Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 74(22): 6598-609, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25217525

RESUMEN

Cell-based drug screenings indicate that tumors displaying c-MET gene amplification are "addicted" to MET signaling and therefore are very sensitive to MET-targeted agents. However, these screenings were conducted in the absence of the MET ligand, hepatocyte growth factor (HGF), which is abundant in the tumor microenvironment. Sensitivity of six MET-addicted human tumor cells to three MET kinase inhibitors (JNJ-38877605, PHA-665752, crizotinib) and one antagonistic anti-MET antibody (DN30 Fab) was analyzed in the absence or presence of HGF, in a stroma-tumor coculture system, and by combining anti-MET drugs with an HGF neutralizing antibody (ficlatuzumab) in human HGF knock-in mice bearing c-MET-amplified tumors. In all models examined, HGF promoted resistance to MET-targeted agents, affecting both their potency and efficacy. HGF-induced resistance was due to restoration of physiologic GAB1-mediated PI3K activation that compensated for loss of aberrant HER3-dependent PI3K signaling. Ficlatuzumab restored sensitivity to MET-targeted agents in coculture systems and overcame resistance to JNJ-38877605, crizotinib, and DN30 Fab in human HGF knock-in mice. These data suggest that c-MET-amplified tumor cells-which normally exhibit ligand-independent, constitutive MET activation-become dependent on HGF for survival upon pharmacologic MET inhibition. Because HGF is frequently overexpressed in human cancer, this mechanism may represent a major cause of resistance to anti-MET therapies. The ability of ficlatuzumab to overcome HGF-mediated resistance generates proof of principle that vertical inhibition of both a tyrosine kinase receptor and its ligand can be therapeutically beneficial and opens new perspectives for the treatment of MET-dependent tumors.


Asunto(s)
Factor de Crecimiento de Hepatocito/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Microambiente Tumoral , Animales , Anticuerpos Monoclonales/farmacología , Ratones , Ratones SCID , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-met/fisiología , Receptor ErbB-3/fisiología , Transducción de Señal
3.
Clin Cancer Res ; 19(9): 2381-92, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23532890

RESUMEN

PURPOSE: MET, the high-affinity receptor for hepatocyte growth factor, is frequently deregulated in human cancer. Tivantinib (ARQ197; Arqule), a staurosporine derivative that binds to the dephosphorylated MET kinase in vitro, is being tested clinically as a highly selective MET inhibitor. However, the mechanism of action of tivantinib is still unclear. EXPERIMENTAL DESIGN: The activity of tivantinib was analyzed in multiple cellular models, including: cells displaying c-MET gene amplification, strictly 'addicted' to MET signaling; cells with normal c-MET gene copy number, not dependent on MET for growth; cells not expressing MET; somatic knockout cells in which the ATP-binding cleft of MET, where tivantinib binds, was deleted by homologous recombination; and a cell system 'poisoned' by MET kinase hyperactivation, where cells die unless cultured in the presence of a specific MET inhibitor. RESULTS: Tivantinib displayed cytotoxic activity independently of c-MET gene copy number and regardless of the presence or absence of MET. In both wild-type and isogenic knockout cells, tivantinib perturbed microtubule dynamics, induced G2/M arrest, and promoted apoptosis. Tivantinib did not rescue survival of cells 'poisoned' by MET kinase hyperactivation, but further incremented cell death. In all cell models analyzed, tivantinib did not inhibit HGF-dependent or -independent MET tyrosine autophosphorylation. CONCLUSIONS: We conclude that tivantinib displays cytotoxic activity via molecular mechanisms that are independent from its ability to bind MET. This notion has a relevant impact on the interpretation of clinical results, on the design of future clinical trials, and on the selection of patients receiving tivantinib treatment.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Pirrolidinonas/farmacología , Quinolinas/farmacología , Moduladores de Tubulina/farmacología , Apoptosis , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Dosificación de Gen , Células Hep G2 , Humanos , Microtúbulos/efectos de los fármacos , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética
4.
J Clin Invest ; 119(4): 865-75, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19307731

RESUMEN

As a tumor grows, it requires increased amounts of oxygen. However, the tumor blood vessels that form to meet this demand are functionally impaired, leading to regions of hypoxia within the tumor. Such hypoxia is one of the hallmarks of malignancy and is thought to promote a number of tumorigenic properties. Here, we sought to determine how tumors without hypoxia would progress by engineering A549 human lung carcinoma cells to ectopically express myoglobin (Mb), a multifunctional heme protein that specializes in oxygen transport, storage, and buffering. Mb expression prevented the hypoxic response in vitro and delayed tumor engraftment and reduced tumor growth following xenotransplantation into mice. Experimental tumors expressing Mb displayed reduced or no hypoxia, minimal HIF-1alpha levels, and a homogeneously low vessel density. Mb-mediated tumor oxygenation promoted differentiation of cancer cells and suppressed both local and distal metastatic spreading. These effects were primarily due to reduced tumor hypoxia, because they were not observed using point-mutated forms of myoglobin unable to bind oxygen and they were abrogated by expression of a constitutively active form of HIF-1alpha. Although limited to xenograft models, these data provide experimental proof of the concept that hypoxia is not just a side effect of deregulated growth but a key factor on which the tumor relies in order to promote its own expansion.


Asunto(s)
Mioglobina/genética , Mioglobina/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Muerte Celular , Diferenciación Celular , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Cartilla de ADN/genética , Femenino , Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Mutación , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Oxígeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Transducción Genética , Trasplante Heterólogo
5.
Cancer Res ; 68(22): 9176-83, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19010889

RESUMEN

Gene therapy provides a still poorly explored opportunity to treat cancer by "active" immunotherapy as it enables the transfer of genes encoding antibodies directed against specific oncogenic proteins. By a bidirectional lentiviral vector, we transferred the cDNA encoding the heavy and light chains of a monoclonal anti-Met antibody (DN-30) to epithelial cancer cells. In vitro, the transduced cells synthesized and secreted correctly assembled antibodies with the expected high affinity, inducing down-regulation of the Met receptor and strong inhibition of the invasive growth response. The inhibitory activity resulted (a) from the interference of the antibody with the Met receptor intracellular processing ("cell autonomous activity," in cis) and (b) from the antibody-induced cleavage of Met expressed at the cell surface ("bystander effect," in trans). The monoclonal antibody gene transferred into live animals by systemic administration or by local intratumor delivery resulted in substantial inhibition of tumor growth. These data provide proof of concept both for targeting the Met receptor and for a gene transfer-based immunotherapy strategy.


Asunto(s)
Anticuerpos Monoclonales/genética , Terapia Genética , Neoplasias/terapia , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/uso terapéutico , Línea Celular Tumoral , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Lentivirus/genética , Ratones , Neoplasias/patología , Fosforilación , Proteínas Proto-Oncogénicas c-met , Transducción de Señal
6.
Mol Cell Biol ; 25(10): 3982-96, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15870272

RESUMEN

The hepatocyte growth factor (HGF) receptor encoded by the Met oncogene controls a genetic program-known as "invasive growth"-responsible for several developmental processes and involved in cancer invasion and metastasis. This program functions through several regulatory gene products, as yet largely unknown, both upstream and downstream of Met. Here we show that activation of the Notch receptor results in transcriptional down-regulation of Met, suppression of HGF-dependent Ras signaling, and impairment of HGF-dependent cellular responses. In turn, Met activation leads to transcriptional induction of the Notch ligand Delta and the Notch effector HES-1, indicating that Met is able to self-tune its own protein levels and the ensuing biochemical and biological outputs through stimulation of the Notch pathway. By using branching morphogenesis of the tracheal system in Drosophila as a readout of invasive growth, we also show that exogenous expression of a constitutively active form of human Met induces enhanced sprouting of the tracheal tree, a phenotype that is further increased in embryos lacking Notch function. These results unravel an in-built mechanism of negative feedback regulation in which Met activation leads to transcriptional induction of Notch function, which in turn limits HGF activity through repression of the Met oncogene.


Asunto(s)
Retroalimentación Fisiológica , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Tráquea/citología , Tráquea/metabolismo , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , División Celular , Línea Celular , Línea Celular Tumoral , Perros , Regulación hacia Abajo/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Activación Enzimática , Retroalimentación Fisiológica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Fenotipo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Notch , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Tráquea/embriología , Factor de Transcripción HES-1 , Transcripción Genética/genética
7.
J Clin Invest ; 114(10): 1418-32, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15545993

RESUMEN

Scatter factor (SF), also known as hepatocyte growth factor, is ubiquitously present in the extracellular matrix of tissues in the form of an inactive precursor (pro-SF). In order to acquire biological activity, pro-SF must be cleaved by specific proteases present on the cell surface. The mature form of SF controls invasive cues in both physiological and pathological processes through activation of its receptor, the Met tyrosine kinase. By substituting a single amino acid in the proteolytic site, we engineered an unprocessable form of pro-SF (uncleavable SF). Using lentivirus vector technology, we achieved local or systemic delivery of uncleavable SF in mice. We provide evidence that (a) uncleavable SF inhibits both protease-mediated pro-SF conversion and active SF-induced Met activation; (b) local expression of uncleavable SF in tumors suppresses tumor growth, impairs tumor angiogenesis, and prevents metastatic dissemination; and (c) systemic expression of uncleavable SF dramatically inhibits the growth of transplanted tumors and abolishes the formation of spontaneous metastases without perturbing vital physiological functions. These data show that proteolytic activation of pro-SF is a limiting step in tumor progression, thus suggesting a new strategy for the treatment or prevention of the malignant conversion of neoplastic lesions.


Asunto(s)
Terapia Genética , Factor de Crecimiento de Hepatocito/fisiología , Metástasis de la Neoplasia/terapia , Neoplasias/terapia , Ingeniería de Proteínas , Sustitución de Aminoácidos , Animales , Neoplasias de la Mama/patología , Carcinoma/patología , Línea Celular Tumoral , Colágeno/metabolismo , Activación Enzimática , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Femenino , Vectores Genéticos , Glutamina/metabolismo , Humanos , Lentivirus/genética , Metionina/metabolismo , Ratones , Ratones Desnudos , Mitosis , Trasplante de Neoplasias , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Transducción Genética , Trasplante Heterólogo , Carga Tumoral
8.
Cancer Cell ; 3(4): 347-61, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12726861

RESUMEN

Hypoxia unleashes the invasive and metastatic potential of tumor cells by largely unknown mechanisms. The Met tyrosine kinase, a high affinity receptor for hepatocyte growth factor (HGF), plays a crucial role in controlling invasive growth and is often overexpressed in cancer. Here we show that: (1) hypoxia activates transcription of the met protooncogene, resulting in higher levels of Met; (2) hypoxic areas of tumors overexpress Met; (3) hypoxia amplifies HGF signaling; (4) hypoxia synergizes with HGF in inducing invasion; (5) the proinvasive effects of hypoxia are mimicked by Met overexpression; and (6) inhibition of Met expression prevents hypoxia-induced invasive growth. These data show that hypoxia promotes tumor invasion by sensitizing cells to HGF stimulation, providing a molecular basis to explain Met overexpression in cancer.


Asunto(s)
Hipoxia/fisiopatología , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Secuencia de Bases , Northern Blotting , Western Blotting , Movimiento Celular/fisiología , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Técnicas de Transferencia de Gen , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Transcripción Genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA