Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Methods Mol Biol ; 467: 189-210, 2009.
Article in English | MEDLINE | ID: mdl-19301672

ABSTRACT

Angiogenesis is a complex sequential process involving endothelial activation, basement membrane degradation, endothelial sprouting from the parent vessel, invasion of the extracellular matrix, endothelial proliferation, vessel elongation, branching, anastomosis, increases in vessel diameter, basement membrane formation, pericyte acquisition, and remodelling. Most in vitro angiogenesis assays are two-dimensional and measure only one facet of this process, generally endothelial proliferation, migration, or tube formation. The two-dimensional nature of the assays also ignores the differences in endothelial phenotype seen in three-dimensional models and in vivo. The in vitro serum-free three-dimensional rat aortic model closely approximates the complexities of angiogenesis in vivo, from endothelial activation to pericyte acquisition and remodelling, and most of these can be quantified by image analysis, immunohistochemistry, and biochemical analysis. It is easily manipulated using molecular biological intervention or exogenous inhibitors and activators in a relatively controlled system.


Subject(s)
Aorta , Cell Culture Techniques/methods , Neovascularization, Physiologic , Tissue Culture Techniques/methods , Animals , Aorta/cytology , Cell Proliferation , Male , Models, Animal , Rats , Rats, Inbred F344
2.
Mol Cancer Ther ; 16(10): 2107-2119, 2017 10.
Article in English | MEDLINE | ID: mdl-28619752

ABSTRACT

Several therapeutic agents targeting HGF/MET signaling are under clinical development as single agents or in combination, notably with anti-EGFR therapies in non-small cell lung cancer (NSCLC). However, despite increasing data supporting a link between MET, irradiation, and cancer progression, no data regarding the combination of MET-targeting agents and radiotherapy are available from the clinic. S49076 is an oral ATP-competitive inhibitor of MET, AXL, and FGFR1-3 receptors that is currently in phase I/II clinical trials in combination with gefitinib in NSCLC patients whose tumors show resistance to EGFR inhibitors. Here, we studied the impact of S49076 on MET signaling, cell proliferation, and clonogenic survival in MET-dependent (GTL16 and U87-MG) and MET-independent (H441, H460, and A549) cells. Our data show that S49076 exerts its cytotoxic activity at low doses on MET-dependent cells through MET inhibition, whereas it inhibits growth of MET-independent cells at higher but clinically relevant doses by targeting Aurora B. Furthermore, we found that S49076 improves the antitumor efficacy of radiotherapy in both MET-dependent and MET-independent cell lines in vitro and in subcutaneous and orthotopic tumor models in vivo In conclusion, our study demonstrates that S49076 has dual antitumor activity and can be used in combination with radiotherapy for the treatment of both MET-dependent and MET-independent tumors. These results support the evaluation of combined treatment of S49076 with radiation in clinical trials without patient selection based on the tumor MET dependency status. Mol Cancer Ther; 16(10); 2107-19. ©2017 AACR.


Subject(s)
Aurora Kinase B/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Proto-Oncogene Proteins c-met/genetics , Aurora Kinase B/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Gefitinib , Gene Expression Regulation, Neoplastic , Humans , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinazolines/administration & dosage , Radiation Tolerance , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
3.
Neoplasia ; 19(1): 35-42, 2017 01.
Article in English | MEDLINE | ID: mdl-27988457

ABSTRACT

The fibroblast growth factor receptor (FGFR) pathway has been implicated both as an escape mechanism from anti-angiogenic therapy and as a driver oncogene in different tumor types. Lucitanib is a small molecule inhibitor of vascular endothelial growth factor (VEGF) receptors 1 to 3 (VEGFR1 to 3), platelet derived growth factor α/ß (PDGFRα/ß) and FGFR1-3 tyrosine kinases and has demonstrated activity in a phase I/II clinical study, with objective RECIST responses in breast cancer patients with FGFR1 or FGF3/4/19 gene amplification, as well as in patients anticipated to benefit from anti-angiogenic agents. We report here the in vitro and in vivo antitumor activity of lucitanib in experimental models with or without FGFR1/2 amplification or mutations. In cell assays, lucitanib potently inhibited the growth of tumor cell lines with amplified FGFR1 or mutated/amplified FGFR2. In all xenograft models studied, lucitanib demonstrated marked tumor growth inhibition due to potent inhibition of angiogenesis. Notably, in two lung cancer models with FGFR1 amplification, the antitumor efficacy was higher, suggesting that the simultaneous inhibition of VEGF and FGF receptors in FGFR1 dependent tumors can be therapeutically advantageous. Similar antitumor activity was observed in FGFR2 wild-type and amplified or mutated xenograft models. Pharmacokinetic studies showed lucitanib plasma concentrations in the micro/sub-micromolar range demonstrated drug accumulation following repeated lucitanib administration.


Subject(s)
Gene Amplification , Mutation , Naphthalenes/pharmacology , Quinolines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Dosage , Humans , Inhibitory Concentration 50 , Mice , Protein Kinase Inhibitors/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 12(9): 1749-62, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804704

ABSTRACT

Aberrant activity of the receptor tyrosine kinases MET, AXL, and FGFR1/2/3 has been associated with tumor progression in a wide variety of human malignancies, notably in instances of primary or acquired resistance to existing or emerging anticancer therapies. This study describes the preclinical characterization of S49076, a novel, potent inhibitor of MET, AXL/MER, and FGFR1/2/3. S49076 potently blocked cellular phosphorylation of MET, AXL, and FGFRs and inhibited downstream signaling in vitro and in vivo. In cell models, S49076 inhibited the proliferation of MET- and FGFR2-dependent gastric cancer cells, blocked MET-driven migration of lung carcinoma cells, and inhibited colony formation of hepatocarcinoma cells expressing FGFR1/2 and AXL. In tumor xenograft models, a good pharmacokinetic/pharmacodynamic relationship for MET and FGFR2 inhibition following oral administration of S49076 was established and correlated well with impact on tumor growth. MET, AXL, and the FGFRs have all been implicated in resistance to VEGF/VEGFR inhibitors such as bevacizumab. Accordingly, combination of S49076 with bevacizumab in colon carcinoma xenograft models led to near total inhibition of tumor growth. Moreover, S49076 alone caused tumor growth arrest in bevacizumab-resistant tumors. On the basis of these preclinical studies showing a favorable and novel pharmacologic profile of S49076, a phase I study is currently underway in patients with advanced solid tumors. Mol Cancer Ther; 12(9); 1749-62. ©2013 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Indoles/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thiazolidinediones/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Bevacizumab , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Indoles/chemistry , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Thiazolidinediones/chemistry , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
5.
Sci Signal ; 2(100): ra80, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19996456

ABSTRACT

Tumor onset and progression require the accumulation of many genetic and epigenetic lesions. In some cases, however, cancer cells rely on only one of these lesions to maintain their malignant properties, and this dependence results in tumor regression upon oncogene inactivation ("oncogene addiction"). Determining which nodes of the many networks operative in the transformed phenotype specifically mediate this response to oncogene neutralization is crucial to identifying the vulnerabilities of cancer. Using the Met receptor as the major model system, we combined multiplex phosphoproteomics, genome-wide expression profiling, and functional assays in various cancer cells addicted to oncogenic receptor tyrosine kinases. We found that Met blockade affected a limited subset of Met downstream signals: Little or no effect was observed for several pathways downstream of Met; instead, only a restricted and pathway-specific signature of transducers and transcriptional effectors downstream of Ras or phosphoinositide 3-kinase (PI3K) was inactivated. An analogous signature was also generated by inhibition of epidermal growth factor receptor in a different cellular context, suggesting a stereotyped response that likely is independent of receptor type or tissue origin. Biologically, Met inhibition led to cell-cycle arrest. Inhibition of Ras-dependent signals and PI3K-dependent signals also resulted in cell-cycle arrest, whereas cells in which Met was inhibited proliferated when Ras or PI3K signaling was active. These findings uncover "dominant" and "recessive" nodes among the numerous oncogenic networks regulated by receptor tyrosine kinases and active in cancer, with the Ras and PI3K pathways as determinants of therapeutic response.


Subject(s)
Oncogenes , Proto-Oncogene Proteins c-met/metabolism , Blotting, Western , Cell Line , ErbB Receptors/metabolism , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Oncogene Protein p21(ras)/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Signal Transduction
6.
Sci Signal ; 2(102): er11, 2009.
Article in English | MEDLINE | ID: mdl-20039471

ABSTRACT

Tumor onset and progression require the accumulation of many genetic and epigenetic lesions. In some cases, however, cancer cells rely on only one of these lesions to maintain their malignant properties, and this dependence results in tumor regression upon oncogene inactivation ("oncogene addiction"). Determining which nodes of the many networks operative in the transformed phenotype specifically mediate this response to oncogene neutralization is crucial to identifying the vulnerabilities of cancer. Using the Met receptor as the major model system, we combined multiplex phosphoproteomics, genome-wide expression profiling, and functional assays in various cancer cells addicted to oncogenic receptor tyrosine kinases. We found that Met blockade affected a limited subset of Met downstream signals: Little or no effect was observed for several pathways downstream of Met; instead, only a restricted and pathway-specific signature of transducers and transcriptional effectors downstream of Ras or phosphoinositide 3-kinase (PI3K) was inactivated. An analogous signature was also generated by inhibition of epidermal growth factor receptor in a different cellular context, suggesting a stereotyped response that likely is independent of receptor type or tissue origin. Biologically, Met inhibition led to cell-cycle arrest. Inhibition of Ras-dependent signals and PI3K-dependent signals also resulted in cell-cycle arrest, whereas cells in which Met was inhibited proliferated when Ras or PI3K signaling was active. These findings uncover "dominant" and "recessive" nodes among the numerous oncogenic networks regulated by receptor tyrosine kinases and active in cancer, with the Ras and PI3K pathways as determinants of therapeutic response.


Subject(s)
Cell Cycle/physiology , Gene Silencing/physiology , Neoplasms/metabolism , Oncogenes/physiology , Proto-Oncogene Proteins c-met/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction/physiology , Blotting, Western , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Oncogene Protein p21(ras)/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Polymerase Chain Reaction , Proteomics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/genetics , Sequence Analysis, DNA
7.
Mol Pharmacol ; 63(6): 1281-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12761337

ABSTRACT

The role of adhesion molecules, such as alphav integrins, in the control of the survival of quiescent tumor cells is unclear. We used S 34961, a novel small molecule alphav integrin antagonist, to investigate the role of integrin-signaling in the survival of populations of quiescent human HT-29 and HCT 116 colon carcinoma cells. S 34961 at 1 microM induced detachment, but cells retained viability, existing as clusters. Nonligated beta-integrins may recruit and activate caspase-8 [J Cell Biol 155:459-470, 2001]. However, congruent with the absence of apoptosis, no activation of caspase-8 in these cells was detected after incubation with S 34961. A rapid (2 h) change in conformation of the N terminus of proapoptotic Bak was observed before detachment, together with a decrease in phosphorylation of focal adhesion kinase (2 h) and subsequent (8 h) decreases in phosphorylation of extracellular signal-regulated kinase-1/2 and Akt. Together, these results suggested that although treatment with S 34961 has no effect on survival per se, it may reduce the survival threshold of the tumor cells, with Bak in an activated state. Indeed, concomitant incubation of S 34961 with 10 microM U-0126 (a mitogen-activated protein kinase kinase inhibitor) was found to lead to apoptosis (at 24 h), whereas U-0126 alone had no effect. Together, these observations could guide the use of combination therapy with integrin antagonists in the clinic.


Subject(s)
Caspases/metabolism , Colonic Neoplasms/pathology , Cycloheptanes/pharmacology , Integrin alpha5/metabolism , Integrins/antagonists & inhibitors , Pyridines/pharmacology , Apoptosis , Caspase 8 , Caspase 9 , Cell Adhesion , Cell Survival/drug effects , Colonic Neoplasms/enzymology , Enzyme Activation , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , HT29 Cells , Humans , Integrin alpha5/drug effects , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , bcl-2 Homologous Antagonist-Killer Protein
SELECTION OF CITATIONS
SEARCH DETAIL