Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 14(3): e0212670, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30913212

RESUMEN

Immunotherapy has fundamentally changed the landscape of cancer treatment. Despite the encouraging results with the checkpoint modulators, response rates vary widely across tumor types, with a majority of patients exhibiting either primary resistance without a significant initial response to treatment or acquired resistance with subsequent disease progression. Hematopoietic progenitor kinase 1 (HPK1) is predominantly expressed in hematopoietic cell linages and serves as a negative regulator in T cells and dendritic cells (DC). While HPK1 gene knockout (KO) studies suggest its role in anti-tumor immune responses, the involvement of kinase activity and thereof its therapeutic potential remain unknown. To investigate the potential of pharmacological intervention using inhibitors of HPK1, we generated HPK1 kinase dead (KD) mice which carry a single loss-of-function point mutation in the kinase domain and interrogated the role of kinase activity in immune cells in the context of suppressive factors or the tumor microenvironment (TME). Our data provide novel findings that HKP1 kinase activity is critical in conferring suppressive functions of HPK1 in a wide range of immune cells including CD4+, CD8+, DC, NK to Tregs, and inactivation of kinase domain was sufficient to elicit robust anti-tumor immune responses. These data support the concept that an HPK1 small molecule kinase inhibitor could serve as a novel agent to provide additional benefit in combination with existing immunotherapies, particularly to overcome resistance to current treatment regimens.


Asunto(s)
Inmunidad Celular , Vigilancia Inmunológica , Linfocitos/inmunología , Neoplasias Experimentales/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Linfocitos/patología , Ratones , Ratones Mutantes , Neoplasias Experimentales/genética , Mutación Puntual , Proteínas Serina-Treonina Quinasas/genética , Microambiente Tumoral/genética
2.
Clin Cancer Res ; 17(12): 4031-41, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21531814

RESUMEN

PURPOSE: The extensive involvement of the HER kinases in epithelial cancer suggests that kinase inhibitors targeting this receptor family have the potential for broad spectrum antitumor activity. BMS-690514 potently inhibits all three HER kinases, and the VEGF receptor kinases. This report summarizes data from biochemical and cellular pharmacology studies, as well as antitumor activity of BMS-690514. EXPERIMENTAL DESIGN: The potency and selectivity of BMS-690514 was evaluated by using an extensive array of enzymatic and binding assays, as well as cellular assays that measure proliferation and receptor signaling. Antitumor activity was evaluated by using multiple xenograft models that depend on HER kinase signaling. The antiangiogenic properties of BMS-690514 were assessed in a matrigel plug assay, and effect on tumor blood flow was measured by dynamic contrast-enhanced MRI. RESULTS: BMS-690514 is a potent and selective inhibitor of epidermal growth factor receptor (EGFR), HER2, and HER4, as well as the VEGF receptor kinases. It inhibits proliferation of tumor cells with potency that correlates with inhibition of receptor signaling, and induces apoptosis in lung tumor cells that have an activating mutation in EGFR. Antitumor activity was observed with BMS-690514 at multiple doses that are well tolerated in mice. There was evidence of suppression of tumor angiogenesis and endothelial function by BMS-690514, which may contribute to its efficacy. CONCLUSIONS: By combining inhibition of two receptor kinase families, BMS-690524 is a novel targeted agent that disrupts signaling in the tumor and its vasculature.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Neoplasias/enzimología , Piperidinas/farmacología , Pirroles/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Triazinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/irrigación sanguínea , Neovascularización Patológica/enzimología , Receptor ErbB-2/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Bioorg Med Chem Lett ; 21(2): 781-5, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21177105
4.
J Med Chem ; 52(21): 6527-30, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19821562

RESUMEN

Structure-activity relationships in a series of 4-[1H-indazol-5-ylamino]pyrrolo[2,1-f][1,2,4]triazine-6-carbamates identified dual human epidermal growth factor receptor (HER)1/HER2 kinase inhibitors with excellent biochemical potency and kinase selectivity. On the basis of its favorable pharmacokinetic profile and robust in vivo activity in HER1 and HER2 driven tumor models, 13 (BMS-599626) was selected as a clinical candidate for treatment of solid tumors.


Asunto(s)
Antineoplásicos/síntesis química , Carbamatos/síntesis química , Receptores ErbB/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Triazinas/síntesis química , Administración Oral , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Disponibilidad Biológica , Carbamatos/farmacocinética , Carbamatos/farmacología , Línea Celular Tumoral , Perros , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Macaca fascicularis , Ratones , Trasplante de Neoplasias , Estereoisomerismo , Relación Estructura-Actividad , Trasplante Heterólogo , Triazinas/farmacocinética , Triazinas/farmacología
5.
J Med Chem ; 52(5): 1251-4, 2009 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-19260711

RESUMEN

Substituted N-(4-(2-aminopyridin-4-yloxy)-3-fluoro-phenyl)-1-(4-fluorophenyl)-2-oxo-1,2-dihydropyridine-3-carboxamides were identified as potent and selective Met kinase inhibitors. Substitution of the pyridine 3-position gave improved enzyme potency, while substitution of the pyridone 4-position led to improved aqueous solubility and kinase selectivity. Analogue 10 demonstrated complete tumor stasis in a Met-dependent GTL-16 human gastric carcinoma xenograft model following oral administration. Because of its excellent in vivo efficacy and favorable pharmacokinetic and preclinical safety profiles, 10 has been advanced into phase I clinical trials.


Asunto(s)
Aminopiridinas/síntesis química , Antineoplásicos/síntesis química , Dihidropiridinas/síntesis química , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Piridonas/síntesis química , Administración Oral , Aminopiridinas/farmacocinética , Aminopiridinas/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Dihidropiridinas/farmacocinética , Dihidropiridinas/farmacología , Perros , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Piridonas/farmacocinética , Piridonas/farmacología , Ratas , Solubilidad , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Bioorg Med Chem Lett ; 17(17): 4947-54, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17606372

RESUMEN

Pyrrolotriazine dual EGFR/HER2 kinase inhibitors with a 5-((4-aminopiperidin-1-yl)methyl) solubilizing group were found to be superior to analogs with previously reported C-5 solubilizing groups. New synthetic methodology was developed for the parallel synthesis of C-4 analogs with the new solubilizing group. Interesting new leads were evaluated in tumor xenograft models and the C-4 aminofluorobenzylindazole, 1c, was found to exhibit the best antitumor activity. It is hypothesized that this solubilizing group extends into the ribose-phosphate portion of the ATP binding pocket and enhances the binding affinity of the inhibitor.


Asunto(s)
Química Farmacéutica/métodos , Receptores ErbB/química , Neoplasias/tratamiento farmacológico , Piperidinas/síntesis química , Pirroles/síntesis química , Receptor ErbB-2/química , Triazinas/síntesis química , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Insectos , Modelos Químicos , Trasplante de Neoplasias , Piperidinas/química , Piperidinas/farmacología , Pirroles/química , Pirroles/farmacología , Triazinas/química , Triazinas/farmacología
7.
Bioorg Med Chem Lett ; 17(10): 2828-33, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17368025

RESUMEN

Novel C-5 aminomethyl pyrrolotriazines were prepared and optimized for dual EGFR and HER2 protein tyrosine kinase inhibition. The homopiperazine, 1p, emerged as a key lead and it showed promising oral efficacy in EGFR and dual EGFR/HER2 driven human tumor xenograft models. It is hypothesized that the C-5 homopiperazine side chain binds in the ribose-phosphate portion of the ATP binding pocket.


Asunto(s)
Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Metilaminas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirroles/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Triazinas/farmacología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Metilaminas/química , Ratones , Modelos Moleculares , Estructura Molecular , Trasplante de Neoplasias , Pirroles/química , Relación Estructura-Actividad , Triazinas/química , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Clin Cancer Res ; 12(20 Pt 1): 6186-93, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17062696

RESUMEN

PURPOSE: The studies described here are intended to characterize the ability of BMS-599626, a small-molecule inhibitor of the human epidermal growth factor receptor (HER) kinase family, to modulate signaling and growth of tumor cells that depend on HER1 and/or HER2. EXPERIMENTAL DESIGN: The potency and selectivity of BMS-599626 were assessed in biochemical assays using recombinant protein kinases, as well as in cell proliferation assays using tumor cell lines with varying degrees of dependence on HER1 or HER2 signaling. Modulation of receptor signaling was determined in cell assays by Western blot analyses of receptor autophosphorylation and downstream signaling. The ability of BMS-599626 to inhibit receptor heterodimer signaling in tumor cells was studied by receptor coimmunoprecipitation. Antitumor activity of BMS-599626 was evaluated using a number of different xenograft models that represent a spectrum of human tumors with HER1 or HER2 overexpression. RESULTS: BMS-599626 inhibited HER1 and HER2 with IC50 of 20 and 30 nmol/L, respectively, and was highly selective when tested against a broad panel of diverse protein kinases. Biochemical studies suggested that BMS-599626 inhibited HER1 and HER2 through distinct mechanisms. BMS-599626 abrogated HER1 and HER2 signaling and inhibited the proliferation of tumor cell lines that are dependent on these receptors, with IC50 in the range of 0.24 to 1 micromol/L. BMS-599626 was highly selective for tumor cells that depend on HER1/HER2 and had no effect on the proliferation of cell lines that do not express these receptors. In tumor cells that are capable of forming HER1/HER2 heterodimers, BMS-599626 inhibited heterodimerization and downstream signaling. BMS-599626 had antitumor activity in models that overexpress HER1 (GEO), as well as in models that have HER2 gene amplification (KPL4) or overexpression (Sal2), and there was good correlation between the inhibition of receptor signaling and antitumor activity. CONCLUSIONS: BMS-599626 is a highly selective and potent inhibitor of HER1 and HER2 kinases and inhibits tumor cell proliferation through modulation of receptor signaling. BMS-599626 inhibits HER1/HER2 receptor heterodimerization and provides an additional mechanism of inhibiting tumors in which receptor coexpression and heterodimerization play a major role in driving tumor growth. The preclinical data support the advancement of BMS-599626 into clinical development for the treatment of cancer.


Asunto(s)
Antineoplásicos/toxicidad , Inhibidores Enzimáticos/toxicidad , Receptores ErbB/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacología , Antígenos CD8/inmunología , División Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Dimerización , Humanos
10.
Mol Cancer Ther ; 5(6): 1571-6, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16818516

RESUMEN

Receptor tyrosine kinases (RTK) remain an area of therapeutic interest because of their role in epithelial tumors, and experimental models specific to these targets are highly desirable. Chimeric receptors were prepared by in-frame fusion of the CD8 extracellular sequence with the cytoplasmic sequences of RTKs. A CD8HER2 fusion protein was shown to form disulfide-mediated homodimers and to transform fibroblasts and epithelial cells. CD8RTK fusion proteins transform rat kidney epithelial cells and impart phenotypes that may reflect signaling specificity inherent in the native receptors. Transgenic expression of CD8HER2 and CD8Met in mice resulted in the formation of salivary and mammary gland tumors. The transgenic tumors allow the derivation of allograft tumors and cell lines that are sensitive to inhibition by small molecule kinase inhibitors. This approach provides excellent cell and tumor models for the characterization of signaling properties of diverse RTKs and for the evaluation of rationally designed antagonists targeting these kinases.


Asunto(s)
Antígenos CD8/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias Mamarias Animales/genética , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/genética , Neoplasias de las Glándulas Salivales/genética , Animales , Western Blotting , Transformación Celular Neoplásica/genética , Dimerización , Modelos Animales de Enfermedad , Disulfuros/farmacología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/etiología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Fragmentos de Péptidos/inmunología , Plásmidos , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Ratas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Neoplasias de las Glándulas Salivales/tratamiento farmacológico , Neoplasias de las Glándulas Salivales/etiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...