Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Pharmacol Exp Ther ; 324(3): 894-901, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18083913

RESUMEN

The alpha(V) integrins are key receptors involved in mediating cell migration and angiogenesis. In age-related macular degeneration (AMD) and diabetic retinopathy, angiogenesis plays a critical role in the loss of vision. These ocular vasculopathies might be treatable with a suitable alpha(V) antagonist, and an oral drug would offer a distinct advantage over current therapies. (3,S,beta,S)-1,2,3,4-Tetrahydro-beta-[[1-[1-oxo-3-(1,5,6,7-tetrahydro-1,8-naphthyridin-2-yl)propyl]-4-piperidinyl]methyl]-3-quinolinepropanoic acid (JNJ-26076713) is a potent, orally bioavailable, nonpeptide alpha(V) antagonist derived from the arginine-glycine-asparagine binding motif in the matrix protein ligands (e.g., vitronectin). This compound inhibits alpha(V)beta(3) and alpha(V)beta(5) binding to vitronectin in the low nanomolar range, it has excellent selectivity over integrins alpha(IIb)beta(3) and alpha(5)beta(1), and it prevents adhesion to human, rat, and mouse endothelial cells. JNJ-26076713 blocks cell migration induced by vascular endothelial growth factor, fibroblast growth factor (FGF), and serum, and angiogenesis induced by FGF in the chick chorioallantoic membrane model. JNJ-26076713 is the first alpha(V) antagonist reported to inhibit retinal neovascularization in an oxygen-induced model of retinopathy of prematurity after oral administration. In diabetic rats, orally administered JNJ-26076713 markedly inhibits retinal vascular permeability, a key early event in diabetic macular edema and AMD. Given this profile, JNJ-26076713 represents a potential therapeutic candidate for the treatment of age-related macular degeneration, macular edema, and proliferative diabetic retinopathy.


Asunto(s)
Permeabilidad Capilar/fisiología , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Integrina alfaV/metabolismo , Naftiridinas/administración & dosificación , Naftiridinas/farmacocinética , Quinolinas/administración & dosificación , Quinolinas/farmacocinética , Neovascularización Retiniana/metabolismo , Administración Oral , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacocinética , Animales , Disponibilidad Biológica , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Embrión de Pollo , Diabetes Mellitus Experimental/tratamiento farmacológico , Retinopatía Diabética/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Naftiridinas/química , Embarazo , Quinolinas/química , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Neovascularización Retiniana/tratamiento farmacológico
2.
Mol Cancer Ther ; 4(8): 1198-204, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16093435

RESUMEN

With the advent of agents directed against specific molecular targets in drug discovery, it has become imperative to show a compound's cellular impact on the intended biomolecule in vivo. The objective of the present study was to determine if we could develop an assay to validate the in vivo effects of a compound. Hence, we investigated the in vivo pharmacodynamic activity of JNJ-10198409, a relatively selective inhibitor of platelet-derived growth factor receptor tyrosine kinase (PDGF-RTK), in tumor tissues after administering the compound orally in a nude mouse xenograft model of human LoVo colon cancer. We developed a novel assay to quantify the in vivo anti-PDGF-RTK activity of the inhibitor in tumor tissue by determining the phosphorylation status of phospholipase Cgamma1 (PLCgamma1), a key downstream cellular molecule in the PDGF-RTK signaling cascade. We used two antibodies, one specific for the total (phosphorylated and unphosphorylated forms) PLCgamma1 (pan-PLCgamma1) and the other, specific for phosphorylated form of PLCgamma1 (ph-PLCgamma1) to immunohistochemically detect their expression in tumor tissues. Computer-assisted image analysis was then used to directly compare the ratio of ph-PLCgamma1 to pan-PLCgamma1 immunolabeling intensities in serial sections (5 mum) of tumors obtained from vehicle- and JNJ-10198409-treated tumor-bearing mice. Our data showed statistically significant, dose-dependent differences in the ph-PLC/pan-PLC ratio among the four treatment groups (vehicle, 25, 50, and 100 mg/kg b.i.d.). These results confirmed this compound's ability to suppress PDGF-RTK downstream signaling in tumor tissues in vivo. In addition to this specific application of this in vivo validation approach to those targets that use PLCgamma as a downstream signaling partner, these methods may also benefit other drug discovery targets.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Indanos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Anticuerpos Fosfo-Específicos/inmunología , Antineoplásicos/química , Neoplasias del Colon/enzimología , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Indanos/química , Ratones , Fosfolipasa C gamma , Fosforilación , Inhibidores de Proteínas Quinasas/química , Pirazoles/química , Fosfolipasas de Tipo C/análisis , Fosfolipasas de Tipo C/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Med Chem ; 48(26): 8163-73, 2005 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-16366598

RESUMEN

A series of (6,7-dimethoxy-2,4-dihydroindeno[1,2-c]pyrazol-3-yl)phenylamines has been optimized to preserve both potent kinase inhibition activity against the angiogenesis target, the receptor tyrosine kinase of Platelet-Derived Growth Factor-BB (PDGF-BB), and to improve the broad tumor cell antiproliferative activity of these compounds. This series culminates in the discovery of 17 (JNJ-10198409), a compound with anti-PDGFR-beta kinase activity (IC(50)=0.0042 microM) and potent antiproliferative activity in six of eight human tumor cell lines (IC(50) < 0.033 microM).


Asunto(s)
Antineoplásicos/farmacología , Indanos/síntesis química , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/síntesis química , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Antineoplásicos/síntesis química , Becaplermina , Línea Celular , Línea Celular Tumoral , Endotelio Vascular/efectos de los fármacos , Humanos , Indanos/farmacología , Concentración 50 Inhibidora , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-sis , Pirazoles/farmacología
4.
J Pharm Sci ; 101(7): 2545-56, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22473491

RESUMEN

Extensive first-pass metabolism can significantly limit a drug's oral exposure levels. In this work, we introduce an innovative approach for increasing the oral bioavailability of a drug that undergoes extensive reversible glucuronidation and enterohepatic recirculation through intraduodenal coadministration of the deconjugating enzyme ß-glucuronidase. Intraduodenal administration of JNJ-10198409 (10 mg/kg) with ß-glucuronidase (34,000-140,000 units/kg) to catheterized rats resulted in a significant increase (p < 0.005) in the mean area under the plasma concentration versus time curve (AUC; approx. threefold) and maximum plasma concentration (C(max); approx. twofold) of JNJ-10198409. The AUC and C(max) were 60 ± 18 ng h/mL and 76 ± 29 ng/mL, respectively, with no enzyme and 177 ± 55 ng h/mL and 129 ± 41 ng/mL, respectively, with ß-glucuronidase coadministered. Moreover, the AUC of the primary glucuronide metabolite increased approximately sevenfold from 1173 ± 361 (ng h)/mL with no enzyme coadministered to 8723 ± 2133 ng h/mL with coadministered enzyme. These pharmacokinetic data support the hypothesis that when the primary glucuronide is secreted into the duodenum via the bile duct, the glucuronide is converted by ß-glucuronidase back to the parent compound. The parent compound is then reabsorbed and reconjugated, resulting in elevated systemic exposures to both parent and glucuronide. Potential clinical and preclinical applications and considerations for this approach are discussed.


Asunto(s)
Glucuronidasa/administración & dosificación , Glucurónidos/metabolismo , Indanos/administración & dosificación , Indanos/farmacocinética , Pirazoles/administración & dosificación , Pirazoles/farmacocinética , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Administración Oral , Animales , Área Bajo la Curva , Disponibilidad Biológica , Glucuronidasa/metabolismo , Indanos/metabolismo , Absorción Intestinal , Masculino , Pirazoles/metabolismo , Ratas , Ratas Sprague-Dawley
5.
Mol Cancer Ther ; 8(11): 3151-61, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19887542

RESUMEN

There is increasing evidence that tumor-associated macrophages promote the malignancy of some cancers. Colony-stimulating factor-1 (CSF-1) is expressed by many tumors and is a growth factor for macrophages and mediates osteoclast differentiation. Herein, we report the efficacy of a novel orally active CSF-1 receptor (CSF-1R) kinase inhibitor, JNJ-28312141, in proof of concept studies of solid tumor growth and tumor-induced bone erosion. H460 lung adenocarcinoma cells did not express CSF-1R and were not growth inhibited by JNJ-28312141 in vitro. Nevertheless, daily p.o. administration of JNJ-28312141 caused dose-dependent suppression of H460 tumor growth in nude mice that correlated with marked reductions in F4/80(+) tumor-associated macrophages and with increased plasma CSF-1, a possible biomarker of CSF-1R inhibition. Furthermore, the tumor microvasculature was reduced in JNJ-28312141-treated mice, consistent with a role for macrophages in tumor angiogenesis. In separate studies, JNJ-28312141 was compared with zoledronate in a model in which MRMT-1 mammary carcinoma cells inoculated into the tibias of rats led to severe cortical and trabecular bone lesions. Both agents reduced tumor growth and preserved bone. However, JNJ-28312141 reduced the number of tumor-associated osteoclasts superior to zoledronate. JNJ-28312141 exhibited additional activity against FMS-related receptor tyrosine kinase-3 (FLT3). To more fully define the therapeutic potential of this new agent, JNJ-28312141 was evaluated in a FLT3-dependent acute myeloid leukemia tumor xenograft model and caused tumor regression. In summary, this novel CSF-1R/FLT3 inhibitor represents a new agent with potential therapeutic activity in acute myeloid leukemia and in settings where CSF-1-dependent macrophages and osteoclasts contribute to tumor growth and skeletal events.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Imidazoles/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Animales , Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Ratas , Ratas Sprague-Dawley , Receptor de Factor Estimulante de Colonias de Macrófagos/sangre , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Especificidad por Sustrato , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa 3 Similar a fms/metabolismo
6.
Angiogenesis ; 7(2): 91-6, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15516829

RESUMEN

The protein processing enzyme, methionine aminopeptidase-2 (MetAP-2), has been identified as a molecular target of fumagillin and its derivative, TNP-470, compounds known to inhibit endothelial cell proliferation and angiogenesis. A high-throughput screening program was undertaken to identify selective, reversible inhibitors of MetAP-2 in an attempt to discover structurally novel anti-angiogenic agents for potential therapeutic use in oncology. Approximately 90 small-molecule, reversible, selective inhibitors of rhMetAP-2 were identified. The most potent of these compounds contained a singly-substituted triazole moiety which exhibited an IC50 of 8 nM (95% confidence limits 5 to 13 nM) and was highly selective for MetAP-2 over MetAP-1 (approximately 60-fold difference in IC50 values). Unlike fumagillin, these MetAP-2 inhibitors failed to significantly inhibit growth factor-stimulated endothelial cell (HUVEC) proliferation or to suppress angiogenesis in the in vitro aortic ring explant model of microvessel outgrowth. The MetAP-2-inhibitory activity of these compounds was dependent on the divalent cation used as the metalloenzyme activating cofactor for MetAP-2. These inhibitors were identified using cobalt(II)-activated recombinant human MetAP-2 for screening compound libraries. When manganese (Mn2+) was substituted for cobalt following EDTA treatment and extensive dialysis of the MetAP-2 protein, these inhibitors were significantly less potent (40-fold increase in IC50) as inhibitors of MetAP-2. These results support the recent hypothesis that cobalt may not be the relevant divalent metal ion cofactor for MetAP-2 in cells and may explain the observed absence of cell-based activity using potent triazole inhibitors of cobalt-activated MetAP-2.


Asunto(s)
Aminopeptidasas/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Proliferación Celular/efectos de los fármacos , Endotelio Vascular/citología , Glicoproteínas/antagonistas & inhibidores , Metaloendopeptidasas/antagonistas & inhibidores , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de la Angiogénesis/química , Aorta , Ciclohexanos , Evaluación Preclínica de Medicamentos , Endotelio Vascular/fisiología , Inhibidores Enzimáticos/farmacología , Ácidos Grasos Insaturados/farmacología , Humanos , Metionil Aminopeptidasas , O-(Cloroacetilcarbamoil) Fumagilol , Sesquiterpenos/farmacología , Relación Estructura-Actividad , Venas Umbilicales
8.
Bioorg Med Chem Lett ; 14(20): 5227-32, 2004 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-15380233

RESUMEN

The synthesis and SAR of a new class of piperidine-based alphavbeta3/alphavbeta5 integrin antagonists is described. Replacement of an amide bond in a prototype isonipecotamide by a C-C isostere, and adjustment of the spacer length between the carboxylic acid and basic moieties, led to low nanomolar antagonists of alphavbeta3 and/or alphavbeta5 integrins with excellent selectivity versus alpha(IIb)beta3.


Asunto(s)
Integrina alfaVbeta3/antagonistas & inhibidores , Integrinas/antagonistas & inhibidores , Piperidinas/síntesis química , Propionatos/síntesis química , Receptores de Vitronectina/antagonistas & inhibidores , Administración Oral , Animales , Disponibilidad Biológica , Humanos , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Piperidinas/química , Piperidinas/farmacología , Propionatos/química , Propionatos/farmacología , Unión Proteica , Ratas , Receptores de Vitronectina/metabolismo , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 14(23): 5937-41, 2004 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-15501072

RESUMEN

Reduction of the quinoline ring in an alpha(v)beta(3) antagonist yielded a 1,2,3,4-tetrahydro derivative as two diastereomers, the four isomers of which were separated by sequential chiral HPLC. Two isomers had significant alpha(V)beta(3) antagonist activity with improved oral bioavailability, relative to the corresponding quinoline derivative.


Asunto(s)
Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/metabolismo , Quinolinas/química , Quinolinas/metabolismo , Administración Oral , Animales , Disponibilidad Biológica , Humanos , Quinolinas/administración & dosificación , Ratas
10.
Mol Pharmacol ; 66(3): 635-47, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15322256

RESUMEN

Inhibition of angiogenesis may have wide use in the treatment of cancer; however, this approach alone will not cause tumor regression but may only slow the growth of solid tumors. The clinical potential of antiangiogenic agents may be increased by combining them with conventional chemotherapeutics. 4-[4-(1-Amino-1-methylethyl)phenyl]-2-[4-(2-morpholin-4-yl-ethyl)phenylamino]pyrimidine-5-carbonitrile (JNJ-17029259) represents a novel structural class of 5-cyanopyrimidines that are orally available, selective, nanomolar inhibitors of the vascular endothelial growth factor receptor-2 (VEGF-R2) and other tyrosine kinases involved in angiogenesis, such as platelet-derived growth factor receptor, fibroblast growth factor receptor, VEGF-R1, and VEGF-R3, but have little activity on other kinase families. At nanomolar levels, JNJ-17029259 blocks VEGF-stimulated mitogen-activated protein kinase signaling, proliferation/migration, and VEGF-R2 phosphorylation in human endothelial cells; inhibits the formation of vascular sprouting in the rat aortic ring model of angiogenesis; and interferes with the development of new veins and arteries in the chorioallantoic membrane assay. At higher concentrations of 1 to 3 microM, this compound shows antiproliferative activity on cells that may contribute to its antitumor effects. JNJ-17029259 delays the growth of a wide range of human tumor xenografts in nude mice when administered orally as single-agent therapy. Histological examination revealed that the tumors have evidence of reduced vascularity after treatment. In addition, JNJ-17029259 enhances the effects of the conventional chemotherapeutic drugs doxorubicin and paclitaxel in xenograft models when administered orally in combination therapy. An orally available angiogenesis inhibitor that can be used in conjunction with standard chemotherapeutic agents to augment their activity may have therapeutic benefit in stopping the progression of cancer and preventing metastasis.


Asunto(s)
Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Neoplasias Experimentales/tratamiento farmacológico , Nitrilos/uso terapéutico , Paclitaxel/uso terapéutico , Pirimidinas/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Quimioterapia Combinada , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ratones , Nitrilos/farmacología , Pirimidinas/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA