Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Sci Signal ; 9(450): rs12, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27811184

ABSTRACT

Patients with lung cancers harboring anaplastic lymphoma kinase (ALK) gene fusions benefit from treatment with ALK inhibitors, but acquired resistance inevitably arises. A better understanding of proximal ALK signaling mechanisms may identify sensitizers to ALK inhibitors that disrupt the balance between prosurvival and proapoptotic effector signals. Using affinity purification coupled with mass spectrometry in an ALK fusion lung cancer cell line (H3122), we generated an ALK signaling network and investigated signaling activity using tyrosine phosphoproteomics. We identified a network of 464 proteins composed of subnetworks with differential response to ALK inhibitors. A small hairpin RNA screen targeting 407 proteins in this network revealed 64 and 9 proteins that when knocked down sensitized cells to crizotinib and alectinib, respectively. Among these, knocking down fibroblast growth factor receptor substrate 2 (FRS2) or coiled-coil and C2 domain-containing protein 1A (CC2D1A), both scaffolding proteins, sensitized multiple ALK fusion cell lines to the ALK inhibitors crizotinib and alectinib. Collectively, our data set provides a resource that enhances our understanding of signaling and drug resistance networks consequent to ALK fusions and identifies potential targets to improve the efficacy of ALK inhibitors in patients.


Subject(s)
Carbazoles/pharmacology , Cell Cycle Proteins , Lung Neoplasms , Microtubule-Associated Proteins , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , RNA Interference , Receptor Protein-Tyrosine Kinases , Serine Endopeptidases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Anaplastic Lymphoma Kinase , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Crizotinib , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
2.
PLoS One ; 11(1): e0147344, 2016.
Article in English | MEDLINE | ID: mdl-26789630

ABSTRACT

Epithelial-mesenchymal transition (EMT) is one mechanism of acquired resistance to inhibitors of the epidermal growth factor receptor-tyrosine kinases (EGFR-TKIs) in non-small cell lung cancer (NSCLC). The precise mechanisms of EMT-related acquired resistance to EGFR-TKIs in NSCLC remain unclear. We generated erlotinib-resistant HCC4006 cells (HCC4006ER) by chronic exposure of EGFR-mutant HCC4006 cells to increasing concentrations of erlotinib. HCC4006ER cells acquired an EMT phenotype and activation of the TGF-ß/SMAD pathway, while lacking both T790M secondary EGFR mutation and MET gene amplification. We employed gene expression microarrays in HCC4006 and HCC4006ER cells to better understand the mechanism of acquired EGFR-TKI resistance with EMT. At the mRNA level, ZEB1 (TCF8), a known regulator of EMT, was >20-fold higher in HCC4006ER cells than in HCC4006 cells, and increased ZEB1 protein level was also detected. Furthermore, numerous ZEB1 responsive genes, such as CDH1 (E-cadherin), ST14, and vimentin, were coordinately regulated along with increased ZEB1 in HCC4006ER cells. We also identified ZEB1 overexpression and an EMT phenotype in several NSCLC cells and human NSCLC samples with acquired EGFR-TKI resistance. Short-interfering RNA against ZEB1 reversed the EMT phenotype and, importantly, restored erlotinib sensitivity in HCC4006ER cells. The level of micro-RNA-200c, which can negatively regulate ZEB1, was significantly reduced in HCC4006ER cells. Our results suggest that increased ZEB1 can drive EMT-related acquired resistance to EGFR-TKIs in NSCLC. Attempts should be made to explore targeting ZEB1 to resensitize TKI-resistant tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Homeodomain Proteins/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Transcription Factors/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition , ErbB Receptors/genetics , ErbB Receptors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mutation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Zinc Finger E-box-Binding Homeobox 1
3.
Cancer Res ; 74(24): 7217-7228, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25348954

ABSTRACT

DDR2 mutations occur in approximately 4% of lung squamous cell cancer (SCC) where the tyrosine kinase inhibitor dasatinib has emerged as a new therapeutic option. We found that ERK and AKT phosphorylation was weakly inhibited by dasatinib in DDR2-mutant lung SCC cells, suggesting that dasatinib inhibits survival signals distinct from other oncogenic receptor tyrosine kinases (RTK) and/or compensatory signals exist that dampen dasatinib activity. To gain better insight into dasatinib's action in these cells, we assessed altered global tyrosine phosphorylation (pY) after dasatinib exposure using a mass spectrometry-based quantitative phosphoproteomics approach. Overlaying protein-protein interaction relationships upon this dasatinib-regulated pY network revealed decreased phosphorylation of Src family kinases and their targets. Conversely, dasatinib enhanced tyrosine phosphorylation in a panel of RTK and their signaling adaptor complexes, including EGFR, MET/GAB1, and IGF1R/IRS2, implicating a RTK-driven adaptive response associated with dasatinib. To address the significance of this observation, these results were further integrated with results from a small-molecule chemical library screen. We found that dasatinib combined with MET and insulin-like growth factor receptor (IGF1R) inhibitors had a synergistic effect, and ligand stimulation of EGFR and MET rescued DDR2-mutant lung SCC cells from dasatinib-induced loss of cell viability. Importantly, we observed high levels of tyrosine-phosphorylated EGFR and MET in a panel of human lung SCC tissues harboring DDR2 mutations. Our results highlight potential RTK-driven adaptive-resistant mechanisms upon DDR2 targeting, and they suggest new, rationale cotargeting strategies for DDR2-mutant lung SCC.


Subject(s)
Lung Neoplasms/drug therapy , Neoplasms, Squamous Cell/drug therapy , Pyrimidines/administration & dosage , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Thiazoles/administration & dosage , Adaptive Immunity/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Dasatinib , Discoidin Domain Receptors , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Drug Synergism , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mutation , Neoplasms, Squamous Cell/immunology , Neoplasms, Squamous Cell/pathology , Proto-Oncogene Proteins c-ret/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Receptors, Somatomedin/antagonists & inhibitors , Signal Transduction/genetics
4.
Mol Cancer Ther ; 13(11): 2751-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25189542

ABSTRACT

A growing number of gene mutations, which are recognized as cancer drivers, can be successfully targeted with drugs. The redundant and dynamic nature of oncogenic signaling networks and complex interactions between cancer cells and the microenvironment, however, can cause drug resistance. While these challenges can be addressed by developing drug combinations or polypharmacology drugs, this benefits greatly from a detailed understanding of the proteome-wide target profiles. Using mass spectrometry-based chemical proteomics, we report the comprehensive characterization of the drug-protein interaction networks for the multikinase inhibitors dasatinib and sunitinib in primary lung cancer tissue specimens derived from patients. We observed in excess of 100 protein kinase targets plus various protein complexes involving, for instance, AMPK, TBK1 (sunitinib), and ILK (dasatinib). Importantly, comparison with lung cancer cell lines and mouse xenografts thereof showed that most targets were shared between cell lines and tissues. Several targets, however, were only present in tumor tissues. In xenografts, most of these proteins were of mouse origin suggesting that they originate from the tumor microenvironment. Furthermore, intersection with subsequent global phosphoproteomic analysis identified several activated signaling pathways. These included MAPK, immune, and integrin signaling, which were affected by these drugs in both cancer cells and the microenvironment. Thus, the combination of chemical and phosphoproteomics can generate a systems view of proteins, complexes, and signaling pathways that are simultaneously engaged by multitargeted drugs in cancer cells and the tumor microenvironment. This may allow for the design of novel anticancer therapies that concurrently target multiple tumor compartments.


Subject(s)
Lung Neoplasms/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Models, Molecular , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Proteomics , Signal Transduction , Tumor Microenvironment , Xenograft Model Antitumor Assays
5.
J Thorac Oncol ; 9(7): 974-982, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24922005

ABSTRACT

INTRODUCTION: Interleukin-6 (IL-6) can activate downstream signaling pathways in lung cancer cells, such as the STAT3 pathway, and is reported to be produced by tumor cells with activating EGFR mutations. We examined IL-6/STAT3 in lung cancer tumor tissues and the effects of siltuximab, a neutralizing antibody to human IL-6, in mouse models of lung cancer. METHODS: IL-6 and STAT3 activation levels were compared with tumor histology and presence of KRAS mutations in snap-frozen, non-small-cell lung cancer tumors. The effects of siltuximab alone or in combination with erlotinib were examined in mouse xenograft models constructed using three cell line xenograft models and one primary explant mouse model. We examined the influence of cancer-associated fibroblasts (CAFs) on tumor growth and siltuximab effects. RESULTS: IL-6 levels were higher in tumors of squamous cell versus adenocarcinoma histology and were not associated with presence of KRAS mutations. Tyrosine phosphorylation status of STAT3 did not correlate with tumor IL-6 levels. Serine phosphorylation of STAT3 was correlated with KRAS mutation status. Both tumor and stromal cells contributed to total IL-6 within tumors. Siltuximab had minimal effect as a single agent in xenografts with tumor cells alone; however, in models coadministered with CAFs, siltuximab had more potent effects on tumor inhibition. We observed no effects of combined erlotinib and siltuximab. CONCLUSIONS: IL-6 is elevated in subsets of human NSCLCs, especially with squamous cell histology. Tumors supported by stromal production of IL-6 seem to be the most vulnerable to tumor growth inhibition by siltuximab.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Lung Neoplasms/drug therapy , Adenocarcinoma/chemistry , Adenocarcinoma/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Disease Models, Animal , Erlotinib Hydrochloride , Female , Fibroblasts , Humans , Interleukin-6/analysis , Interleukin-6/immunology , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Mice , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Quinazolines/administration & dosage , STAT3 Transcription Factor/metabolism , Serine/metabolism , Tyrosine/metabolism , ras Proteins/genetics
6.
Mol Syst Biol ; 9: 705, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24189400

ABSTRACT

We hypothesized that elucidating the interactome of epidermal growth factor receptor (EGFR) forms that are mutated in lung cancer, via global analysis of protein-protein interactions, phosphorylation, and systematically perturbing the ensuing network nodes, should offer a new, more systems-level perspective of the molecular etiology. Here, we describe an EGFR interactome of 263 proteins and offer a 14-protein core network critical to the viability of multiple EGFR-mutated lung cancer cells. Cells with acquired resistance to EGFR tyrosine kinase inhibitors (TKIs) had differential dependence of the core network proteins based on the underlying molecular mechanisms of resistance. Of the 14 proteins, 9 are shown to be specifically associated with survival of EGFR-mutated lung cancer cell lines. This included EGFR, GRB2, MK12, SHC1, ARAF, CD11B, ARHG5, GLU2B, and CD11A. With the use of a drug network associated with the core network proteins, we identified two compounds, midostaurin and lestaurtinib, that could overcome drug resistance through direct EGFR inhibition when combined with erlotinib. Our results, enabled by interactome mapping, suggest new targets and combination therapies that could circumvent EGFR TKI resistance.


Subject(s)
Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Mutation , Neoplasm Proteins/metabolism , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride , Furans , Humans , Neoplasm Proteins/genetics , Phosphorylation , Protein Interaction Maps , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
7.
Eur J Cancer Prev ; 21(5): 407-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22156994

ABSTRACT

Multiple studies from independent groups find evidence for signal transducer and activator of transcription 3 (Stat3) activation in nearly 50% of lung cancers, suggesting a functional role for this target in subsets of lung cancer. On the basis of the existing evidence, we hypothesized that bioavailable curcuminoid complex may modulate lung carcinogenesis, primarily by inhibiting Stat3 activation. With the safety of this being botanically well established, the objective of these studies was to test our hypothesis in vitro and in vivo in an effort to inform the design of a phase II chemoprevention trial in former smokers. We treated non-tumor-derived, normal (but immortalized) human bronchial epithelial cells (AALE) (Lundberg et al., 2002; Pillai et al., 2011) and lung adenocarcinoma-derived cells (H441) with bioactive curcumin C3 complex. Asynchronous cells in each case were treated with curcumin for 24 h, followed by immunoblotting for Stat3 and activated Stat3-P, prior signal of which was used for normalization. We also completed a preclinical trial in which 12 mice were randomly divided into three groups and subjected to 3 days or 9 days of curcumin intraperitoneal injections, followed by analysis of lung tissues for Stat3-P changes and growth suppressive effects of the curcumin. The growth suppressive effects were measured using Cyclin D1 and the replicative helicase subunit, Mcm2, as surrogates for the proliferative capacity of the tissues. In-vitro studies with curcuminoid complex demonstrated that the activity of Stat3 in both normal bronchoepithelial cells and lung cancer-derived cells is sensitive to curcumin exposure. In a dose-dependent manner, curcumin treatment resulted in significant suppression of Stat3 phosphorylation and reduction in the proliferative capacity of both cell types. In the preclinical trial with rodent models, curcumin reduced Stat3-P and the proliferative markers CycD1 and Mcm2 in mice lung tissues in vivo. These culture and preclinical studies indicate that the activity of the Stat3 pathway can be suppressed by curcumin treatment, concomitant with a reduction in cell proliferation, supporting our hypothesis that inhibition of the Stat3 pathway represents at least one important mechanism by which curcumin elicits its effects on the bronchoepithelium. These data provide a rationale for the use of curcumin as a promising chemopreventive agent in high-risk populations such as former smokers.


Subject(s)
Curcumin/therapeutic use , Enzyme Inhibitors/therapeutic use , Lung Neoplasms/prevention & control , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cell Line , Female , Humans , Mice , Mice, Nude , Random Allocation
8.
Mol Cancer Ther ; 10(3): 481-94, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21216930

ABSTRACT

Members of the signal transducer and activator of transcription (STAT) family of transcription factors are potential targets for the treatment and prevention of cancers including non-small-cell lung cancer. STAT proteins can be phosphorylated and activated by diverse upstream kinases including cytokine receptors and tyrosine kinases. We examined STAT protein activation in lung cancer cell lines including those with activating mutations in the EGFR and examined upstream kinases responsible for STAT3 phosphorylation and activation using small molecules, antibodies, and RNA interference. We found more pronounced STAT3 activation in cells with activating EGFR mutations, yet inhibition of EGFR activity had no effect on STAT3 activation. Inhibition of JAK1 with small molecules or RNA interference resulted in loss of STAT3 tyrosine phosphorylation and inhibition of cell growth. An interleukin-6 neutralizing antibody, siltuximab (CNTO 328) could inhibit STAT3 tyrosine phosphorylation in a cell-dependent manner. Siltuximab could completely inhibit STAT3 tyrosine phosphorylation in H1650 cells, and this resulted in inhibition of lung cancer cell growth in vivo. Combined EGFR inhibition with erlotinib and siltuximab resulted in dual inhibition of both tyrosine and serine STAT3 phosphorylation, more pronounced inhibition of STAT3 transcriptional activity, and translated into combined effects on lung cancer growth in a mouse model. Our results suggest that JAK1 is responsible for STAT3 activation in lung cancer cells and that indirect attacks on JAK1-STAT3 using an IL-6 neutralizing antibody with or without EGFR inhibition can inhibit lung cancer growth in lung cancer subsets.


Subject(s)
Antibodies, Neutralizing/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Interleukin-6/immunology , Janus Kinase 1/metabolism , Lung Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cell Proliferation/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Immunoblotting , Immunoprecipitation , Interleukin-6/antagonists & inhibitors , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Mice , Mice, Nude , Mutation , Phosphorylation , RNA Interference , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Cells, Cultured
9.
J Thorac Oncol ; 5(11): 1806-14, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20881637

ABSTRACT

BACKGROUND: Targeted agents such as tyrosine kinase inhibitors have been extensively studied in preclinical systems and in advanced-stage patients. Little is known about levels of kinase inhibitors found in tumors as opposed to plasma. Similarly, effects of inhibitors on tumor signaling pathways in patient-based materials are unclear. To explore these questions, we conducted a trial of a brief course of preoperative gefitinib, an epidermal growth factor receptor (EGFR) inhibitor, in early-stage non-small cell lung cancer. METHODS: Patient with early-stage non-small cell lung cancer received 4 weeks of gefitinib 250 mg daily before surgical resection. Pre- and posttreatment computerized tomography scans and positron emission tomography scans were used to assess clinical response. Gefitinib and surgical toxicity were evaluated. Tumor tissue was evaluated for gefitinib levels and was compared with plasma gefitinib levels. Activated signaling molecules including EGFR, STAT3, ERK, and AKT were examined in surgically resected tumor tissue. RESULTS: Twenty-three patients participated in the study, and all had surgical resection of tumors. No toxicities unrelated to known effects of gefitinib or surgery were encountered. Twenty-two patients had stable disease, and one had progression in tumor size. There was no correlation with positron emission tomography response and computerized tomography response. Tumor levels of gefitinib were approximately 40-fold higher than plasma levels, indicating potential tumor concentration of gefitinib. Tyrosine phosphorylated STAT3 was abundant in the surgically resected tumor tissue, indicating potential role in primary resistance in vivo. CONCLUSIONS: This study confirms previous preclinical observations that tumor tissues concentrate gefitinib. Persistent STAT3 may be leading to primary resistance to EGFR inhibitors in vivo.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Signal Transduction/drug effects , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Aged , Antineoplastic Agents/pharmacokinetics , Blotting, Western , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gefitinib , Humans , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Neoplasm Staging , Phosphorylation/drug effects , Pilot Projects , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacokinetics , STAT3 Transcription Factor/metabolism , Survival Rate , Tissue Distribution , Tyrosine/metabolism
10.
Nat Chem Biol ; 6(4): 291-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20190765

ABSTRACT

We describe a strategy for comprehending signaling pathways that are active in lung cancer cells and that are targeted by dasatinib using chemical proteomics to identify direct interacting proteins combined with immunoaffinity purification of tyrosine-phosphorylated peptides corresponding to activated tyrosine kinases. We identified nearly 40 different kinase targets of dasatinib. These include SRC-family kinase (SFK) members (LYN, SRC, FYN, LCK and YES), nonreceptor tyrosine kinases (FRK, BRK and ACK) and receptor tyrosine kinases (Ephrin receptors, DDR1 and EGFR). Using quantitative phosphoproteomics, we identified peptides corresponding to autophosphorylation sites of these tyrosine kinases that are inhibited in a concentration-dependent manner by dasatinib. Using drug-resistant gatekeeper mutants, we show that SFKs (particularly SRC and FYN), as well as EGFR, are relevant targets for dasatinib action. The combined mass spectrometry-based approach described here provides a system-level view of dasatinib action in cancer cells and suggests both functional targets and a rationale for combinatorial therapeutic strategies.


Subject(s)
Lung Neoplasms/drug therapy , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Proteomics/methods , Pyrimidines/pharmacology , Thiazoles/pharmacology , Apoptosis , Cell Line, Tumor , Dasatinib , Gene Knockdown Techniques , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mass Spectrometry , Peptides/chemistry , Peptides/metabolism , Phenotype , Phosphorylation/drug effects , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Thiazoles/metabolism , Thiazoles/therapeutic use
11.
J Clin Oncol ; 28(8): 1387-94, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20142592

ABSTRACT

PURPOSE: Src family kinase (SFK) proteins are frequently activated in cancer and can coordinate tumor cell growth, survival, invasion, and angiogenesis. Given the importance of SFK signaling in cancer, known cooperation between SFK and epidermal growth factor receptor (EGFR) signaling, and efficacy of EGFR inhibitors, we performed a phase I trial combining dasatinib, an SFK and multikinase inhibitor, with erlotinib, an EGFR inhibitor, in patients with advanced non-small-cell lung cancer. PATIENTS AND METHODS: Patients received erlotinib for 1 week before addition of dasatinib; pharmacokinetics were performed after weeks 1 and 2. Four cohorts were examined, including twice-daily and daily dasatinib dosing. Responses were assessed after 8 weeks. Plasma levels of angiogenic markers (vascular endothelial growth factor [VEGF], interleukin-8, and basic fibroblast growth factor [bFGF]) were determined before and during treatment. RESULTS: Thirty-four patients were enrolled. The average duration of treatment was 73 days. The main adverse events include GI (diarrhea, anorexia, and nausea), skin rash, cytopenias, pleural effusions, and fatigue. No effect of escalating doses of dasatinib was observed on erlotinib pharmacokinetics. Two partial responses and one bone response were observed, and the disease control rate was 63%. Reductions in plasma VEGF and bFGF were observed, and reductions in VEGF correlated with disease control. CONCLUSION: The combination of erlotinib and dasatinib is tolerable, with adverse effects consistent with the two agents. Disease control and inhibition of plasma angiogenesis markers were observed. Personalized strategies for deployment of SFK should receive further attention.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Pharmacological/blood , Dasatinib , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Female , Fibroblast Growth Factor 2/blood , Humans , Male , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Quinazolines/administration & dosage , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Thiazoles/administration & dosage , Thiazoles/adverse effects , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Vascular Endothelial Growth Factor A/blood
12.
Nat Rev Clin Oncol ; 6(10): 587-95, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19787002

ABSTRACT

Src family kinases (SFKs) have a critical role in cell adhesion, invasion, proliferation, survival, and angiogenesis during tumor development. SFKs comprise nine family members that share similar structure and function. Overexpression or high activation of SFKs occurs frequently in tumor tissues and they are central mediators in multiple signaling pathways that are important in oncogenesis. SFKs can interact with tyrosine kinase receptors, such as EGFR and the VEGF receptor. SFKs can affect cell proliferation via the Ras/ERK/MAPK pathway and can regulate gene expression via transcription factors such as STAT molecules. SFKs can also affect cell adhesion and migration via interaction with integrins, actins, GTPase-activating proteins, scaffold proteins, such as p130(CAS) and paxillin, and kinases such as focal adhesion kinases. Furthermore, SFKs can regulate angiogenesis via gene expression of angiogenic growth factors, such as fibroblast growth factor, VEGF, and interleukin 8. On the basis of these important findings, small-molecule SFK inhibitors have been developed and are undergoing early phase clinical testing. In preclinical studies these agents can suppress tumor growth and metastases. The agents seem to be safe in humans and could add to the therapeutic arsenal against subsets of cancers.


Subject(s)
Neoplasms/drug therapy , src-Family Kinases/genetics , src-Family Kinases/metabolism , Actins/metabolism , Animals , Cell Adhesion , Cell Movement , Cell Proliferation , Cell Survival , Clinical Trials as Topic , Crk-Associated Substrate Protein/metabolism , Drug Evaluation, Preclinical , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Integrins/metabolism , MAP Kinase Signaling System , Neoplasm Metastasis/drug therapy , Neoplasms/metabolism , Paxillin/metabolism , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Time Factors , Transcription Factors/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry
13.
J Interferon Cytokine Res ; 27(3): 209-20, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17348819

ABSTRACT

Stat transcription factors are activated by cytokines and can activate pathways important in oncogenesis. Although previous studies have identified an oncogenic role of Stat3 in lung cancer cells, the role of Stat1 is unclear. Using a mutant of Stat1 with constitutive activity (Stat1C), we examined the effect of persistent Stat1 activity on lung cancer cell growth, survival and gene expression. We identified no significant effect of Stat1C alone or with interferon-gamma (IFN-gamma) on lung cancer cell growth or survival. Consistent with prior reports, Stat1C expression alone elicited minimal changes in gene expression and required costimulatory IFN-gamma for full activity. Using oligonucleotide gene arrays and quantitative real-time PCR, we identified numerous proinflammatory gene products and chemokines regulated by IFN-gamma/Stat1C signaling. These results suggest the major role of IFN-gamma and Stat1 in lung cells is to direct a proinflammatory gene expression program rather than have major effects on cell growth or survival or both.


Subject(s)
Antiviral Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Interferon-gamma/pharmacology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Chemokines/biosynthesis , Chemokines/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics
14.
Cancer Res ; 66(11): 5542-8, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740687

ABSTRACT

Mutations of the epidermal growth factor receptor (EGFR) selectively activate Akt and signal transducer and activator of transcription (STAT) pathways that are important in lung cancer cell survival. Src family kinases can cooperate with receptor tyrosine kinases to signal through downstream molecules, such as phosphatidylinositol 3-kinase/PTEN/Akt and STATs. Based on the importance of EGFR signaling in lung cancer, the known cooperation between EGFR and Src proteins, and evidence of elevated Src activity in human lung cancers, we evaluated the effectiveness of a novel orally bioavailable Src inhibitor dasatinib (BMS-324825) in lung cancer cell lines with defined EGFR status. Here, we show that cell fate (death versus growth arrest) in lung cancer cells exposed to dasatinib is dependent on EGFR status. In cells with EGFR mutation that are dependent on EGFR for survival, dasatinib reduces cell viability through the induction of apoptosis while having minimal apoptotic effect on cell lines with wild-type (WT) EGFR. The induction of apoptosis in these EGFR-mutant cell lines corresponds to down-regulation of activated Akt and STAT3 survival proteins. In cell lines with WT or resistant EGFR mutation that are not sensitive to EGFR inhibition, dasatinib induces a G(1) cell cycle arrest with associated changes in cyclin D and p27 proteins, inhibits activated FAK, and prevents tumor cell invasion. Our results show that dasatinib could be effective therapy for patients with lung cancers through disruption of cell growth, survival, and tumor invasion. Our results suggest EGFR status is important in deciding cell fate in response to dasatinib.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Dasatinib , ErbB Receptors/genetics , Gene Deletion , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Signal Transduction , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
15.
Clin Cancer Res ; 11(23): 8288-94, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322287

ABSTRACT

PURPOSE: Signal transducers and activators of transcription 3 (Stat3), a member of the STAT family of transcription factors, regulates multiple oncogenic pathways, including pathways regulating tumor cell survival. We evaluated Stat3 activation in early stage non-small cell lung cancers (NSCLC) and how this relates to upstream epidermal growth factor receptor (EGFR) activation, tumor apoptosis, and prognosis. EXPERIMENTAL DESIGN: High-density tissue microarrays using tissues from 176 surgically resected NSCLC were evaluated for expression of phosphorylated Stat3 (pStat3) and epidermal growth factor receptor (pEGFR) along with tumor apoptosis. Using NSCLC cell lines, we evaluated how pStat3 expression relates to EGFR mutations and sensitivity of cells to gefitinib. RESULTS: We identified nuclear pStat3 expression in 54% of tumors. pStat3 expression was correlated with smaller tumors (P < 0.0001) and with limited smoking history (P = 0.02). We identified a trend toward higher pStat3 expression in adenocarcinomas compared with other tumor histology (P = 0.09). No relationship was found between pStat3 and prognosis following surgical resection. Importantly, we found a strong positive correlation between pEGFR expression and pStat3 expression (P <0.0001) and an inverse correlation between pStat3 and apoptosis (P = 0.01) consistent with less apoptosis in tumors expressing high amounts of pStat3. Cell lines with mutant EGFR have increased levels of pStat3 compared with cell lines without mutant EGFR and this correlates with their sensitivity to gefitinib. Finally, antisense-mediated knockdown of Stat3 induces apoptosis in EGFR mutant lung cancer cells. CONCLUSIONS: Early-stage NSCLC tumors have activated EGFR-Stat3 signaling with low apoptosis. Our findings suggest that pStat3 expression may be helpful in identifying patients appropriate for treatment with EGFR tyrosine kinase inhibitors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Female , Gefitinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Neoplasm Staging , Phosphorylation , Prognosis , Quinazolines/therapeutic use , Survival Rate , Tissue Array Analysis
16.
Cancer Biol Ther ; 4(3): 267-76, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15753661

ABSTRACT

Overexpression of anti-apoptotic Bcl-2 family members and deregulation of the pathways that regulate pro-apoptotic family members have been observed in non-small cell lung cancers (NSCLC). Previous reports have identified both Bcl-2 and Bcl-x(L) proteins as survival factors in lung cancer cells since reductions in these proteins can induce apoptosis and sensitize lung cancer cells to apoptosis induced by chemotherapy agents. Myeloid cell leukemia-1 (Mcl-1), another member of the Bcl-2 family, has been found to be a critical survival factor in hematopoietic cells, yet little data exists for a role of Mcl-1 in human lung cancers. We used NSCLC cell lines to explore how Mcl-1 levels affect lung cancer cell survival and studied tumors from patients to determine expression patterns of Mcl-1. NSCLC cells express abundant Mcl-1 protein and depletion of Mcl-1 levels by antisense Mcl-1 oligonucleotides induces apoptosis in A549 and H1299 lung cancer cells. Reduction in Mcl-1 levels can sensitize lung cancer cells to apoptosis induced by cytotoxic agents as well as by ionizing radiation. Lung cancer cells overexpressing Mcl-1 are less sensitive to apoptosis induced by chemotherapeutic agents, ZD1839 (an inhibitor of EGFR tyrosine kinase) and Bcl-2 or Bcl-x(L) antisense oligonucleotides. We find that epidermal growth factor (EGF) can enhance Mcl-1 protein levels in an ERK-dependent manner. Signal transduction agents that reduce Mcl-1 levels correlated with their individual ability to induce apoptosis in lung cancer cells. Finally, NSCLC tumors taken directly from patients have elevated levels of Mcl-1 protein compared with normal adjacent lung tissue. Therefore, agents that target Mcl-1 can induce apoptosis and sensitize cells to apoptosis induced by cytotoxic agents. Mcl-1 protein is overexpressed in a subset of human NSCLC and enhanced levels of Mcl-1 may protect lung cancer cells from death induced by a variety of pro-apoptotic stimuli.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Lung Neoplasms/genetics , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinazolines/pharmacology , Radiation Tolerance , Transcriptional Activation , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics
17.
Oncogene ; 24(21): 3397-408, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15735721

ABSTRACT

Wound healing and cancer are both characterized by cell proliferation, remodeling of extracellular matrix, cell invasion and migration, new blood vessel formation, and modulation of blood coagulation. The mechanisms that link wound healing and cancer are poorly understood. We report here that Stat3, a common signaling mechanism involved in oncogenesis and tissue injury, regulates a common set of genes involved in wound healing and cancer. Using oligonucleotide gene arrays and quantitative real-time PCR, we evaluated changes in global gene expression resulting from expression of Stat3 in lung epithelial cells. We report here previously uncharacterized genes induced by Stat3 implicated in signaling pathways common to both wound healing and cancer including cell invasion and migration, angiogenesis, modulation of coagulation, and repression of interferon-inducible genes. Consistent with these results, we found increased Stat3 activity associated with wound healing in chronically inflamed mouse lungs and increased Stat3 activity was identified at the leading edge of lung tumors invading adjacent nontumor stroma. These findings provide a molecular basis for understanding cancer as a deregulation of normal wound healing processes.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacology , Gene Expression Profiling , Neoplasms/genetics , Neoplasms/physiopathology , Trans-Activators/genetics , Trans-Activators/pharmacology , Wound Healing/genetics , Acute-Phase Proteins , Animals , Cell Movement , Cell Transformation, Neoplastic , Disease Models, Animal , Epithelial Cells , Inflammation , Lung/cytology , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , STAT3 Transcription Factor , Signal Transduction
18.
Immunology ; 109(4): 572-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12871225

ABSTRACT

We reported previously that low-density lipoprotein (LDL)-containing immune complexes (LDL-IC) stimulated matrix metalloproteinase-1 (MMP-1) expression in U937 histiocytes through Fc gamma receptor (FcgammaR)-mediated extracellular signal-regulated kinase pathway. The present study has explored the transcriptional mechanisms involved in the stimulation. Deletion analysis showed that LDL-IC stimulated MMP-1 promoter activity in cells transfected with the Construct 1 that contained a 4,334-bp MMP-1 promoter fragment, but had no effect in cells transfected with other constructs that had shorter MMP-1 promoter (2685-bp or less), suggesting that cis-acting elements located between -4334 and -2685 are required for the promoter stimulation. The mutation study further indicated that the activator protein-1 (AP-1) (-3471) or Ets (-3836) motifs in this distal region were essential for the LDL-IC-stimulated MMP-1 expression. Moreover, although above deletion analysis showed that LDL-IC did not stimulate MMP-1 promoter activity in cells transfected with constructs that contained the proximal AP-1 (-72) and Ets (-88) in the promoter fragments that are 2685-bp or less, the mutations of the -72 AP-1 or the -88 Ets motif in the construct 1 abolished the stimulation of MMP-1 expression by LDL-IC, suggesting that a long promoter sequence is required for the -72 AP-1 and -88 Ets motifs to be involved in the stimulation. Finally, electrophoretic mobility shift assay showed that LDL-IC stimulated the activities of transcription factors AP-1 and Ets. In conclusion, the present study shows that both the distal and proximal AP-1 and Ets motifs are required for LDL-IC-stimulated MMP-1 expression in U937 histiocytes.


Subject(s)
Antigen-Antibody Complex/immunology , Lipoproteins, LDL/immunology , Matrix Metalloproteinase 1/genetics , Promoter Regions, Genetic/immunology , Transcription Factor AP-1/immunology , Up-Regulation/immunology , Base Sequence , Blotting, Northern/methods , DNA/analysis , Electrophoretic Mobility Shift Assay/methods , Enzyme-Linked Immunosorbent Assay/methods , Gene Deletion , Humans , Matrix Metalloproteinase 1/immunology , Mutation/genetics , Mutation/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-ets , Transcription Factors/genetics , Transcription Factors/immunology , Transcription, Genetic , Transfection , U937 Cells
19.
Oncogene ; 22(27): 4150-65, 2003 Jul 03.
Article in English | MEDLINE | ID: mdl-12833138

ABSTRACT

Overexpression of receptor tyrosine kinases including the epidermal growth factor receptor (EGF-R) as well as nonreceptor tyrosine kinases, such as Src, have been implicated in the formation of human lung cancers. In addition, cytokines like interleukin-6 (IL-6) have been demonstrated to modulate lung cancer cell growth and elevated levels of IL-6 have been shown to be an adverse prognostic factor for patients with lung cancer. Despite a large body of evidence pointing to their potential importance, few direct studies into the role of signal transducers and activators of transcription (STAT) pathways in human lung cancer have been undertaken. Here we demonstrate that multiple nonsmall cell lung cancer cell lines demonstrate constitutive Stat3 DNA-binding activity. Stat3 DNA-binding activity is specifically upregulated by the addition of epidermal growth factor (EGF), IL-6, and hepatocyte-derived growth factor (HGF). Furthermore, the stimulation of Stat3 DNA-binding activity by EGF requires the activity of EGF-R tyrosine kinase as well as Src-kinase, while the upregulation of Stat3 activity by IL-6 or HGF requires only Src-kinase activity. Treatment of A549 lung cancer cells with PD180970 or SU6656, both pharmacological inhibitors of Src-kinase, resulted in reduced Src and Stat3 activity, cell cycle arrest in G2, and reduced viability of cells accompanied by induction of apoptosis. Treatment of Stat3-positive A549 and H358 cells with antisense Stat3 oligonucleotides results in complete loss of Stat3 DNA-binding activity and apoptosis, while Stat3-positive H1299 cells remained healthy. Finally, an adenoviral vector expressing a dominant-negative Stat3 isoform results in loss of Stat3 DNA-binding activity, apoptosis, and reduced cellular viability. These results demonstrate a role of Stat3 in transducing survival signals downstream of tyrosine kinases such as Src, EGF-R, and c-Met, as well as cytokines such as IL-6, in human nonsmall cell lung cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Lung Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , Adenoviridae/genetics , Apoptosis , Blotting, Western , Cell Cycle , Cell Nucleus/metabolism , Cell Survival , DNA/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , G2 Phase , Genes, Dominant , Hepatocyte Growth Factor/metabolism , Humans , Interleukin-6/metabolism , Models, Biological , Oligonucleotides, Antisense/pharmacology , Protein Binding , Protein-Tyrosine Kinases/antagonists & inhibitors , STAT3 Transcription Factor , Time Factors , Transfection , Tumor Cells, Cultured
20.
Oncogene ; 21(13): 2000-8, 2002 Mar 27.
Article in English | MEDLINE | ID: mdl-11960372

ABSTRACT

Non-receptor and receptor tyrosine kinases, such as Src and EGF receptor (EGFR), are major inducers of vascular endothelial growth factor (VEGF), one of the most potent mediators of angiogenesis. While tyrosine kinases signal through multiple pathways, signal transducer and activation of transcription 3 (Stat3) is a point of convergence for many of these and is constitutively activated with high frequency in a wide range of cancer cells. Here, we show that VEGF expression correlates with Stat3 activity in diverse human cancer cell lines. An activated Stat3 mutant (Stat3C) up-regulates VEGF expression and stimulates tumor angiogenesis. Stat3C-induced VEGF up-regulation is abrogated when a Stat3-binding site in the VEGF promoter is mutated. Furthermore, interrupting Stat3 signaling with dominant-negative Stat3 protein or Stat3 antisense oligonucleotide in tumor cells down-regulates VEGF expression. Consistent with an important role of Stat3 in VEGF up-regulation induced by various oncogenic tyrosine kinases, v-Src-mediated VEGF expression is inhibited when Stat3 signaling is blocked. Moreover, chromatin immunoprecipitation assays indicate that Stat3 protein binds to the VEGF promoter in vivo and mutation of a Stat3-binding site in the VEGF promoter abrogates v-Src-induced VEGF promoter activity. These studies provide evidence that the VEGF gene is regulated directly by Stat3 protein, and indicate that Stat3 represents a common molecular target for blocking angiogenesis induced by multiple signaling pathways in human cancers.


Subject(s)
DNA-Binding Proteins/metabolism , Endothelial Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Lymphokines/metabolism , Neovascularization, Pathologic , Trans-Activators/metabolism , Up-Regulation , 3T3 Cells , Animals , Base Sequence , Cell Line, Transformed , Collagen , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Down-Regulation , Drug Combinations , Electrophoretic Mobility Shift Assay , Endothelial Growth Factors/genetics , Humans , Laminin , Lymphokines/genetics , Mice , Mutagenesis, Site-Directed , Neoplasm Transplantation , Oncogene Protein pp60(v-src)/metabolism , Promoter Regions, Genetic/genetics , Proteoglycans , STAT3 Transcription Factor , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...