects against vascular inflammation, we sought to assess the role of endogenous HGF in the pathogenesis of intracranial aneurysms. Circulating HGF concentrations in blood samples drawn from the lumen of human intracranial aneurysms or femoral arteries were compared in 16 patients. Tissue from superficial temporal arteries and ruptured or unruptured intracranial aneurysms collected from patients undergoing clipping (n=10) were immunostained with antibodies to HGF and its receptor c-Met. Intracranial aneurysms were induced in mice treated with PF-04217903 (a c-Met antagonist) or vehicle. Expression of inflammatory molecules was also measured in cultured human endothelial, smooth muscle cells and monocytes treated with lipopolysaccharides in presence or absence of HGF and PF-04217903. We found that HGF concentrations were significantly higher in blood collected from human intracranial aneurysms (1076±656 pg/mL) than in femoral arteries (196±436 pg/mL; P<0.001). HGF and c-Met were detected by immunostaining in superficial temporal arteries and in both ruptured and unruptured human intracranial aneurysms. A c-Met antagonist did not alter the formation of intracranial aneurysms (P>0.05), but significantly increased the prevalence of subarachnoid hemorrhage and decreased survival in mice (P<0.05). HGF attenuated expression of vascular cell adhesion molecule-1 (P<0.05) and E-Selectin (P<0.05) in human aortic endothelial cells. In conclusion, plasma HGF concentrations are elevated in intracranial aneurysms. HGF and c-Met are expressed in superficial temporal arteries and in intracranial aneurysms. HGF signaling through c-Met may decrease inflammation in endothelial cells and protect against intracranial aneurysm rupture., (© 2014 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
15. Molecular targets in the treatment of non-small-cell lung cancer: is there hope on the horizon?
- Author
-
Carter CA, Nations JA, and Lazarus A
- Subjects
- Adenocarcinoma diagnostic imaging, Adenocarcinoma drug therapy, Adenocarcinoma genetics, Carcinoma, Non-Small-Cell Lung diagnostic imaging, Carcinoma, Non-Small-Cell Lung genetics, ErbB Receptors genetics, Humans, Lung Neoplasms diagnostic imaging, Lung Neoplasms genetics, Proto-Oncogene Proteins drug effects, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins B-raf drug effects, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins c-ret drug effects, Proto-Oncogene Proteins c-ret genetics, Proto-Oncogene Proteins p21(ras), Proto-Oncogenes genetics, Tomography, X-Ray Computed, ras Proteins drug effects, ras Proteins genetics, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Molecular Targeted Therapy, Proto-Oncogenes drug effects
- Abstract
Non-small-cell lung cancer (NSCLC) is a growing concern worldwide, and its incidence continues to increase in developing countries. It has a strong association with smoking. Lung cancer remains the leading cause of cancer-related deaths in most industrialized countries and in the United States. In the last 10 years, there have been significant advancements in the understanding of molecular oncogenes and how they play a role in driving lung cancer to both grow and metastasize. Understanding this rapidly expanding field has the potential to extend life, and it is an important field for all providers to conceptualize if they are treating patients with lung cancer. Currently, > 50% of all NSCLC is linked to 1 of several known genetic driver mutations. Using online databases, expert opinion, and practice-changing trials, we review the current standards of molecular testing of NSCLC and the expanding evidence of oncogenic drivers in nonsquamous NSCLC.
- Published
- 2014
- Full Text
- View/download PDF
16. Silencing Met receptor tyrosine kinase signaling decreased oral tumor growth and increased survival of nude mice.
- Author
-
Tao X, Hill KS, Gaziova I, Sastry SK, Qui S, Szaniszlo P, Fennewald S, Resto VA, and Elferink LA
- Subjects
- Animals, Apoptosis drug effects, Cell Movement drug effects, Cell Proliferation drug effects, Indoles pharmacology, Lymphatic Metastasis, Mice, Mice, Nude, Neoplasms, Experimental, Piperazines pharmacology, Proto-Oncogene Proteins c-met drug effects, RNA, Small Interfering pharmacology, Signal Transduction drug effects, Sulfonamides pharmacology, Proto-Oncogene Proteins c-met metabolism, Tongue Neoplasms metabolism
- Abstract
Objectives: The hepatocyte growth factor receptor (Met) is frequently overexpressed in Head and Neck Squamous Cell Carcinoma (HNSCC), correlating positively with high-grade tumors and shortened patient survival. As such, Met may represent an important therapeutic target. The purpose of this study was to explore the role of Met signaling for HNSCC growth and locoregional dissemination., Materials and Methods: Using a lentiviral system for RNA interference, we knocked down Met in established HNSCC cell lines that express high levels of the endogenous receptor. The effect of Met silencing on in vitro proliferation, cell survival and migration was examined using western analysis, immunohistochemistry and live cell imaging. In vivo tumor growth, dissemination and mouse survival was assessed using an orthotopic tongue mouse model for HNSCC., Results: We show that Met knockdown (1) impaired activation of downstream MAPK signaling; (2) reduced cell viability and anchorage independent growth; (3) abrogated HGF-induced cell motility on laminin; (4) reduced in vivo tumor growth by increased cell apoptosis; (5) caused reduced incidence of tumor dissemination to regional lymph nodes and (6) increased the survival of nude mice with orthotopic xenografts., Conclusion: Met signaling is important for HNSCC growth and locoregional dissemination in vivo and that targeting Met may be an important strategy for therapy., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
17. Liver protective effect of ursodeoxycholic acid includes regulation of ADAM17 activity.
- Author
-
Buryova H, Chalupsky K, Zbodakova O, Kanchev I, Jirouskova M, Gregor M, and Sedlacek R
- Subjects
- ADAM17 Protein, Animals, Bile Ducts surgery, Cholestasis, Hep G2 Cells, Humans, Ligation, MAP Kinase Signaling System drug effects, Mice, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Transforming Growth Factor alpha drug effects, Transforming Growth Factor alpha metabolism, Tumor Necrosis Factor-alpha drug effects, Tumor Necrosis Factor-alpha metabolism, ADAM Proteins drug effects, Cholagogues and Choleretics pharmacology, Hepatocytes drug effects, Liver drug effects, Ursodeoxycholic Acid pharmacology
- Abstract
Background: Ursodeoxycholic acid (UDCA) is used to treat primary biliary cirrhosis, intrahepatic cholestasis, and other cholestatic conditions. Although much has been learned about the molecular basis of the disease pathophysiology, our understanding of the effects of UDCA remains unclear. Possibly underlying its cytoprotective, anti-apoptotic, anti-oxidative effects, UDCA was reported to regulate the expression of TNFα and other inflammatory cytokines. However, it is not known if this effect involves also modulation of ADAM family of metalloproteinases, which are responsible for release of ectodomains of inflammatory cytokines from the cell surface. We hypothesized that UDCA modulates ADAM17 activity, resulting in amelioration of cholestasis in a murine model of bile duct ligation (BDL)., Methods: The effect of UDCA on ADAM17 activity was studied using the human liver hepatocellular carcinoma cell line HepG2. Untransfected cells or cells ectopically expressing human ADAM17 were cultured with or without UDCA and further activated using phorbol-12-myristate-13-acetate (PMA). The expression and release of ADAM17 substrates, TNFα, TGFα, and c-Met receptor (or its soluble form, sMet) were evaluated using ELISA and quantitative real-time (qRT) PCR. Immunoblotting analyses were conducted to evaluate expression and activation of ADAM17 as well as the level of ERK1/2 phosphorylation after UDCA treatment. The regulation of tissue inhibitor of metalloproteinases-1 (TIMP-1) by UDCA was studied using zymography and qRT-PCR. A mouse model of acute cholestasis was induced by common BDL technique, during which mice received daily orogastric gavage with either UDCA or vehicle only. Liver injury was quantified using alkaline phosphatase (ALP), relative liver weight, and confirmed by histological analysis. ADAM17 substrates in sera were assessed using a bead multiplex assay., Results: UDCA decreases amount of shed TNFα, TGFα, and sMet in cell culture media and the phosphorylation of ERK1/2. These effects are mediated by the reduction of ADAM17 activity in PMA stimulated cells although the expression ADAM17 is not affected. UDCA reduced the level of the mature form of ADAM17. Moreover, UDCA regulates the expression of TIMP-1 and gelatinases activity in PMA stimulated cells. A BDL-induced acute cholangitis model was characterized by increased relative liver weight, serum levels of ALP, sMet, and loss of intracellular glycogen. UDCA administration significantly decreased ALP and sMet levels, and reduced relative liver weight. Furthermore, hepatocytes of UDCA-treated animals retained their metabolic activity as evidenced by the amount of glycogen storage., Conclusions: The beneficial effect of UDCA appears to be mediated in part by the inhibition of ADAM17 activation and, thus, the release of TNFα, a strong pro-inflammatory factor. The release of other ADAM17 substrates, TGFα and sMet, are also regulated this way, pointing to a general impact on the release of ADAM17 substrates, which are pivotal for liver regeneration and function. In parallel, UDCA upregulates TIMP-1 that in turn inhibits matrix metalloproteinases, which destroy the hepatic ECM in diseased liver. This control of extracellular matrix turnover represents an additional beneficial path of UDCA treatment.
- Published
- 2013
- Full Text
- View/download PDF
18. The impact of genomic changes on treatment of lung cancer.
- Author
-
Cardarella S and Johnson BE
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma of Lung, Anaplastic Lymphoma Kinase, Antineoplastic Agents therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, ErbB Receptors therapeutic use, Genomics, Humans, Lung Neoplasms drug therapy, Molecular Targeted Therapy methods, Mutation drug effects, Mutation genetics, Oncogene Proteins drug effects, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins drug effects, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins B-raf drug effects, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins c-ret drug effects, Proto-Oncogene Proteins c-ret genetics, Proto-Oncogene Proteins p21(ras), Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor Protein-Tyrosine Kinases therapeutic use, Receptor, ErbB-2 drug effects, Receptor, ErbB-2 genetics, ras Proteins drug effects, ras Proteins genetics, Adenocarcinoma genetics, Carcinoma, Non-Small-Cell Lung genetics, ErbB Receptors genetics, Lung Neoplasms genetics, Oncogene Proteins genetics, Proto-Oncogene Proteins B-raf genetics, Receptor Protein-Tyrosine Kinases genetics
- Abstract
The remarkable success of epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors in patients with EGFR mutations and ALK rearrangements, respectively, introduced the era of targeted therapy in advanced non-small cell lung cancer (NSCLC), shifting treatment from platinum-based combination chemotherapy to molecularly tailored therapy. Recent genomic studies in lung adenocarcinoma identified other potential therapeutic targets, including ROS1 rearrangements, RET fusions, MET amplification, and activating mutations in BRAF, HER2, and KRAS in frequencies exceeding 1%. Lung cancers that harbor these genomic changes can potentially be targeted with agents approved for other indications or under clinical development. The need to generate increasing amounts of genomic information should prompt health-care providers to be mindful of the amounts of tissue needed for these assays when planning diagnostic procedures. In this review, we summarize oncogenic drivers in NSCLC that can be currently detected, highlight their potential therapeutic implications, and discuss practical considerations for successful application of tumor genotyping in clinical decision making.
- Published
- 2013
- Full Text
- View/download PDF
19. HGF and IGF-1 synergize with SDF-1α in promoting migration of myeloma cells by cooperative activation of p21-activated kinase.
- Author
-
Rø TB, Holien T, Fagerli UM, Hov H, Misund K, Waage A, Sundan A, Holt RU, and Børset M
- Subjects
- Autocrine Communication, Cell Line, Tumor, Cell Movement drug effects, Chemokine CXCL12 physiology, Dose-Response Relationship, Drug, Drug Synergism, Enzyme Activation, Hepatocyte Growth Factor physiology, Humans, Insulin-Like Growth Factor I physiology, Neoplasm Proteins physiology, Proto-Oncogene Proteins c-met drug effects, RNA, Small Interfering pharmacology, Receptors, CXCR4 physiology, Recombinant Fusion Proteins physiology, Transfection, p21-Activated Kinases antagonists & inhibitors, p21-Activated Kinases genetics, Chemokine CXCL12 pharmacology, Hepatocyte Growth Factor pharmacology, Insulin-Like Growth Factor I pharmacology, Multiple Myeloma pathology, p21-Activated Kinases physiology
- Abstract
Stromal-derived factor (SDF)-1α, insulin-like growth factor (IGF)-1 and hepatocyte growth factor (HGF) are potent mediators of cell migration. We studied the effect of combinations of these cytokines on the migration of myeloma cells. When SDF-1α was combined with either HGF or IGF-1, we found a striking synergy in the cytokines' ability to guide cells across a transwell membrane. Between HGF and IGF-1 there was no cooperativity. However, the effects of HGF and IGF-1 were not redundant. HGF and SDF-1 caused concentration gradient-directed migration, as opposed to IGF-1, which apparently caused randomly directed cell movement. The SDF-1α-driven migration of JJN-3 cells, a myeloma cell line secreting large amounts of HGF, was reduced when JJN-3 cells were given an inhibitor of the HGF receptor, demonstrating a cooperative activity between autocrine HGF and exogenous SDF-1α. There was a clear positive correlation between the degree of cytokine-induced migration and phosphorylation of p21-activated kinase (PAK) both in primary myeloma cells and in cell lines including INA-6 and IH-1. Downregulation of PAK with small interfering RNA in INA-6 cells resulted in decreased cytokine-driven migration. This study shows synergy between SDF-1α and HGF/IGF-1 in inducing migration of myeloma cells, yet each cytokine has distinct properties in the way it regulates cell migration. These findings are likely to be of clinical relevance because multiple myeloma cells are located in an environment containing HGF and IGF-1 and are exposed to an SDF-1α gradient between the bone marrow and peripheral blood., (Copyright © 2013 ISEH - Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
20. Non-small cell lung cancer--genetic predictors.
- Author
-
Koudelakova V, Kneblova M, Trojanec R, Drabek J, and Hajduch M
- Subjects
- Carcinoma, Non-Small-Cell Lung drug therapy, Drug Resistance, Neoplasm, ErbB Receptors drug effects, ErbB Receptors genetics, Humans, Lung Neoplasms drug therapy, Oncogene Proteins, Fusion drug effects, Oncogene Proteins, Fusion genetics, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases drug effects, Protein-Tyrosine Kinases genetics, Proto-Oncogene Proteins drug effects, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins p21(ras), ras Proteins drug effects, ras Proteins genetics, Biomarkers, Tumor genetics, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms genetics
- Abstract
Background: Non-small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer that is the leading cause of cancer-related mortality worldwide. Several predictive markers have been found in NSCLC patients to date but only a few are currently used for tailored therapy., Methods and Results: PubMed and Web of Science online databases were used to search review and original articles on the most important predictive markers in NSCLC., Conclusion: EGFR activating mutations (exons 18 to 21) and EML4-ALK rearrangement are clinically important markers able to select NSCLC patients which benefit from EGFR or ALK tyrosine kinase inhibitors (gefitinib, erlotinib, crizotinib). Other markers, such as KRAS mutation, EGFR T790M mutation and C-MET amplification, are responsible for resistance to these inhibitors. Overcoming of this resistance as well as discovery of new potential markers and inhibitors is the main goal of ongoing research and clinical trials in NSCLC.
- Published
- 2013
- Full Text
- View/download PDF
21. Underexpression of miR-34a in hepatocellular carcinoma and its contribution towards enhancement of proliferating inhibitory effects of agents targeting c-MET.
- Author
-
Dang Y, Luo D, Rong M, and Chen G
- Subjects
- Adult, Aged, Aged, 80 and over, DNA Primers, Female, Humans, Male, Middle Aged, Phenotype, Polymerase Chain Reaction, Carcinoma, Hepatocellular genetics, Liver Neoplasms genetics, MicroRNAs genetics, Proto-Oncogene Proteins c-met drug effects
- Abstract
Aberrant expression of microRNA-34a (miR-34a) has been reported to be involved in the tumorigenesis and progression of various classes of malignancies. However, its role in hepatocellular carcinoma (HCC) has not been completely clarified. In the current study, we have investigated the clinical significance and the in vitro contribution of miR-34a on biological functions of human HCCs. miR-34a expression in eighty-three cases of HCC formalin-fixed paraffin-embedded (FFPE) tissues decreased significantly compared to that in the adjacent liver tissues (P<0.01), as detected by real-time quantitative RT-PCR (RT-qPCR). miR-34a expression in the groups of TNM stage I and II, without metastasis and without portal vein tumor embolus, was significantly higher than that of their corresponding groups (P<0.05). In functional experiments, miR-34a mimic suppressed cell growth, migration and invasion, meanwhile it increased cellular apoptosis and caspase activity in HCC cells. miR-34a mimic also reduced phospho-ERK1/2 and phospho-stat5 signaling. In addition, miR-34a mimic enhanced the effect of cell proliferation inhibition and caspase activity induction of agents targeting c-MET (siRNAs and small molecular inhibitor su11274). In conclusion, miR-34a may act as a tumor suppressor miRNA of HCC. The strategies to increase miR-34a level might be a critical targeted therapy for HCC in future.
- Published
- 2013
- Full Text
- View/download PDF
22. 17AEP-GA, an HSP90 antagonist, is a potent inhibitor of glioblastoma cell proliferation, survival, migration and invasion.
- Author
-
Miekus K, Kijowski J, Sekuła M, and Majka M
- Subjects
- Benzoquinones chemistry, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Glioblastoma pathology, Hepatocyte Growth Factor metabolism, Humans, Lactams, Macrocyclic chemistry, Mitogen-Activated Protein Kinases metabolism, Neoplasm Invasiveness, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Antibiotics, Antineoplastic pharmacology, Benzoquinones pharmacology, Glioblastoma drug therapy, Glioblastoma metabolism, HSP90 Heat-Shock Proteins antagonists & inhibitors, Lactams, Macrocyclic pharmacology
- Abstract
Glioblastoma multiforme (GBM) is the most frequent and the most malignant human brain tumor. The expression of receptor tyrosine kinase MET and its ligand hepatocyte growth factor (HGF) is strongly increased in GBM, where they promote tumor proliferation, cell survival, migration, invasion and angiogenesis. We used geldanamycins (GAs) (inhibitors of HSP90) in order to block glioblastoma growth and HGF-dependent cell migration and invasion. The effect of GAs on three GBM cell lines was tested and we found their antiproliferative effect on tumor cells. The maximum level of inhibition reached 70%. After treatment with GAs, cells also became apoptotic as determined by Annexin V-positive staining and activation of the caspase-3 pathway. We examined the expression and activity of the MET receptor on GBM cell lines and we observed phosphorylation of AKT and MAPK after HGF stimulation by western blot analysis. Since GBM cells express high level of MET receptor and were shown to respond to HGF by increased motility we tested if GAs could negatively affect GBM cell movement. In our study, we found that GAs inhibited the chemotaxis of glioblastoma cells toward the hepatocyte growth factor gradient. The GAs also blocked migration of tumor cells through a Matrigel layer in invasion assays. The strongest inhibitory effect was observed for GA and its analog, 17AEP-GA. Based on our results, GAs, particularly 17AEP-GA, could be considered as a new potential agent to treat glioblastoma multiforme.
- Published
- 2012
- Full Text
- View/download PDF
23. Targeted therapy of hepatocellular carcinoma: present and future.
- Author
-
Chan SL and Yeo W
- Subjects
- Angiogenesis Inhibitors pharmacology, Angiogenesis Inhibitors therapeutic use, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Carcinoma, Hepatocellular blood supply, Epigenesis, Genetic drug effects, ErbB Receptors drug effects, Hepatocyte Growth Factor antagonists & inhibitors, Humans, Liver Neoplasms blood supply, Neovascularization, Pathologic prevention & control, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-met antagonists & inhibitors, Proto-Oncogene Proteins c-met drug effects, Signal Transduction drug effects, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy, Molecular Targeted Therapy
- Abstract
Following the encouraging results of sorafenib in advanced hepatocellular carcinoma (HCC), targeted therapy has become a new direction of research in the treatment of HCC. Emerging data provide evidence that the pathogenesis and progression of HCC are mediated by a number of molecular defects and dysregulated pathways. Novel targeted therapies are designed to inhibit the aberrant pathways at a molecular level with an aim to improve the clinical outcome. For the past few years, an increasing number of targeted agents have been tested in HCC in the clinical setting. This review aims to summarize the current status of clinical development of targeted therapy in HCC, with focus on novel agents targeting angiogenesis, signal transduction and epigenetic dysregulation of tumors. The review also discusses the lessons learned from outcomes of completed clinical trials and provides perspectives on future clinical trials in HCC., (© 2012 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
24. Gene-selective histone H3 acetylation in the absence of increase in global histone acetylation in liver of rats chronically fed alcohol.
- Author
-
Park PH, Lim RW, and Shukla SD
- Subjects
- Acetylation drug effects, Alcohol Dehydrogenase drug effects, Alcohol Dehydrogenase genetics, Animals, Genes, jun drug effects, Hepatocyte Growth Factor genetics, Histones chemistry, Histones metabolism, Liver metabolism, Lysine, Male, Nitric Oxide Synthase Type II drug effects, Nitric Oxide Synthase Type II genetics, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics, RNA, Messenger analysis, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, bcl-2-Associated X Protein drug effects, bcl-2-Associated X Protein genetics, Central Nervous System Depressants pharmacology, Ethanol pharmacology, Gene Expression drug effects, Histones drug effects, Liver drug effects, Promoter Regions, Genetic drug effects, Protein Processing, Post-Translational drug effects
- Abstract
Aims: The aim of this study was to determine the effect of chronic ethanol feeding on acetylation of histone H3 at lysine 9 (H3-Lys9) at promoter and coding regions of genes for class I alcohol dehydrogenase (ADH I), inducible nitric oxide synthase (iNOS), Bax, p21, c-met and hepatocyte growth factor in the rat liver., Methods: Rats were fed ethanol-containing liquid diet (5%, w/v) for 1-4 weeks. The global level of acetylation of H3-Lys9 in the liver was examined by western blot analysis. The levels of mRNA for various genes were measured by real-time reverse transcriptase-polymerase chain reaction. The association of acetylated histone H3-Lys9 with the different regions of genes was monitored by chromatin immunoprecipitation assay., Results: Chronic ethanol treatment increased mRNA expression of genes for iNOS, c-jun and ADH 1. Chronic ethanol treatment did not cause increase in global acetylation of H3-Lys9, but significantly increased the association of acetylated histone H3-Lys9 in the ADH I gene, both in promoter and in coding regions. In contrast, chronic ethanol treatment did not significantly increase the association of acetylated histone H3-Lys9 with iNOS and c-jun genes., Conclusion: Chronic ethanol exposure increased the gene-selective association of acetylated H3-Lys9 in the absence of global histone acetylation. Thus, not all genes expressed by ethanol are linked to transcription via histone H3 acetylation at Lys9.
- Published
- 2012
- Full Text
- View/download PDF
25. New promising molecular targets in head and neck squamous cell carcinoma.
- Author
-
Bauman JE, Michel LS, and Chung CH
- Subjects
- Alcohol Drinking adverse effects, Alphapapillomavirus, Antineoplastic Agents therapeutic use, Carcinoma, Squamous Cell etiology, Female, Head and Neck Neoplasms etiology, Humans, Male, Molecular Targeted Therapy trends, Papillomavirus Infections complications, Phosphatidylinositol 3-Kinases drug effects, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Receptor, Notch1 drug effects, Receptor, Notch1 metabolism, Smoking adverse effects, Squamous Cell Carcinoma of Head and Neck, TOR Serine-Threonine Kinases drug effects, TOR Serine-Threonine Kinases metabolism, Tumor Suppressor Protein p53 drug effects, Tumor Suppressor Protein p53 metabolism, Antineoplastic Agents pharmacology, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell metabolism, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms metabolism, Molecular Targeted Therapy methods
- Abstract
Purpose of Review: Despite advances in multimodality therapy, the overall 5-year survival rate is 40-50% in patients with head and neck squamous cell carcinoma (HNSCC) and current multimodality approaches impart significant toxicities. This review highlights promising targets with the potential to improve clinical outcomes in HNSCC., Recent Findings: In addition to mutagenic exposure to tobacco and alcohol as risk factors, recent studies have shown that human papillomavirus is one of the main causes of HNSCC and as such is being investigated as a therapeutic target. Furthermore, recent data generated from whole exome sequencing of HNSCC, new insights into the biology of DNA damage repair, and increased understanding of tumor hypoxia responses are pointing to new therapeutic possibilities for treating HNSCC., Summary: HNSCC is a heterogeneous disease. Improved treatment will require a rapid translation of basic science research, and the simultaneous development of novel therapeutics and corresponding biomarkers to guide their application.
- Published
- 2012
- Full Text
- View/download PDF
26. Targeting the Met pathway in lung cancer.
- Author
-
Belalcazar A, Azaña D, Perez CA, Raez LE, and Santos ES
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung pathology, Humans, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Carcinoma, Non-Small-Cell Lung metabolism, Lung Neoplasms metabolism, Proto-Oncogene Proteins c-met drug effects
- Abstract
Dysregulation of Met signaling has been implicated in the initiation, progression and metastasis of human cancers, and therefore represents an attractive target for anticancer drug development. Met is overexpressed in non-small-cell lung cancer and its lack of staining in normal lung tissue makes it an attractive target. To date, erlotinib and gefitinib have established themselves as first-line therapy for non-small-cell lung cancer patients whose tumors harbor an EGF receptor gene mutation, and hence, it is crucial that we identify mechanisms of resistance that could be targeted by novel agents, while keeping an acceptable toxicity profile at the same time; something very important when we develop these new drugs. Inhibitors of the Met pathway represent a therapeutic alternative in this setting. In this review, we discuss the early clinical studies reported using two Met inhibitors, a monoclonal antibody (MetMAb) and a small molecule tyrosine kinase inhibitor (MGCD265).
- Published
- 2012
- Full Text
- View/download PDF
27. Wnt, osteosarcoma, and future therapy.
- Author
-
Hoang BH
- Subjects
- Bone Neoplasms metabolism, Cell Transformation, Neoplastic drug effects, DNA Methylation, Disease Progression, Humans, Low Density Lipoprotein Receptor-Related Protein-5 metabolism, Neoplasm Metastasis genetics, Osteosarcoma metabolism, Prognosis, Proto-Oncogene Proteins c-met drug effects, Wnt Signaling Pathway drug effects, Bone Neoplasms drug therapy, Bone Neoplasms pathology, Osteosarcoma drug therapy, Osteosarcoma pathology, Wnt Signaling Pathway physiology
- Published
- 2012
- Full Text
- View/download PDF
28. Induction of MET by ionizing radiation and its role in radioresistance and invasive growth of cancer.
- Author
-
De Bacco F, Luraghi P, Medico E, Reato G, Girolami F, Perera T, Gabriele P, Comoglio PM, and Boccaccio C
- Subjects
- Animals, Apoptosis radiation effects, Ataxia Telangiectasia Mutated Proteins, Blotting, Northern, Cell Cycle Proteins genetics, Cell Cycle Proteins radiation effects, Cell Line, Tumor, Cell Movement radiation effects, Cell Survival radiation effects, Chromatin Immunoprecipitation, DNA-Binding Proteins genetics, DNA-Binding Proteins radiation effects, Enzyme-Linked Immunosorbent Assay, Gene Expression Regulation, Neoplastic drug effects, Gene Silencing, Humans, In Situ Nick-End Labeling, Indoles pharmacology, Mice, Mitogen-Activated Protein Kinases metabolism, NF-kappa B genetics, NF-kappa B radiation effects, Neoplasm Invasiveness prevention & control, Neoplasms pathology, Neoplasms radiotherapy, Phosphorylation radiation effects, Polymerase Chain Reaction, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases radiation effects, Proto-Oncogene Proteins c-met antagonists & inhibitors, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins c-met radiation effects, RNA, Messenger metabolism, RNA, Small Interfering, Radiation Tolerance, Radiation, Ionizing, Radiation-Sensitizing Agents pharmacology, Receptors, Growth Factor drug effects, Receptors, Growth Factor genetics, Receptors, Growth Factor radiation effects, Sulfones pharmacology, Transcription, Genetic radiation effects, Transplantation, Heterologous, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins radiation effects, Up-Regulation radiation effects, Cell Cycle Proteins metabolism, DNA Damage radiation effects, DNA-Binding Proteins metabolism, NF-kappa B metabolism, Neoplasms metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-met metabolism, Receptors, Growth Factor antagonists & inhibitors, Receptors, Growth Factor metabolism, Signal Transduction radiation effects, Tumor Suppressor Proteins metabolism
- Abstract
Background: Ionizing radiation (IR) is effectively used in cancer therapy. However, in subsets of patients, a few radioresistant cancer cells survive and cause disease relapse with metastatic progression. The MET oncogene encodes the hepatocyte growth factor (HGF) receptor and is known to drive "invasive growth", a regenerative and prosurvival program unduly activated in metastasis., Methods: Human tumor cell lines (MDA-MB-231, MDA-MB-435S, U251) were subjected to therapeutic doses of IR. MET mRNA, and protein expression and signal transduction were compared in treated and untreated cells, and the involvement of the DNA-damage sensor ataxia telangiectasia mutated (ATM) and the transcription factor nuclear factor kappa B (NF-κB) in activating MET transcription were analyzed by immunoblotting, chromatin immunoprecipitation, and use of NF-κB silencing RNA (siRNA). Cell invasiveness was measured in wound healing and transwell assays, and cell survival was measured in viability and clonogenic assays. MET was inhibited by siRNA or small-molecule kinase inhibitors (PHA665752 or JNJ-38877605). Combinations of MET-targeted therapy and radiotherapy were assessed in MDA-MB-231 and U251 xenografts (n = 5-6 mice per group). All P values were from two-sided tests., Results: After irradiation, MET expression in cell lines was increased up to fivefold via activation of ATM and NF-κB. MET overexpression increased ligand-independent MET phosphorylation and signal transduction, and rendered cells more sensitive to HGF. Irradiated cells became more invasive via a MET-dependent mechanism that was further enhanced in the presence of HGF. MET silencing by siRNA or inhibition of its kinase activity by treatment with PHA665752 or JNJ-38877605 counteracted radiation-induced invasiveness, promoted apoptosis, and prevented cells from resuming proliferation after irradiation in vitro. Treatment with MET inhibitors enhanced the efficacy of IR to stop the growth of or to induce the regression of xenografts (eg, at day 13, U251 xenografts, mean volume increase relative to mean tumor volume at day 0: vehicle = 438%, 5 Gy IR = 151%, 5 Gy IR + JNJ-38877605 = 76%; difference, IR vs JNJ-38877604 + IR = 75%, 95% CI = 59% to 91%, P = .01)., Conclusion: IR induces overexpression and activity of the MET oncogene through the ATM-NF-κB signaling pathway; MET, in turn, promotes cell invasion and protects cells from apoptosis, thus supporting radioresistance. Drugs targeting MET increase tumor cell radiosensitivity and prevent radiation-induced invasiveness.
- Published
- 2011
- Full Text
- View/download PDF
29. Effect of globin digest on the liver injury and hepatic gene expression profile in galactosamine-induced liver injury in SD rats.
- Author
-
Yamamoto K, Sasakawa Y, Nakaoka F, Nakao M, Nakamura M, Kominami A, Abe M, Fukuhama C, and Kagawa K
- Subjects
- Animals, Antioxidants metabolism, Chemical and Drug Induced Liver Injury etiology, Chemical and Drug Induced Liver Injury pathology, Disease Models, Animal, Galactosamine toxicity, Hepatocytes drug effects, Hepatocytes metabolism, Male, Mice, PPAR gamma drug effects, PPAR gamma genetics, Phosphorylation drug effects, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Rats, Rats, Sprague-Dawley, Chemical and Drug Induced Liver Injury prevention & control, Gene Expression Regulation drug effects, Globins pharmacology
- Abstract
Aims: We investigated the effect of globin digest (GD) on the liver injury and hepatic gene expression profile in galactosamine (GalN)-induced liver injury., Main Methods: The effect of GD on the liver injury was examined by measuring the activities of serum transferases and hepatic antioxidant enzymes, histopathological analysis, gene expression profile, and proteins of the peroxisome proliferator-activated receptor alpha (PPARα) and met proto-oncogene (c-Met) in SD rats at 24 h after GalN administration. The effect of GD on the expression of PPARα and its target gene in AML-12 mouse hepatocytes was also examined., Key Findings: GD suppressed the elevated activities of serum transferases in GalN-induced liver injury in SD rats. The thiobarbituric acid reactive substance content in GalN-injured liver was a decreasing tendency by GD. GD suppressed the increased oxidized glutathione content, and increased the decreased protein, reduced glutathione contents, and catalase activity in GalN-injured liver. GD may improve the antioxidant defense system and protein synthesis in GalN-injured liver. GD suppressed the elevated expression of the genes related to the inflammation, and decreased the histopathological grade value of inflammatory cell infiltration in GalN-injured liver. GD increased the expression of PPARα protein in GalN-injured liver, and also increased the expression of PPARα and its target gene in AML-12 hepatocytes. The total and phosphorylated c-Met proteins in GalN-injured liver were the increasing tendencies by GD., Significance: These findings indicate that GD has the hepatoprotective effect on GalN-induced liver injury in SD rats., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
30. [Study of motogenic signal in human melanoma cells].
- Author
-
Kenessey I
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, ErbB Receptors drug effects, Extracellular Matrix metabolism, Gene Expression Regulation, Neoplastic, Humans, Melanoma drug therapy, Mice, Mice, SCID, Proto-Oncogene Proteins c-met drug effects, Receptors, Purinergic P2X metabolism, Signal Transduction drug effects, Skin Neoplasms metabolism, Skin Neoplasms pathology, Antineoplastic Agents pharmacology, Calcium metabolism, Cell Movement drug effects, Enzyme Inhibitors pharmacology, ErbB Receptors metabolism, Melanoma metabolism, Melanoma pathology, Proto-Oncogene Proteins c-met metabolism, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor Protein-Tyrosine Kinases metabolism
- Abstract
The components of the extracellular matrix (ECM) are more than just adhesion sites for migrating tumor cells: following enzymatic degradation of the ECM, the release of sequestrated growth factors increases, thus they become available for tumor cells. In a number of cancers dysfunction of epidermal growth factor receptor (EGFR) or hepatocyte growth factor receptor (c-Met) contribute to the malignant transformation that directly regulates cell proliferation, survival and motility. Furthermore, intracellular calcium level plays an important role in the regulation of the tyrosine kinase pathway. In our preclinical experiments, by administering heparin-derived oligosaccharides we influenced the interaction between human melanoma cells and ECM. In vitro cell migration was inhibited by heparin fragments. Moreover, two of the effective oligosaccharides reduced the number of lung colonies formed in SCID mice. In human melanoma cells an important element of Ca2+ homeostasis, the purinergic Ca2+ channel P2X7 proved to be an anti-apoptotic protein. EGFR and c-Met showed constitutive activity in human melanoma cells, and their inhibition in vitro caused decreased proliferation, migration and elevated apoptosis. Administration of a selective c-Met-TKI significantly decreased primary tumor growth in vivo as well as the capacity for liver colony formation in SCID mice. Selective EGFR-TKI had less inhibitory effect on metastasis formation, and had no effect on the primary tumor. Our results suggest the necessity of a rational dual-specific drug design for the purpose in the therapy of malignant melanoma.
- Published
- 2011
- Full Text
- View/download PDF
31. Aspergiolides C and D: spirocyclic aromatic polyketides with potent protein kinase c-Met inhibitory effects.
- Author
-
Du L, Ai J, Li D, Zhu T, Wang Y, Knauer M, Bruhn T, Liu H, Geng M, Gu Q, and Bringmann G
- Subjects
- Anthraquinones isolation & purification, Aspergillus chemistry, Cell Movement drug effects, Hepatocyte Growth Factor metabolism, Humans, Macrolides isolation & purification, Molecular Structure, Phosphorylation, Proto-Oncogene Proteins c-met chemistry, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Quantum Theory, Stereoisomerism, Anthraquinones chemistry, Anthraquinones pharmacology, Aspergillus isolation & purification, Biological Products chemistry, Biological Products isolation & purification, Hepatocyte Growth Factor chemistry, Macrolides chemistry, Macrolides pharmacology, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met agonists, Spiro Compounds chemistry, Spiro Compounds pharmacology
- Abstract
Variation of the cultivation conditions for Aspergillus glaucus led to the discovery of two novel spirocyclic aromatic polyketides, aspergiolides C (3) and D (4). Their constitutions were elucidated by a combination of spectroscopic methods and isotope-labeling experiments. Aspergiolides C (3) and D (4) occur as racemic mixtures, the resolution of which was succeeded by HPLC on a chiral phase. The absolute configurations of their enantiomers were assigned online, from the peaks in the chromatogram, by a combination of HPLC-CD and quantum chemical CD calculations. Both compounds were found to inhibit the kinase activities of the receptor tyrosine kinases (RTKs) c-Met, Ron, and c-Src with low-micromolar IC(50)s. The enantiomers of 3 were resolved by HPLC on a chiral phase. Both enantiomers showed a comparable inhibition of the HGF-induced autophosphorylation of c-Met and of subsequent cell migration., (Copyright © 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2011
- Full Text
- View/download PDF
32. Acquired endocrine resistance in breast cancer: implications for tumour metastasis.
- Author
-
Hayes E, Nicholson RI, and Hiscox S
- Subjects
- Antineoplastic Agents, Hormonal pharmacology, Breast Neoplasms blood supply, Breast Neoplasms pathology, Cell Adhesion physiology, Estradiol analogs & derivatives, Estradiol pharmacology, Female, Fulvestrant, Humans, Hyaluronan Receptors biosynthesis, Proto-Oncogene Proteins c-met drug effects, src-Family Kinases metabolism, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm drug effects, Neoplasm Metastasis physiopathology
- Abstract
Endocrine therapy is the treatment of choice in hormone receptor-positive breast cancer. However, the effectiveness of these agents is limited by the development of drug resistance, ultimately leading to disease progression and patient mortality. Whilst pre-clinical cell models of acquired endocrine resistance have demonstrated a role for altered growth factor signalling in the development of an endocrine insensitive phenotype, it is becoming apparent that acquisition of endocrine resistance in breast cancer is also accompanied by the development of an adverse cellular phenotype, with resistant cells exhibiting altered adhesive interactions, enhanced migratory and invasive behaviour, and a capacity to induce angiogenic responses in endothelium. Since invasion and metastasis of cancer cells is a major cause of mortality in breast cancer patients, elucidation of molecular mechanisms underlying the adverse cellular features that accompany acquired endocrine resistance and their subsequent targeting may provide a means of limiting the progression of such tumours in vivo.
- Published
- 2011
- Full Text
- View/download PDF
33. Cordycepin inhibits renal interstitial myofibroblast activation probably by inducing hepatocyte growth factor expression.
- Author
-
Li L, He D, Yang J, and Wang X
- Subjects
- Actins genetics, Animals, Cell Proliferation drug effects, Cells, Cultured, Deoxyadenosines administration & dosage, Dose-Response Relationship, Drug, Fibroblasts metabolism, Hepatocyte Growth Factor metabolism, Kidney cytology, Kidney drug effects, Kidney metabolism, Myofibroblasts drug effects, Myofibroblasts metabolism, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Rats, Smad2 Protein metabolism, Smad3 Protein metabolism, Time Factors, Transforming Growth Factor beta1 pharmacology, Deoxyadenosines pharmacology, Fibroblasts drug effects, Gene Expression Regulation drug effects, Hepatocyte Growth Factor genetics
- Abstract
Renal interstitial fibrosis is the common end point of progressive renal diseases leading to the deterioration and eventual loss of renal function. This study investigated the effect and potential mechanism of cordycepin on activation of renal interstitial fibroblast cells. The time and dose-responses of cordycepin in rat renal interstitial fibroblast (NRK-49F) cells were analyzed. The proliferation of NRK-49F and the expression of α-smooth muscle actin (α-SMA) and fibronectin (FN) were examined. The expression and translocation of Smad proteins also were measured by western blot and indirect immunofluorescence staining. The mRNA level of hepatocyte growth factor (HGF) and the expression of HGF receptor c-Met and its phosphorylation (p-Met) were also detected. Cordycepin suppressed the proliferation of NRK-49F and the expression of α-SMA and FN induced by transforming growth factor-β1 (TGF-β1). The pretreatment of cordycepin markedly attenuated the nuclear translocation and accumulation of activated Smad2/3 in NRK-49F cells. Furthermore, cordycepin not only increased HGF expression, but also induced HGF secretion, as well as HGF receptor phosphorylation in NRK-49F cells. Cordycepin possesses renoprotective activity through suppression myofibroblast activation. This action is mediated, at least in part, by blocking nuclear translocation and accumulation of activated Smad2/3 protein and up-regulating anti-fibrotic HGF expression and secretion and HGF receptor activation.
- Published
- 2011
- Full Text
- View/download PDF
34. A disintegrin and metalloproteinase-10 (ADAM-10) mediates DN30 antibody-induced shedding of the met surface receptor.
- Author
-
Schelter F, Kobuch J, Moss ML, Becherer JD, Comoglio PM, Boccaccio C, and Krüger A
- Subjects
- ADAM10 Protein, Antibodies pharmacology, Cell Line, Tumor, Hepatocyte Growth Factor pharmacology, Humans, Neoplasm Invasiveness prevention & control, Neoplasm Metastasis drug therapy, Neoplasm Metastasis prevention & control, Proto-Oncogene Proteins c-met drug effects, Receptors, Growth Factor drug effects, ADAM Proteins physiology, Amyloid Precursor Protein Secretases physiology, Antibodies therapeutic use, Membrane Proteins physiology, Proto-Oncogene Proteins c-met metabolism, Receptors, Growth Factor metabolism
- Abstract
Met, the tyrosine kinase receptor for the hepatocyte growth factor is a prominent regulator of cancer cell invasiveness and has emerged as a promising therapeutic target. Binding of the anti-Met monoclonal antibody DN30 to its epitope induces the proteolytic cleavage of Met, thereby impairing the invasive growth of tumors. The molecular mechanism controlling this therapeutic shedding process has so far been unknown. Here, we report that A Disintegrin And Metalloproteinase (ADAM)-10, but not ADAM-17, is required for DN30-induced Met shedding. Knockdown of ADAM-10 in different tumor cell lines or abrogation of its proteolytic activity by natural or synthetic inhibitors abolished Met down-regulation on the cell surface as well as reduction of Met activation. Moreover, hepatocyte growth factor-induced tumor cell migration and invasion were impaired upon ADAM-10 knockdown. Thus, the therapeutic effect of DN30 involves ADAM-10-dependent Met shedding, linking for the first time a specific metalloprotease to target therapy against a receptor tyrosine kinase.
- Published
- 2010
- Full Text
- View/download PDF
35. Agonist monoclonal antibodies against HGF receptor protect cardiac muscle cells from apoptosis.
- Author
-
Pietronave S, Forte G, Locarno D, Merlin S, Zamperone A, Nicotra G, Isidoro C, Nardo PD, and Prat M
- Subjects
- Animals, Antibodies, Monoclonal immunology, Cell Line, Cell Movement drug effects, Cell Proliferation drug effects, Dogs, Extracellular Signal-Regulated MAP Kinases metabolism, Hydrogen Peroxide pharmacology, Mice, Models, Animal, Myocytes, Cardiac metabolism, NIH 3T3 Cells, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt drug effects, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-met immunology, Proto-Oncogene Proteins c-met metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Rats, Signal Transduction drug effects, Antibodies, Monoclonal pharmacology, Apoptosis drug effects, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Proto-Oncogene Proteins c-met drug effects
- Abstract
Hepatocyte growth factor (HGF), a pleiotropic cytokine with mitogenic, motogenic, morphogenic, and antiapoptotic effects in various cell types, is a cardioprotective growth factor that can counteract the loss of cardiomyocytes usually observed in cardiac diseases. HGF is a quite unstable molecule in its biologically active heterodimeric form. Since all HGF-induced biological responses are mediated by its high-affinity tyrosine kinase receptor (Met/HGF-R) encoded by the Met gene, we asked whether a monoclonal antibody (MAb) that displays receptor full agonist activity could protect cardiac muscle cell lines from hydrogen peroxide-induced apoptosis. We report that the MAb efficiently inhibited hydrogen peroxide-induced cell shrinkage, DNA fragmentation, annexin V positivity, mitochondrial translocation of bax, and caspase activation. The MAb was thus able to counteract apoptosis evaluated by both morphological and biochemical criteria. The agonist activity of the MAb was mediated by Met/HGF-R, since a Met/HGF-R-specific short hairpin RNA (shRNA) inhibited both activation of transduction pathways and motility triggered by MAb DO-24. The protective antiapoptotic effect of MAb DO-24 was dependent on activation of the ras-MAPK Erk1/2 and phosphatidylinositol 3-kinase (PI3-kinase)-Akt transduction pathways, since it was abrogated by treatments with their specific pharmacological inhibitors, PD-98059 and wortmannin. Moreover, the MAb induced a motogenic, but not mitogenic, response in these cells, mimicking in all aspects the natural ligand HGF but displaying a significant higher stability than HGF in culture. This MAb may thus be a valuable substitute for HGF, being more easily available in a biologically active, highly stable, and purified form.
- Published
- 2010
- Full Text
- View/download PDF
36. Facilitated tendon-bone healing by local delivery of recombinant hepatocyte growth factor in rabbits.
- Author
-
Nakase J, Kitaoka K, Matsumoto K, and Tomita K
- Subjects
- Animals, Bone Remodeling drug effects, Bone Remodeling physiology, Calcification, Physiologic drug effects, Chondrocytes ultrastructure, Drug Evaluation, Fibrocartilage ultrastructure, Hepatocyte Growth Factor administration & dosage, Hepatocyte Growth Factor pharmacology, Peptide Fragments administration & dosage, Peptide Fragments pharmacology, Peptide Fragments therapeutic use, Proto-Oncogene Proteins c-met drug effects, Rabbits, Recombinant Proteins administration & dosage, Recombinant Proteins pharmacology, Recombinant Proteins therapeutic use, Regeneration drug effects, Tendon Transfer, Tendons ultrastructure, Tensile Strength, Weight-Bearing, Anterior Cruciate Ligament surgery, Hepatocyte Growth Factor therapeutic use, Plastic Surgery Procedures, Tendons surgery, Tibia surgery, Wound Healing drug effects
- Abstract
Purpose: This study was performed to evaluate the therapeutic effect of hepatocyte growth factor (HGF) on tendon-bone healing in a rabbit model., Methods: In adult rabbits the long digital extensor tendon was detached from the lateral femoral condyle, and the free end of the tendon was inserted into a tunnel drilled into the proximal tibial metaphysis. Cancellous bone obtained during drilling of the tibial hole was soaked in saline solution or solution containing 100-microg/mL human recombinant HGF and then transplanted into the bone tunnel. Junctional healing between the tendon and the bone was evaluated by histologic analysis and uniaxial load-to-failure testing at 2, 4, 6, 8, and 12 weeks after surgery., Results: In the saline solution-treated control group, Sharpey-like fibers, which connected the tendon graft and the bone tissue, appeared 6 weeks after treatment. At 8 weeks after treatment, maturation of lamellar bone was seen, and at 12 weeks, the adhesion between tendon and bone appeared to be supported by indirect insertion of fibrocartilaginous tissue, wherein the border between the fibrocartilaginous tissue and tendon or bone was significant. In the HGF-treated group, the fibrous tissues were parallel to the load axis, and lamellar bone and Sharpey-like fibers appeared as early as 4 weeks after treatment. At 12 weeks, junctional tissue, characterized by a continuous 4-layer structure of bone, calcified cartilage, fibrocartilage, and tendon, was regenerated by a direct insertion. On biomechanical testing, the HGF-treated group had significantly better biomechanical properties than the control group at 2 and 4 weeks. The histologic improvement caused by HGF treatment was associated with the biomechanical improvement., Conclusions: Local administration of recombinant HGF promotes the adhesive healing process at the tendon-bone junction, both histologically and mechanically, after ligament reconstruction in a rabbit model., Clinical Relevance: Application of HGF may be considered as a new therapeutic approach to accelerate healing and rehabilitation after ligament reconstruction., ((c) 2010 Arthroscopy Association of North America. Published by Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
37. PHA665752, a small-molecule inhibitor of c-Met, inhibits hepatocyte growth factor-stimulated migration and proliferation of c-Met-positive neuroblastoma cells.
- Author
-
Crosswell HE, Dasgupta A, Alvarado CS, Watt T, Christensen JG, De P, Durden DL, and Findley HW
- Subjects
- Blotting, Western, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Humans, Hypoglycemic Agents pharmacology, MAP Kinase Signaling System drug effects, Neoplasm Staging, Neuroblastoma pathology, PTEN Phosphohydrolase metabolism, Proto-Oncogene Proteins c-met drug effects, Reverse Transcriptase Polymerase Chain Reaction, Rosiglitazone, Thiazolidinediones pharmacology, Transfection, Antineoplastic Agents pharmacology, Hepatocyte Growth Factor metabolism, Indoles pharmacology, Neuroblastoma metabolism, Proto-Oncogene Proteins c-met metabolism, Sulfones pharmacology
- Abstract
Background: c-Met is a tyrosine kinase receptor for hepatocyte growth factor/scatter factor (HGF/SF), and both c-Met and its ligand are expressed in a variety of tissues. C-Met/HGF/SF signaling is essential for normal embryogenesis, organogenesis, and tissue regeneration. Abnormal c-Met/HGF/SF signaling has been demonstrated in different tumors and linked to aggressive and metastatic tumor phenotypes. In vitro and in vivo studies have demonstrated inhibition of c-Met/HGF/SF signaling by the small-molecule inhibitor PHA665752. This study investigated c-Met and HGF expression in two neuroblastoma (NBL) cell lines and tumor tissue from patients with NBL, as well as the effects of PHA665752 on growth and motility of NBL cell lines. The effect of the tumor suppressor protein PTEN on migration and proliferation of tumor cells treated with PHA665752 was also evaluated., Methods: Expression of c-Met and HGF in NBL cell lines SH-EP and SH-SY5Y and primary tumor tissue was assessed by immunohistochemistry and quantitative RT-PCR. The effect of PHA665752 on c-Met/HGF signaling involved in NBL cell proliferation and migration was evaluated in c-Met-positive cells and c-Met-transfected cells. The transwell chemotaxis assay and the MTT assay were used to measure migration and proliferation/cell-survival of tumor cells, respectively. The PPAR-gamma agonist rosiglitazone was used to assess the effect of PTEN on PHA665752-induced inhibition of NBL cell proliferation/cell-survival and migration, Results: High c-Met expression was detected in SH-EP cells and primary tumors from patients with advanced-stage disease. C-Met/HGF signaling induced both migration and proliferation of SH-EP cells. Migration and proliferation/cell-survival were inhibited by PHA665752 in a dose-dependent manner. We also found that induced overexpression of PTEN following treatment with rosiglitazone significantly enhanced the inhibitory effect of PHA665752 on NBL-cell migration and proliferation., Conclusion: c-Met is highly expressed in most tumors from patients with advanced-stage, metastatic NBL. Furthermore, using the NBL cell line SH-EP as a model, PHA665752 was shown to inhibit cMet/HGF/SF signaling in vitro, suggesting c-Met inhibitors may have efficacy for blocking local progression and/or metastatic spread of c-Met-positive NBL in vivo. These are novel findings for this disease and suggest that further studies of agents targeting the c-Met/HGF axis in NBL are warranted.
- Published
- 2009
- Full Text
- View/download PDF
38. Targeting the Met signaling pathway in renal cancer.
- Author
-
Giubellino A, Linehan WM, and Bottaro DP
- Subjects
- Animals, Carcinoma, Renal Cell mortality, Carcinoma, Renal Cell physiopathology, Drug Delivery Systems, Humans, Kidney Neoplasms mortality, Kidney Neoplasms physiopathology, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Signal Transduction drug effects, Antineoplastic Agents pharmacology, Carcinoma, Renal Cell drug therapy, Kidney Neoplasms drug therapy
- Abstract
Renal cell carcinoma (RCC), the most common form of kidney cancer, accounts for 3% of all adult malignancies and its incidence has significantly increased over the last 20 years. RCC claims 13,000 lives annually in the USA and more than 100,000 worldwide. A better understanding of the molecular basis of RCC has facilitated the development of novel and more selective therapeutic approaches. An important role in RCC oncogenesis is played by the receptor for HGF, Met, which has attracted considerable attention, more recently as a molecular target for cancer therapy, and several drugs selectively targeting this pathway are now in clinical trials. This review will focus on efforts to understand the role of the Met signaling pathway in renal cancer and how this has contributed to the development of potent and selective drug candidates.
- Published
- 2009
- Full Text
- View/download PDF
39. A special key for unlocking the door to targeted therapies of breast cancer.
- Author
-
Lindemann K, Harbeck N, Lengyel E, and Resau JH
- Subjects
- Humans, Proto-Oncogene Proteins c-met genetics, Breast Neoplasms therapy, Carcinoma, Intraductal, Noninfiltrating therapy, Hepatocyte Growth Factor therapeutic use, Proto-Oncogene Proteins c-met drug effects, Receptor, ErbB-2 metabolism
- Abstract
Personalized medicine and targeted therapy is the best hope for patients with cancer. C-Met-HGF/SF inhibition may be an effective cancer treatment for ductal carcinoma of the breast as it is expected to be an important signalling target in a large number of malignancies.
- Published
- 2008
- Full Text
- View/download PDF
40. beta-Catenin and actin reorganization in HGF/SF response of ST14A cells.
- Author
-
Soldati C, Biagioni S, Poiana G, and Augusti-Tocco G
- Subjects
- Animals, Cadherins metabolism, Cell Adhesion drug effects, Cell Adhesion physiology, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Line, Cell Movement drug effects, Cell Movement physiology, Cells, Cultured, Central Nervous System cytology, Central Nervous System metabolism, Cytoskeleton metabolism, Enzyme Inhibitors pharmacology, Hepatocyte Growth Factor pharmacology, Mice, Neurons drug effects, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Rats, Stem Cells drug effects, Actins metabolism, Central Nervous System embryology, Hepatocyte Growth Factor metabolism, Neurons metabolism, Stem Cells metabolism, beta Catenin metabolism
- Abstract
Hepatocyte growth factor/scatter factor (HGF/SF) is a pleiotropic factor that activates proliferation, differentiation, and migration of various cell types. Its action is mediated by c-Met, a receptor endowed with tyrosine kinase activity that activates complex signaling cascades and mediates diverse cell responses. Although HGF action was first demonstrated in epithelial cells, expression of HGF and c-Met receptor has also been described in developing and adult mammalian brain. In the developing central nervous system, areas of HGF and c-Met expression are coincident with the migratory pathway of precursor cells. In the present article we report that the interaction between c-Met and HGF/SF in striatal progenitor ST14A cells triggers a signaling cascade that induces modification of cell morphology, with decreased cell-cell interactions and increased cell motility; in particular, we analyzed the reorganization of the actin cytoskeleton and the delocalization of beta-catenin and N-cadherin. The testing of other neurotrophic factors (NGF, BDNF, NT3, and CNTF) showed that the observed modifications were peculiar to HGF. We show that phosphoinositide 3-kinase inhibitor treatment, which blocks cell scattering induced by HGF/SF, does not abolish actin and beta-catenin redistribution. The effects of HGF/SF on primary spinal cord cell cultures were also investigated, and HGF/SF was found to have a possible motogenic effect on these cells. The data reported suggest that HGF could play a role in the early steps of neurogenesis as a motogenic factor.
- Published
- 2008
- Full Text
- View/download PDF
41. An in vivo model of Met-driven lymphoma as a tool to explore the therapeutic potential of Met inhibitors.
- Author
-
Accornero P, Lattanzio G, Mangano T, Chiarle R, Taulli R, Bersani F, Forni PE, Miretti S, Scuoppo C, Dastrù W, Christensen JG, Crepaldi T, and Ponzetto C
- Subjects
- Animals, Blotting, Western, Gene Transfer Techniques, Humans, Immunohistochemistry, Indoles pharmacology, Lymphoma pathology, Mice, Mice, Transgenic, Proto-Oncogene Proteins c-met drug effects, Sulfones pharmacology, Disease Models, Animal, Lymphoma drug therapy, Lymphoma genetics, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met genetics
- Abstract
Purpose: Met, the tyrosine kinase receptor for hepatocyte growth factor, is frequently deregulated in human cancer. Recent evidence indicates that Met amplification may confer resistance to treatments directed toward other receptor tyrosine kinases. Thus, there is a need to develop Met inhibitors into therapeutic tools, to be used alone or in combination with other molecularly targeted drugs. Preclinical validation of Met inhibitors has thus far been done in nude mice bearing cancer cells xenografts. A far superior model would be a transgenic line developing spontaneous Met-driven tumors with high penetrance and short latency., Experimental Design: To this end, we introduced into the mouse genome TPR-MET, the oncogenic form of MET. The Tpr-Met protein ensures deregulation of Met signaling because dimerization motifs in the Tpr moiety cause ligand-independent activation of the Met kinase., Results: Here, we describe a TPR-MET transgenic line that develops thymic T-cell lymphoma with full penetrance and very short latency. In the tumors, Tpr-Met and its effectors were phosphorylated. Treatment of tumor-derived T lymphocytes with the selective Met inhibitor PHA-665752 at nanomolar concentrations abolished phosphorylation of Met and downstream effectors and led to caspase-mediated apoptosis. I.v. administration of PHA-665752 to transgenic mice bearing lymphomas in exponential growth phase led to a significant decrease in tumor growth and, in some cases, to tumor regression., Conclusions: Our transgenic line, which within 2 months reliably develops Tpr-Met-driven T-cell lymphoma, represents a valuable tool to explore the efficacy and therapeutic potential of Met kinase inhibitors as anticancer drugs.
- Published
- 2008
- Full Text
- View/download PDF
42. Hepatocyte growth factor-modulated rat Leydig cell functions.
- Author
-
Del Bravo J, Catizone A, Ricci G, and Galdieri M
- Subjects
- Animals, Apoptosis drug effects, Cell Culture Techniques, Culture Media, In Situ Nick-End Labeling, Leydig Cells drug effects, Male, Organ Culture Techniques, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics, RNA genetics, RNA isolation & purification, Rats, Rats, Wistar, Reverse Transcriptase Polymerase Chain Reaction, Testis drug effects, Testis physiology, Testosterone metabolism, Hepatocyte Growth Factor pharmacology, Leydig Cells physiology
- Abstract
Hepatocyte growth factor (HGF) regulates many cellular functions acting through c-Met, its specific tyrosine kinase receptor. We previously reported that in prepuberal rats HGF is secreted by the peritubular myoid cells during the entire postnatal testicular development and by the Sertoli cells only at puberty. We have also demonstrated that germ cells at different stages of development express c-Met and that HGF modulates germ cell proliferation and apoptosis. In the present article, we extend our study to the interstitial compartment of the testis and demonstrate that the c-Met protein is present on Leydig cells. The receptor is functionally active as demonstrated by the detected effects of HGF. We report in this article that HGF significantly increases the amount of testosterone secreted by the Leydig cells and decreases the number of Leydig cells undergoing apoptosis. The antiapoptotic effect of HGF is mediated by caspase-3 activity because the amount of the active fragment of the enzyme is decreased in Leydig cells cultured in the presence of HGF. However, treatment with the growth factor does not modify the expression levels of caspase-3 mRNA. These data indicate that HGF regulates the functional activities of Leydig cells. Interestingly, the steroidogenetic activity of the cells is increased by HGF in cultured explants of testicular tissues as well as the antiapoptotic effect of HGF. Therefore, our data indicate that HGF has a crucial role in the regulation of male fertility.
- Published
- 2007
- Full Text
- View/download PDF
43. Fragments 2007--what has fragment-based drug discovery delivered for medicinal chemistry? 7 March 2007, Cambridge, UK.
- Author
-
Norman P
- Subjects
- Amyloid Precursor Protein Secretases antagonists & inhibitors, Aspartic Acid Endopeptidases antagonists & inhibitors, Crystallography, X-Ray, Drug Evaluation, Preclinical, Enzyme Inhibitors pharmacology, Fusion Proteins, bcr-abl antagonists & inhibitors, Glycine analogs & derivatives, Glycine chemistry, Humans, Ligands, Magnetic Resonance Spectroscopy, Piperazines chemistry, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism, Quantitative Structure-Activity Relationship, Drug Delivery Systems, Drug Design, Drugs, Investigational
- Published
- 2007
44. Review of clinic trials: agents targeting c-Met.
- Author
-
Abidoye O, Murukurthy N, and Salgia R
- Subjects
- Clinical Trials as Topic, Humans, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Neoplasms metabolism, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met metabolism
- Abstract
Receptor tyrosine kinases are a group of molecules that can enhance cellular proliferation, cell motility and migration, and eventual metastasis. c-Met receptor tyrosine kinase has a significant biological and biochemical effect on cancer cells, and appears to be an important therapeutic target. In many cancers, c-Met (which can be activated by its ligand hepatocyte growth factor, HGF) can be overexpressed, activated, amplified, and/or mutated. The mutations of c-Met had initially been described in the tyrosine kinase domain, and we have described them in other "hot-spots" such as the juxtamembrane and semaphorin domains. Targeting c-Met has been very fruitful pre-clinically, and currently, there are several clinical trials for advanced cancers. Described in this review are some of the biological and biochemical aspects of c-Met, and detailed are a number of therapeutic strategies. With our understanding of c-Met biology and role in cancer, we should be able to arrive at a unique strategy to eradicate cancers in which c-Met plays a significant role.
- Published
- 2007
- Full Text
- View/download PDF
45. c-Met is a potentially new therapeutic target for treatment of human melanoma.
- Author
-
Puri N, Ahmed S, Janamanchi V, Tretiakova M, Zumba O, Krausz T, Jagadeeswaran R, and Salgia R
- Subjects
- Amino Acid Sequence, Base Sequence, Cell Differentiation drug effects, Cell Line, Tumor, Fluorescent Antibody Technique, Hepatocyte Growth Factor metabolism, Humans, Immunohistochemistry, Melanoma metabolism, Microphthalmia-Associated Transcription Factor, Molecular Sequence Data, Mutation, Polymerase Chain Reaction, Proto-Oncogene Proteins c-met drug effects, Proto-Oncogene Proteins c-met genetics, RNA, Small Interfering, Reactive Oxygen Species metabolism, Skin Neoplasms metabolism, Transfection, Indoles pharmacology, Melanoma drug therapy, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met metabolism, Skin Neoplasms drug therapy, Sulfonamides pharmacology
- Abstract
Purpose: c-Met is a receptor tyrosine kinase involved in cell growth, invasion, metastases, and angiogenesis. In this study, we investigated the role of c-Met in melanoma biology using a novel small-molecule tyrosine kinase inhibitor SU11274 and small interfering (si) RNA against the receptor., Experimental Design: The effects of SU11274 and c-Met siRNA were studied on proliferation, apoptosis, differentiation, reactive oxygen species, and intracellular signaling. c-Met mutations were examined, and the expression of c-Met and activated c-Met was studied in nevi, primary, and metastatic melanoma., Results: c-Met was expressed in 6:7 melanoma cell lines by immunoblotting. SU11274 inhibited cell growth in all melanoma cell lines by 85% to 98% with an IC(50) between 1 and 2.5 mumol/L and caused apoptosis (12-58%) in five out of six cell lines. siRNA against c-Met inhibited proliferation of melanoma cells by 60%. This is the first study that shows that SU11274 and siRNA induced microphthalmia-associated transcription factor (MITF) and several other melanoma differentiation proteins and a morphologically differentiated phenotype. SU11274 also inhibited reactive oxygen species formation and phosphorylation of c-Met receptor, AKT and S-6 kinase by the hepatocyte growth factor. A new missense c-Met mutation N948S was identified in cell lines and R988C in tumor tissue in the juxtamembrane domain of c-Met. It was found that c-Met was expressed in 88% of melanomas and 15% of nevi, and that c-Met (pY1003) was activated in 21% of human melanomas., Conclusion: These results support the role of c-Met in proliferation, apoptosis, differentiation, and tumor progression of melanoma. SU11274 could be used in the therapeutic inhibition of melanoma.
- Published
- 2007
- Full Text
- View/download PDF
46. A Hepatocyte Growth Factor (HGF)/receptor autocrine loop regulates constitutive self-renewal of human periodontal ligament cells but reduces sensitivity to exogenous HGF.
- Author
-
Kawase T, Okuda K, and Yoshie H
- Subjects
- Adolescent, Autocrine Communication drug effects, Cell Membrane chemistry, Cell Proliferation drug effects, Cells, Cultured, Child, Cytoplasm chemistry, DNA biosynthesis, Down-Regulation, Female, Hepatocyte Growth Factor analysis, Hepatocyte Growth Factor pharmacology, Humans, Male, Mitogen-Activated Protein Kinases analysis, Mitogens pharmacology, Periodontal Ligament physiology, Phosphorylation, Proto-Oncogene Proteins c-met analysis, Proto-Oncogene Proteins c-met drug effects, Autocrine Communication physiology, Hepatocyte Growth Factor physiology, Periodontal Ligament cytology, Proto-Oncogene Proteins c-met physiology
- Abstract
Background: In addition to its prominent role in liver regeneration, hepatocyte growth factor (HGF) is now generally thought to be produced by mesenchymal cells to promote the regeneration of epithelial tissue by a paracrine mechanism. However, it is not known how or if HGF could be involved in the regeneration of periodontal tissues. The purpose of this study was to characterize the ability of normal human periodontal ligament (PDL) cells to produce or respond to HGF., Methods: PDL cells derived from healthy young volunteers were used from passages four through 10. HGF receptors were detected both by immunocytochemical staining and Western-blotting analysis. Both DNA synthesis (by bromo-deoxyuridine [BrdU]-incorporation) and secreted HGF were quantified by enzyme-linked immunosorbent assays. Mitogen-activated protein kinase (MAPK) phosphorylation was also analyzed by Western blot., Results: Despite the immunocytochemical demonstration of HGF receptor protein in the cytoplasm and on the plasma membrane of PDL cells, exogenous recombinant human HGF did not exert the mitogenic effects expected. As reported for other mesenchymal cells, PDL cells were found to secrete HGF. Treatments with neutralizing anti-HGF antibody significantly suppressed constitutive PDL cell proliferation and sustained the receptor protein at higher levels than in non-treated cells. Under these conditions, exogenous HGF rapidly phosphorylated extracellular signal-regulated kinase (ERK), an action linked to the cell proliferation and downregulation of cell-surface receptors., Conclusions: Unlike other known mesenchymal or epithelial cells, these findings suggest that normal PDL cells from young donors possess a constitutive HGF/receptor autocrine loop that normally regulates their replacement self-proliferation but reduces sensitivity to exogenously applied HGF by acute receptor downregulation.
- Published
- 2006
- Full Text
- View/download PDF
47. Hepatocyte growth factor/c-met signaling in regulating urokinase plasminogen activator in human stomach cancer: A potential therapeutic target for human stomach cancer.
- Author
-
Lee KH, Choi EY, Hyun MS, Jang BI, Kim TN, Kim SW, Song SK, Kim JH, and Kim JR
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma enzymology, Disease Progression, Humans, In Vitro Techniques, Neoplasm Metastasis, Stomach Neoplasms drug therapy, Urokinase-Type Plasminogen Activator antagonists & inhibitors, Hepatocyte Growth Factor metabolism, Proto-Oncogene Proteins c-met drug effects, Receptor Protein-Tyrosine Kinases drug effects, Receptors, Growth Factor drug effects, Signal Transduction drug effects, Stomach Neoplasms enzymology, Urokinase-Type Plasminogen Activator metabolism
- Abstract
Background: Up-regulation of the hepatocyte growth factor (HGF), its transmembrane tyrosine kinase receptor (c-Met), and urokinase type plasminogen activator (uPA), is associated with the development and metastasis of various types of cancers. However, the mechanisms by which HGF/c-Met signaling mediates cancer progression and metastasis are unclear., Methods: We investigated the roles of HGF/c-Met in tumor progression and metastasis in NUGC-3 and MKN-28 stomach cancer cell lines., Results: Treatment with HGF increased c-Met phosphorylation in a dose-dependent manner, as well as increasing cell proliferation. HGF treatment also increased the protein level and the activity of uPA in NUGC-3 and MKN-28 cells. A monoclonal antibody against human uPA receptor (uPAR), mAb 3936, inhibited HGF-mediated tumor cell invasion in a dose-dependent manner. Down-regulation of uPA using uPA-shRNA induced a decrease in in vitro cell invasion in NUGC-3 cells., Conclusions: These results suggest that NUGC-3 and MKN-28 cells express functional c-Met, which may provide a therapeutic target for interfering with metastases of cancer cells by inhibiting uPA and uPAR-mediated proteolysis.
- Published
- 2006
- Full Text
- View/download PDF
48. Neoadjuvant selective COX-2 inhibition down-regulates important oncogenic pathways in patients with esophageal adenocarcinoma.
- Author
-
Tuynman JB, Buskens CJ, Kemper K, ten Kate FJ, Offerhaus GJ, Richel DJ, and van Lanschot JJ
- Subjects
- Adenocarcinoma enzymology, Blotting, Western, Celecoxib, Cyclooxygenase 1 drug effects, Cyclooxygenase 2 drug effects, Down-Regulation, Esophageal Neoplasms enzymology, Humans, Immunohistochemistry, In Vitro Techniques, Neoadjuvant Therapy, Proto-Oncogene Proteins c-met drug effects, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured drug effects, Adenocarcinoma drug therapy, Cyclooxygenase Inhibitors pharmacology, Cyclooxygenase Inhibitors therapeutic use, Esophageal Neoplasms drug therapy, Pyrazoles pharmacology, Pyrazoles therapeutic use, Sulfonamides pharmacology, Sulfonamides therapeutic use
- Abstract
Objectives: To evaluate the effects of neoadjuvant therapy with the selective cyclooxygenase-2 (COX-2) inhibitor celecoxib in vitro and in patients with esophageal adenocarcinoma on COX-2 and MET expression., Summary Background Data: High COX-2 and/or MET expression levels are negative prognostic factors for adenocarcinoma of the esophagus. Nonsteroidal anti-inflammatory drugs (NSAIDs) and selective COX-2 inhibitors exert anticancer mechanisms as is evident from epidemiologic studies and from experimental models for esophageal cancer. The mechanisms and the significance of these findings in patients with adenocarcinoma of the esophagus are unknown., Methods: Esophageal adenocarcinoma cell lines were used to asses the effects in vitro. To study the clinical effects 12 patients with esophageal adenocarcinoma were included for neoadjuvant treatment (4 weeks) with celecoxib at 400 mg twice daily. Fifteen patients not receiving NSAIDs or celecoxib were included as a control. Effects were evaluated using the MTT-cell viability test, Western blot analysis, immunohistochemistry, and RT-PCR., Results: In vitro celecoxib administration resulted in decreased cell viability, increased apoptosis, and decreased COX-2 and MET expression levels. In patients, neoadjuvant treatment with celecoxib significantly down-regulated COX-2 and MET expression in the tumor when compared with the nontreated control group and when compared with pretreatment measurements., Conclusions: This is the first study to show in vitro and in patients with esophageal adenocarcinoma that selective COX-2 inhibition down-regulates COX-2 and MET expression, both important proteins involved in cancer progression and dissemination. Therefore, (neo)adjuvant therapy with celecoxib might have clinical potential for patients with esophageal adenocarcinoma.
- Published
- 2005
- Full Text
- View/download PDF
49. Possible role of hepatocyte growth factor in regeneration of human peritoneal mesothelial cells.
- Author
-
Naiki Y, Matsuo K, Matsuoka T, and Maeda Y
- Subjects
- Cell Proliferation, Cells, Cultured, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Glucose Solution, Hypertonic, Humans, Matrix Metalloproteinase 2 drug effects, Peritoneal Dialysis, Continuous Ambulatory, Proto-Oncogene Proteins c-met drug effects, Tissue Inhibitor of Metalloproteinase-2 drug effects, Transforming Growth Factor beta pharmacology, Transforming Growth Factor beta1, Epithelial Cells physiology, Hepatocyte Growth Factor pharmacology, Peritoneum cytology, Regeneration
- Abstract
Human peritoneal mesothelial cells (HPMCs) play an important role in peritoneal functions. During long term peritoneal dialysis, it has been reported that HPMCs are damaged by high glucose solution via the signal of transforming growth factor (TGF)-beta1 produced by HPMCs. In this study, we focused on the effect of hepatocyte growth factor (HGF), known as an anti-fibrotic and anti-TGF-beta1 agent, on HPMCs damaged by high glucose solution. HPMCs were isolated from specimens of the omentum from nonuremic patients after informed consent had been obtained. After confirming adhesion for 6 hours, 100 microL of DMEM with 0.5%FCS were added at different concentrations (D-glucose; 6, 30 mM) with or without HGF (10, 30, 100 ng/mL) for 48 hours. We examined the effects of a high concentration of glucose and then focused on following four critical points: 1) the production of HGF from HPMCs exposed to a high concentration of glucose, 2) the expression of c-Met on HPMCs, 3) the viability of those cells, and 4) matrix metalloproteinase-2 (MMP-2) and tissue inhibitors of metalloproteinase-2 (TIMP-2). The following significant changes are described herein: high glucose solution and TGF-beta1 i) decreased HGF production from HPMCs and ii) up-regulated expression of c-Met on HPMCs, and addition of HGF iii) restored viability of HPMCs damaged by glucose, iv) suppressed TGF-beta1 production by HGF, and v) induced up-regulation of MMP-2 and decreased TIMP-2 production by HPMCs. Levels of HGF decreased by high concentrations of glucose in the peritoneal cavity may induce the loss of HPMCs and thereby result in peritoneal fibrosis. These results suggest that HGF is an effective agent in the regeneration of peritoneal membrane damaged by high glucose solution.
- Published
- 2005
- Full Text
- View/download PDF
50. [Experimental inhibition of SF factor/HGF linkage to its receptor in the treatment of metastatic dissemination...].
- Subjects
- Animals, Humans, Hepatocyte Growth Factor antagonists & inhibitors, Neoplasm Metastasis drug therapy, Proto-Oncogene Proteins c-met drug effects
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.