Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 13(3): 576-84, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24327519

ABSTRACT

Small cell lung cancer (SCLC) is a devastating disease, and current therapies have not greatly improved the 5-year survival rates. Topoisomerase (Top) inhibition is a treatment modality for SCLC; however, the response is short lived. Consequently, our research has focused on improving SCLC therapeutics through the identification of novel targets. Previously, we identified MNNG HOS transforming gene (MET) to be overexpressed and functional in SCLC. Herein, we investigated the therapeutic potential of combinatorial targeting of MET using SU11274 and Top1 using 7-ethyl-10-hydroxycamptothecin (SN-38). MET and TOP1 gene copy numbers and protein expression were determined in 29 patients with limited (n = 11) and extensive (n = 18) disease. MET gene copy number was significantly increased (>6 copies) in extensive disease compared with limited disease (P = 0.015). Similar TOP1 gene copy numbers were detected in limited and extensive disease. Immunohistochemical staining revealed a significantly higher Top1 nuclear expression in extensive (0.93) versus limited (0.15) disease (P = 0.04). Interestingly, a significant positive correlation was detected between MET gene copy number and Top1 nuclear expression (r = 0.5). In vitro stimulation of H82 cells revealed hepatocyte growth factor (HGF)-induced nuclear colocalization of p-MET and Top1. Furthermore, activation of the HGF/MET axis enhanced Top1 activity, which was abrogated by SU11274. Combination of SN-38 with SU11274 dramatically decreased SCLC growth as compared with either drug alone. Collectively, these findings suggest that the combinatorial inhibition of MET and Top1 is a potentially efficacious treatment strategy for SCLC.


Subject(s)
Carcinoma, Small Cell/drug therapy , DNA Topoisomerases, Type I/genetics , Proto-Oncogene Proteins c-met/genetics , Small Cell Lung Carcinoma/drug therapy , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Dosage/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/administration & dosage , Irinotecan , Middle Aged , Piperazines/administration & dosage , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Signal Transduction/drug effects , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Sulfonamides/administration & dosage , Topoisomerase I Inhibitors/administration & dosage
2.
Cancer Res ; 73(1): 184-94, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23100466

ABSTRACT

Esophageal cancer incidence is increasing and has few treatment options. In studying receptor tyrosine kinases associated with esophageal cancers, we have identified EPHB4 to be robustly overexpressed in cell lines and primary tumor tissues. In total, 94 squamous cell carcinoma, 82 adenocarcinoma, 25 dysplasia, 13 Barrett esophagus, and 25 adjacent or unrelated normal esophageal tissues were evaluated by immunohistochemistry. EPHB4 expression was significantly higher in all the different histologic categories than in adjacent normal tissues. In 13 esophageal cancer cell lines, 3 of the 9 SCC cell lines and 2 of the 4 adenocarcinomas expressed very high levels of EPHB4. An increased gene copy number ranging from 4 to 20 copies was identified in a subset of the overexpressing patient samples and cell lines. We have developed a novel 4-nitroquinoline 1-oxide (4-NQO)-induced mouse model of esophageal cancer that recapitulates the EPHB4 expression in humans. A specific small-molecule inhibitor of EPHB4 decreased cell viability in a time- and dose-dependent manner in 3 of the 4 cell lines tested. The small-molecule inhibitor and an EPHB4 siRNA also decreased cell migration (12%-40% closure in treated vs. 60%-80% in untreated), with decreased phosphorylation of various tyrosyl-containing proteins, EphB4, and its downstream target p125FAK. Finally, in a xenograft tumor model, an EPHB4 inhibitor abrogated tumor growth by approximately 60% compared with untreated control. EphB4 is robustly expressed and potentially serves as a novel biomarker for targeted therapy in esophageal cancers.


Subject(s)
Biomarkers, Tumor/analysis , Esophageal Neoplasms/enzymology , Receptor, EphB4/biosynthesis , Adenocarcinoma/enzymology , Animals , Barrett Esophagus/enzymology , Carcinoma, Squamous Cell/enzymology , Cell Line, Tumor , Disease Models, Animal , Gene Dosage , Humans , Immunoblotting , Immunohistochemistry , Mice , Real-Time Polymerase Chain Reaction , Receptor, EphB4/analysis , Tissue Array Analysis , Xenograft Model Antitumor Assays
3.
Cancer Biol Ther ; 12(1): 9-46, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21543897

ABSTRACT

RON (MST1R) is one of two members of the MET receptor tyrosine kinase family, along with parent receptor MET. RON has a putative role in several cancers, but its expression and function is poorly characterized in gastroesophageal adenocarcinoma. A recognized functional role of MET tyrosine kinase in gastroesophageal cancer has led to early phase clinical trials using MET inhibitors, with unimpressive results. Therefore, the role of RON in gastroesophageal cancer, as well as its role in cooperative signaling with MET and as a mechanism of resistance to MET inhibition, was studied in gastroesophageal tissues and cell lines. By IHC, RON was highly over-expressed in 74% of gastroesophageal samples (n=94), and over-expression was prognostic of poor survival (p=0.008); RON and MET co-expression occurred in 43% of samples and was prognostic of worst survival (p=0.03). High MST1R gene copy number by quantitative polymerase chain reaction, and confirmed by fluorescence in situ hybridization and/or array comparative genomic hybridization, was seen in 35.5% (16/45) of cases. High MST1R gene copy number correlated with poor survival (p=0.01), and was associated with high MET and ERBB2 gene copy number. A novel somatic MST1R juxtamembrane mutation R1018G was found in 11% of samples. RON signaling was functional in cell lines, activating downstream effector STAT3, and resulted in increased viability over controls. RON and MET co-stimulation assays led to enhanced malignant phenotypes over stimulation of either receptor alone. Growth inhibition as evidenced by viability and apoptosis assays was optimal using novel blocking monoclonal antibodies to both RON and MET, versus either alone. SU11274, a classic MET small molecule tyrosine kinase inhibitor, blocked signaling of both receptors, and proved synergistic when combined with STAT3 inhibition (combination index < 1). These preclinical studies define RON as an important novel prognostic marker and therapeutic target for gastroesophageal cancer warranting further investigation.


Subject(s)
Adenocarcinoma/genetics , Esophageal Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Cell Line, Tumor , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Gene Dosage , Hepatocyte Growth Factor/metabolism , Humans , In Situ Hybridization, Fluorescence , Indoles/pharmacology , Mutation , Piperazines/pharmacology , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Small Interfering , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/genetics , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Sulfonamides/pharmacology
4.
Clin Cancer Res ; 17(10): 3112-22, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21364031

ABSTRACT

PURPOSE: Fyn is a member of the Src family of kinases that we have previously shown to be overexpressed in prostate cancer. This study defines the biological impact of Fyn inhibition in cancer using a PC3 prostate cancer model. EXPERIMENTAL DESIGN: Fyn expression was suppressed in PC3 cells using an shRNA against Fyn (PC3/FYN-). Knockdown cells were characterized using standard growth curves and time-lapse video microscopy of wound assays and Dunn Chamber assays. Tissue microarray analysis was used to verify the physiologic relevance of the HGF/MET axis in human samples. Flank injections of nude mice were performed to assess in vivo growth characteristics. RESULTS: HGF was found to be sufficient to drive Fyn-mediated events. Compared to control transductants (PC3/Ctrl), PC3/FYN- showed a 21% decrease in growth at 4 days (P = 0.05). PC3/FYN- cells were 34% longer than control cells (P = 0.018) with 50% increase in overall surface area (P < 0.001). Furthermore, when placed in a gradient of HGF, PC3/FYN- cells showed impaired directed chemotaxis down an HGF gradient in comparison to PC3/Ctrl (P = 0.001) despite a 41% increase in cellular movement speed. In vivo studies showed 66% difference of PC3/FYN- cell growth at 8 weeks using bidimensional measurements (P = 0.002). CONCLUSIONS: Fyn plays an important role in prostate cancer biology by facilitating cellular growth and by regulating directed chemotaxis-a key component of metastasis. This finding bears particular translational importance when studying the effect of Fyn inhibition in human subjects.


Subject(s)
Cell Shape , Hepatocyte Growth Factor/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-fyn/physiology , Proto-Oncogene Proteins c-met/metabolism , Receptors, Growth Factor/metabolism , Tropism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Shape/drug effects , Cell Shape/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Growth Factor/genetics , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Proto-Oncogene Proteins c-met/genetics , RNA, Small Interfering/pharmacology , Receptors, Growth Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Tropism/drug effects , Tropism/genetics , Xenograft Model Antitumor Assays
5.
J Carcinog ; 8: 15, 2009.
Article in English | MEDLINE | ID: mdl-19955662

ABSTRACT

BACKGROUND: Treatment of non-small cell lung cancer (NSCLC) remains a difficult task in oncology. Targeted inhibition of oncogenic proteins is promising. In this study, we evaluate the expression of MET and PKCbeta and in vitro effects of their inhibition using SU11274 and enzastaurin (LY317615.HCl) respectively. MATERIALS AND METHODS: Patient samples were analyzed by immunohistochemistry for expression of PKCbeta and MET, utilizing tissue microarrays under an IRB-approved protocol. Expression of PKCbeta and MET was evaluated in cell lines by immunoblotting. Treatment with SU1174 against MET and enzastaurin against PKCbeta was performed in H1993 and H358 cell lines, and cell proliferation and downstream signaling (phosphorylation of MET, AKT, FAK, and GSK3beta) were evaluated by immunoblotting. Statistical analysis was performed using SPSS 16.0. RESULTS: Expression of MET positively correlated with lymph node metastases (p=.0004), whereas PKCbeta showed no correlation (p=0.204). MET and PKCbeta expression were also strongly correlated (p<0.001). Expression of MET was observed in 5/8 cell lines (H358, H1703, A549, H1993, H2170; absent from H522, H661, or SW1573), whereas PKCbeta expression was observed in 8/8 cell lines. Cell proliferation was significantly impaired by treatment with SU11274 and enzastaurin, and their effects were synergistic in combination (CI=0.32 and 0.09). Phosphorylation of MET, FAK, AKT, and GSK3beta were strongly inhibited with both agents in combination. CONCLUSIONS: Concomitant inhibition of MET and PKCbeta significantly increased cytotoxicity in vitro against NSCLC, disrupting important downstream signaling pathways. Further evaluation in animal models is warranted.

6.
J Environ Pathol Toxicol Oncol ; 28(2): 89-98, 2009.
Article in English | MEDLINE | ID: mdl-19817696

ABSTRACT

Gene copy number analysis for some of the important molecules in lung tumorogenesis, such as MET, hepatocyte growth factor [(HGF), ligand for MET), epidermal growth factor receptor (EGFR), and paxillin (PXN), is likely to determine both the type of treatment and prognosis. Formalin-fixed paraffin-embedded (FFPE) archival tumor tissue samples are an excellent source for determining key molecular changes in the OncoGenome; however, existing extraction procedures yield relatively poor quality genomic DNA fragments. Although FISH is the method of choice for determining amplification of a gene, a more rapid quantitative poly-merase chain reaction (qPCR) technique to determine gene copy number can be used when reasonably good quality genomic DNA is available. We report here a relatively rapid method based on microwave/chelex-100 treatment that gives rise to genomic DNA fragments ranging from 1 to 12 Kb and beyond, thereby attesting to its superior quality. Genomic PCR for beta-globin gene gave reliable and reproducible results. The number of steps for extracting the DNA was kept to a minimum, and instead of precipitating the DNA, we preserved the genomic DNA extracts so as to prevent a loss in DNA yield. We found the extracts to be stable and amenable to qPCR and mutational analysis. Using lung adenocarcinoma FFPE samples and cell lines derived from lung adenocarcinomas, we demonstrated that the gene copy number for MET in lung adenocarcinoma tissue samples was preferentially increased over EGFR, HGF, and PXN and that it positively correlated with a better prognosis. In contrast, the genomic DNA extracted from 25 NSCLC cell lines gave a relatively higher gene copy number for all four genes evaluated. Our results indicate that the microwave/chelex-100-based method yields good-quality genomic DNA extracts that can be used for complex DNA analysis, such as determination of gene copy number. In addition, our data demonstrated that the adenocarcinoma cell lines potentially evolved under ex vivo conditions, and therefore, in genetic studies it is imperative to use primary tumors for generalized conclusions about lung tumors.


Subject(s)
Adenocarcinoma/genetics , DNA, Neoplasm/genetics , ErbB Receptors/genetics , Gene Dosage , Hepatocyte Growth Factor/genetics , Lung Neoplasms/genetics , Paxillin/genetics , Proto-Oncogene Proteins c-met/genetics , Adenocarcinoma/pathology , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Lung Neoplasms/pathology , Polymerase Chain Reaction , Prognosis
7.
Cancer Res ; 69(7): 3021-31, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318576

ABSTRACT

Recurrent/metastatic head and neck cancer remains a devastating disease with insufficient treatment options. We investigated the MET receptor tyrosine kinase as a novel target for the treatment of head and neck squamous cell carcinoma (HNSCC). MET/phosphorylated MET and HGF expression was analyzed in 121 tissues (HNSCC/normal) by immunohistochemistry, and in 20 HNSCC cell lines by immunoblotting. The effects of MET inhibition using small interfering RNA/two small-molecule inhibitors (SU11274/PF-2341066) on signaling, migration, viability, and angiogenesis were determined. The complete MET gene was sequenced in 66 head and neck cancer tissue samples and eight cell lines. MET gene copy number was determined in 14 cell lines and 23 tumor tissues. Drug combinations of SU11274 with cisplatin or erlotinib were tested in SCC35/HN5 cell lines. Eighty-four percent of the HNSCC samples showed MET overexpression, whereas 18 of 20 HNSCC cell lines (90%) expressed MET. HGF overexpression was present in 45% of HNSCC. MET inhibition with SU11274/PF-2341066 abrogated MET signaling, cell viability, motility/migration in vitro, and tumor angiogenesis in vivo. Mutational analysis of 66 tumor tissues and 8 cell lines identified novel mutations in the semaphorin (T230M/E168D/N375S), juxtamembrane (T1010I/R988C), and tyrosine kinase (T1275I/V1333I) domains (incidence: 13.5%). Increased MET gene copy number was present with >10 copies in 3 of 23 (13%) tumor tissues. A greater-than-additive inhibition of cell growth was observed when combining a MET inhibitor with cisplatin or erlotinib and synergy may be mediated via erbB3/AKT signaling. MET is functionally important in HNSCC with prominent overexpression, increased gene copy number, and mutations. MET inhibition abrogated MET functions, including proliferation, migration/motility, and angiogenesis. MET is a promising, novel target for HNSCC and combination approaches with cisplatin or EGFR inhibitors should be explored.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Head and Neck Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cisplatin/administration & dosage , ErbB Receptors/antagonists & inhibitors , Gene Dosage , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Immunohistochemistry , Indoles/administration & dosage , Indoles/pharmacology , Mice , Mice, Nude , Mutation , Piperazines/administration & dosage , Piperazines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Structure, Tertiary , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met , RNA, Small Interfering/genetics , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...