Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cancer Res ; 65(19): 9038-46, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16204078

RESUMEN

Modulation of aberrant cell cycle regulation is a potential therapeutic strategy applicable to a wide range of tumor types. JNJ-7706621 is a novel cell cycle inhibitor that showed potent inhibition of several cyclin-dependent kinases (CDK) and Aurora kinases and selectively blocked proliferation of tumor cells of various origins but was about 10-fold less effective at inhibiting normal human cell growth in vitro. In human cancer cells, treatment with JNJ-7706621 inhibited cell growth independent of p53, retinoblastoma, or P-glycoprotein status; activated apoptosis; and reduced colony formation. At low concentrations, JNJ-7706621 slowed the growth of cells and at higher concentrations induced cytotoxicity. Inhibition of CDK1 kinase activity, altered CDK1 phosphorylation status, and interference with downstream substrates such as retinoblastoma were also shown in human tumor cells following drug treatment. Flow cytometric analysis of DNA content showed that JNJ-7706621 delayed progression through G1 and arrested the cell cycle at the G2-M phase. Additional cellular effects due to inhibition of Aurora kinases included endoreduplication and inhibition of histone H3 phosphorylation. In a human tumor xenograft model, several intermittent dosing schedules were identified that produced significant antitumor activity. There was a direct correlation between total cumulative dose given and antitumor effect regardless of the dosing schedule. These results show the therapeutic potential of this novel cell cycle inhibitor and support clinical evaluation of JNJ-7706621.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Triazoles/farmacología , Animales , Aurora Quinasas , Línea Celular Tumoral , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Femenino , Células HeLa , Humanos , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cancer Res ; 63(19): 6162-5, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14559797

RESUMEN

Tumor oxygenation is known to be an important predictive/prognostic marker in a variety of tumors, including cervix, head/neck, sarcoma, non-small cell of the lung, and breast. Tumor oxygenation is influenced by many interactions, including oxygen delivery (angiogenesis, permeability, and HgB) and consumption (metabolic and growth rates). This study randomized 30 nonanemic, female Fischer 344 rats into three treatment arms to examine the effects of recombinant human erythropoietin (EPO) on R3230 rodent mammary carcinoma oxygenation. The three treatment arms were: (a) placebo; (b) EPO after tumor implantation (2000 units/kg/SQdose, M/W/F for six doses); and (c) EPO before tumor implantation (2000 units/kg/SQdose, M/W/F for six doses). Tumors were implanted in the hindflank, and in vivo oxygenation was measured at day 22 after implantation using the Oxylite system (Oxford Optronix, Oxford, England). An average of 180 measurements/animal were performed. On day 22, median tumor volume was 399 mm(3) (range: 65-950 mm(3)), and no differences in tumor volume were seen between treatment arms. Mean hematocrit was equal between arms at therapy initiation but were significantly higher for both arms receiving EPO at day 22 (placebo versus Arm B versus Arm C; Wilcoxon P = 0.052). EPO-treated tumors had significantly less hypoxic measurements when compared with either the placebo or those receiving EPO before implantation. These data confirm that tumor oxygenation in nonanemic individuals may be improved through the administration of EPO, and this improvement appears to be independent of HgB effects.


Asunto(s)
Adenocarcinoma/sangre , Adenocarcinoma/tratamiento farmacológico , Eritropoyetina/farmacología , Hemoglobinas/metabolismo , Neoplasias Mamarias Experimentales/sangre , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Oxígeno/sangre , Animales , Femenino , Humanos , Trasplante de Neoplasias , Oxígeno/metabolismo , Presión Parcial , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes
3.
J Med Chem ; 48(13): 4208-11, 2005 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15974571

RESUMEN

A series of 1-acyl-1H-[1,2,4]triazole-3,5-diamine analogues were synthesized as cyclin-dependent kinase (CDK) inhibitors. These compounds showed potent and selective CDK1 and CDK2 inhibitory activities and inhibited in vitro cellular proliferation in various human tumor cells. Representative compound 3b demonstrated in vivo efficacy in a human melanoma A375 xenograft model in nude mice.


Asunto(s)
Antineoplásicos/síntesis química , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Diaminas/síntesis química , Inhibidores Enzimáticos/síntesis química , Triazoles/síntesis química , Administración Oral , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Disponibilidad Biológica , Diaminas/farmacocinética , Diaminas/farmacología , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Ratones , Ratones Desnudos , Estructura Molecular , Ratas , Relación Estructura-Actividad , Triazoles/farmacocinética , Triazoles/farmacología
4.
J Med Chem ; 48(14): 4535-46, 2005 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-15999992

RESUMEN

On the basis of previous studies, we identified pyrazine-pyridine A as a potent vascular endothelial growth factor inhibitor and pyrimidine-pyridine B as a moderately potent cyclin dependent kinase (CDK) inhibitor. A proposed combination of CGP-60474 and compound B led to the discovery of [1,3,5]triazine-pyridine as a new series of potent CDK inhibitors. Palladium-catalyzed C-C bond formation reactions, particularly the Negishi coupling reaction, were used to assemble various triazine-heteroaryl analogues effectively. Among them, compound 20 displayed high inhibitory potency at CDK1 (IC(50) = 0.021 microM), CDK2, and CDK5 and submicromolar potency at CDK4, CDK6, and CDK7. Compound 20 also displayed high potency at GSK-3beta. It demonstrated potent antiproliferative activity on various tumor cell lines, including HeLa, HCT-116, U937, and A375. When 20 was administered intraperitoneally at 150 and 125 mg/kg to nude mice bearing human A375 xenografts, the compound produced a significant survival increase. Molecular docking studies were conducted in an attempt to enhance the understanding of the observed structure-activity relationship.


Asunto(s)
Aminopiridinas/síntesis química , Antineoplásicos/síntesis química , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Piridinas/síntesis química , Tiazinas/síntesis química , Triazinas/síntesis química , Aminopiridinas/química , Aminopiridinas/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Piridinas/química , Piridinas/farmacología , Tiazinas/química , Tiazinas/farmacología , Trasplante Heterólogo , Triazinas/química , Triazinas/farmacología
5.
J Med Chem ; 48(6): 1886-900, 2005 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-15771433

RESUMEN

Pathological angiogenesis is associated with disease states such as cancer, diabetic retinopathy, rheumatoid arthritis, endometriosis, and psoriasis. There is much evidence that direct inhibition of the kinase activity of vascular endothelial growth factor receptor-2 (VEGFR-2) will result in the reduction of angiogenesis and the suppression of tumor growth. Attempts to optimize a cyclin-dependent kinase-1 (CDK1) inhibitor by using palladium-catalyzed C-C bond, C-N bond formation reactions to assemble diverse biheteroaryl molecules led to the unexpected discovery of a pyrazine-pyridine biheteroaryl as a novel series of potent VEGFR-2 inhibitors. Compound 15, which had IC(50) = 0.084 microM at VEGFR-2, showed very modest selectivity against fibroblast growth factor receptor-2 (IC(50) = 0.21 microM), platelet-derived growth factor receptor (IC(50) = 0.36 microM), and glycogen synthase kinase-3 (IC(50) = 0.478 microM), while it exhibited more than 10-fold selectivity against epidermal growth factor receptor (IC(50) = 1.36 microM) and insulin-R kinase (IC(50) = 1.69 microM). On the other hand, compound 15 exhibited more than 100-fold selectivity against calmodulin kinase 2; casein kinase-1 and -2; CDK1 and -4; mitogen-activated protein kinase; and protein kinase A, Cbeta2, and Cgamma (IC(50) >10 microM). Compound 15 also displayed high inhibitory potency on VEGF-stimulated human umbilical vein endothelial cell (HUVEC) proliferation (IC(50) = 0.005 microM) and good selectivity against cell lines such as HUVEC, human aortic smooth muscle cells, and MRC5 lung fibroblasts. Molecular docking studies were conducted in an attempt to rationalize the unexpected high VEGFR-2 selectivity of 15.


Asunto(s)
Inhibidores de la Angiogénesis/síntesis química , Compuestos Heterocíclicos con 2 Anillos/síntesis química , Pirazinas/síntesis química , Piridinas/síntesis química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Compuestos Heterocíclicos con 2 Anillos/química , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Modelos Moleculares , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Pirazinas/química , Pirazinas/farmacología , Piridinas/química , Piridinas/farmacología , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química
6.
J Med Chem ; 48(15): 4892-909, 2005 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-16033269

RESUMEN

There is much evidence that direct inhibition of the kinase activity of vascular endothelial growth factor receptor-2 (VEGFR-2) will result in the reduction of angiogenesis and the suppression of tumor growth. Palladium-catalyzed C-C bond, C-N bond formation reactions were used to assemble various pyrazine-pyridine biheteroaryls as potent VEGFR-2 inhibitors. Among them, 4-{5-[6-(3-chloro-phenylamino)-pyrazin-2-yl]-pyridin-3-ylamino}-butan-1-ol (39) and N-{5-[6-(3-chloro-phenylamino)-pyrazin-2-yl]-pyridin-3-yl}-N',N'-dimethyl-ethane-1,2-diamine (41) exhibited the highest kinase selectivity against fibroblast growth factor receptor kinase, platelet-derived growth factor receptor kinase, and glycogen synthase kinase-3. All of these compounds showed good cellular potency to inhibit VEGF-stimulated proliferation of human umbilical vein endothelial cells (HUVEC) but with modest effects on the unstimulated growth of HUVEC. The low inhibition of these compounds to the growth of tumor cell lines, such as HeLa, HCT-116, and A375 further confirms that these VEGFR-2 inhibitors are not cytotoxic agents. The in vivo antitumor activity of 39 and 41 were demonstrated in the A375 human melanoma xenograft nude mice model. Molecular modeling (QSAR analysis) was conducted in an attempt to rationalize the observed structure-activity relationship.


Asunto(s)
Aminopiridinas/síntesis química , Pirazinas/síntesis química , Piridinas/síntesis química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Aminopiridinas/química , Aminopiridinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Endotelio Vascular/citología , Femenino , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Pirazinas/química , Pirazinas/farmacología , Piridinas/química , Piridinas/farmacología , Relación Estructura-Actividad Cuantitativa , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Med Chem ; 46(19): 4021-31, 2003 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-12954055

RESUMEN

Attempts to design the macrocyclic maleimides as selective protein kinase C gamma inhibitors led to the unexpected discovery of a novel series of potent and highly selective glycogen synthase kinase-3beta (GSK-3beta) inhibitors. Palladium-catalyzed cross-coupling reactions were used to synthesize the key intermediates 17 and 22 that resulted in the synthesis of novel macrocycles. All three macrocyclic series (bisindolyl-, mixed 7-azaindoleindolyl-, and bis-7-azaindolylmaleimides) were found to have submicromolar inhibitory potency at GSK-3beta with various degrees of selectivity toward other protein kinases. To gain the inhibitory potency at GSK-3beta, the ring sizes of these macrocycles may play a major role. To achieve the selectivity at GSK-3beta, the additional nitrogen atoms in the indole rings may contribute to a significant degree. Overall, the bis-7-azaindolylmaleimides 28 and 29 exhibited little or no inhibitions to a panel of 50 protein kinases. Compound 29 almost behaved as a GSK-3beta specific inhibitor. Both 28 and 29 displayed good potency in GS cell-based assay. Molecular docking studies were conducted in an attempt to rationalize the GSK-3beta selectivity of azaindolylmaleimides.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Éteres Cíclicos/síntesis química , Éteres Cíclicos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Maleimidas/síntesis química , Maleimidas/farmacología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Células Cultivadas , Diseño de Fármacos , Éteres Cíclicos/química , Glucógeno Sintasa Quinasa 3/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Maleimidas/química , Modelos Moleculares , Datos de Secuencia Molecular , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/metabolismo , Ratas , Alineación de Secuencia , Relación Estructura-Actividad , Especificidad por Sustrato
8.
Brain Res Mol Brain Res ; 104(1): 86-95, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12117554

RESUMEN

Erythropoietin (EPO) is the primary modulator of red blood cell production. Recently EPO has received considerable attention for its functions outside of hematopoiesis, including its effects in the nervous system where it has been shown to act as a neuroprotectant. To understand the function of EPO in the nervous system and to determine if EPO functions through the same signaling pathways identified in hematopoietic cells, we used cDNA array hybridization and RT-PCR to investigate the changes in gene expression induced by EPO in the neuronal-like PC-12 cell line. PC-12 cells cultured in the presence of EPO (10 U/ml) showed significant changes in gene expression by 3 h with a return to basal expression levels for the vast majority of genes by 24 h. The genes influenced by EPO included genes with known functions in cell proliferation, differentiation and apoptosis. Semi-quantitative RT-PCR confirmed that 24 h pre-treatment with EPO (10 pM) resulted in a 2.5-fold increase in the expression of the anti-apoptotic gene bcl(XL) and a 4-fold decrease in the expression of the pro-apoptotic gene bak. In addition to supporting the current models of EPO function these results suggest previously unidentified mechanisms by which EPO may function in neurons.


Asunto(s)
Diferenciación Celular/genética , División Celular/genética , Supervivencia Celular/genética , Eritropoyetina/metabolismo , Regulación de la Expresión Génica/fisiología , Neuronas/metabolismo , Transducción de Señal/genética , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Eritropoyetina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de la Membrana/genética , Neuronas/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Células PC12 , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Proteína Destructora del Antagonista Homólogo bcl-2 , Proteína bcl-X
9.
Growth Factors ; 24(4): 268-78, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17381068

RESUMEN

Chondrogenic promotion by rhGDF5 with or without rhTGFbeta3 was studied in pellet culture of human mesenchymal stem cells (HMSCs). A synergy between rhGDF5 and rhTGFbeta3 was observed in promoting chondrogenesis. rhBMP2, rhBMP6, rhBMP7 and rhTGFbeta1 were further tested and showed the same effect. To explore the mechanism, the expression of TGFbetatype I and II receptors, ALK5, ALK2, ALK3, ALK6, TGFbetaRII, BMPRII, ActRII was studied. ALK6 showed increase by the rhTGFbeta1 or rhTGFbeta3 treatment. ALK6 protein expression also showed increase by rhTGFbeta3. rhTGFbeta1/rhTGFbeta3 induced ALK6 up-regulation was inhibited by SD-208, a TGFbeta type I receptor inhibitor. Chondrogenesis by rhTGFbetal/rhTGFbeta3 or the combination between rhTGFbetal/rhTGFbeta3 and rhGDF5 also was diminished by SD-208. SMAD1/5/8 phosphorylation in nascent human mesenchymal stem cells (HMSCs) was stimulated weakly by rhGDF5 but strongly by rhBMP7. The rhGDF5 stimulated SMAD1/5/8 phosphorylation was enhanced by rhTGFbetal/rhTGFbeta3 but inhibited by SD-208. The rhBMP7 stimulated SMAD1/5/8 phosphorylation did not show influence by rhTGFbeta3 and SD-208. Our results indicated the potential involvement of ALK6 activation by rhTGFbetas in the synergy between rhTGFbetas and rhBMPs.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Condrogénesis , Células Madre Mesenquimatosas/fisiología , Factor de Crecimiento Transformador beta3/farmacología , Receptores de Activinas Tipo I/metabolismo , Proteína Morfogenética Ósea 7 , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/fisiología , Sinergismo Farmacológico , Factor 5 de Diferenciación de Crecimiento , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Fosforilación , Pteridinas/farmacología , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba
11.
Oncologist ; 9 Suppl 5: 41-7, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15591421

RESUMEN

Within the past decade, clinical trials have shown that the presence of anemia can diminish the physical status, functional abilities, and overall quality of life (QOL) of cancer patients and can negatively influence the outcome of their treatment. However, recent preclinical and clinical studies have also shown that increasing hemoglobin levels by administering recombinant human erythropoietin (rHuEPO, epoetin alfa) may ameliorate anemia and, in doing so, improve QOL and possibly result in better treatment outcomes following radiotherapy, chemotherapy, or a combination of these modalities. Several mechanisms by which rHuEPO may improve treatment outcome have been proposed, including correction of tumor hypoxia, increased sensitivity of tumor cells to radiotherapy and chemotherapy, correction of anemia and its associated symptoms (particularly fatigue), and immune-modulated effects of rHuEPO on tumor growth. Improvement of tumor oxygenation by rHuEPO could affect treatment outcome in two ways. First, correction of hypoxia results in the downregulation of hypoxia-inducible factor 1 (HIF-1), a key regulator of cellular adaptive responses to hypoxia (e.g., angiogenesis), including many pathways that are important for tumor growth and metastasis. Interruption of the HIF-1 pathway not only limits growth of the primary tumor but also reduces the potential for the development of more aggressive tumors and metastatic spread, which could ultimately improve treatment outcome. Second, within the tumor, it is the hypoxic cells that are resistant to oxygen-dependent radiotherapy and chemotherapy, and improvement in their oxygenation would increase their sensitivity to the cytotoxic effects of such treatment. Correction of anemia and its associated symptoms, particularly fatigue, can have a beneficial effect on patient QOL, and this in turn may translate into greater tolerance of radiotherapy and chemotherapy, allowing patients to receive full doses and on-schedule dosing, and thus have an increased likelihood of a therapeutic response. Lastly, results of a study using a murine model of multiple myeloma have indicated that rHuEPO may induce an immune-mediated antitumor effect. Therefore, additional research is warranted to further explore the biologic actions of rHuEPO and to determine their relevance to therapeutic outcome.


Asunto(s)
Anemia/tratamiento farmacológico , Anemia/etiología , Hipoxia de la Célula , Eritropoyetina/farmacología , Animales , Ensayos Clínicos como Asunto , Terapia Combinada , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Resistencia a Antineoplásicos , Eritropoyetina/uso terapéutico , Fatiga , Humanos , Ratones , Mieloma Múltiple/complicaciones , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Calidad de Vida , Tolerancia a Radiación , Proteínas Recombinantes , Linfocitos T/inmunología , Resultado del Tratamiento
12.
Bioorg Med Chem Lett ; 12(16): 2145-8, 2002 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-12127524

RESUMEN

The syntheses and biological activities of a novel series of 2,4- and 2,5-disubstituted thiophenes are reported. These analogues have shown excellent affinity and selectivity against alpha(1)-adrenoreceptor subtypes.


Asunto(s)
Hiperplasia Prostática/tratamiento farmacológico , Tiofenos/síntesis química , Tiofenos/farmacología , Antagonistas Adrenérgicos alfa/síntesis química , Antagonistas Adrenérgicos alfa/química , Antagonistas Adrenérgicos alfa/farmacología , Sitios de Unión , Humanos , Masculino , Estructura Molecular , Relación Estructura-Actividad , Tiofenos/química
13.
Biol Neonate ; 85(2): 138-44, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14639039

RESUMEN

Erythropoietin (Epo) decreases neuronal injury and cell death in vitro and in vivo. To lay the groundwork for use of Epo as a potential therapy for brain injury, we tested the hypothesis that systemic dosing of high-dose recombinant Epo (rEpo) would result in neuroprotective rEpo concentrations in the spinal fluid of adult and developing animals. This report characterizes the pharmacokinetics of high-dose rEpo in the blood and spinal fluid of juvenile and adult nonhuman primates (n = 7) and fetal sheep (n = 37) following a single injection. Timed blood and spinal fluid samples were collected following rEpo injection. Epo accumulation in spinal fluid was dependent on peak serum concentration and time following injection. We demonstrate that high-dose rEpo was well tolerated and results in neuroprotective concentrations in spinal fluid of adult and developing animal models by 2-2.5 h after injection.


Asunto(s)
Envejecimiento/metabolismo , Eritropoyetina/administración & dosificación , Eritropoyetina/líquido cefalorraquídeo , Feto/metabolismo , Envejecimiento/sangre , Animales , Barrera Hematoencefálica , Relación Dosis-Respuesta a Droga , Eritropoyetina/sangre , Sangre Fetal , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Macaca nemestrina , Masculino , Concentración Osmolar , Proteínas Recombinantes , Ovinos
14.
J Rheumatol ; 29(9): 1907-13, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12233885

RESUMEN

OBJECTIVE: Treatment of autoimmune diseases with therapies that tolerize pathogenic lymphocytes may obviate the need for longterm global immunosuppression. In vitro, non-Fc receptor binding derivatives of anti-murine CD3 monoclonal antibodies tolerize type 1 T cells and stimulate type 2 T cells. Recently, a humanized non-FcR binding derivative of the anti-human CD3 Mab OKT3, huOKT3gamma1(ala-ala), has been described. We hypothesized that this Mab may be safe and efficacious in the treatment of type 1 T lymphocyte mediated chronic autoimmune diseases such as psoriatic arthritis (PsA). METHODS: In a Phase I/II trial, 7 patients with PsA were treated with escalating daily doses of huOKT3gamma1(ala-ala) for 12 to 14 days. Number of tender and swollen joints and a visual analog pain scale were used to rate disease activity at entry and Day 30 and Day 90 after treatment. RESULTS: At Day 30, 6 of 7 patients had > or = 75% improvement in the number of inflamed joints and an average 63% improvement on the patient pain scale. Two of 6 responders had sustained improvement at Day 90. No patient treated with an initial dose < or = 1 mg had significant side effects, nor did they have detectable increases in serum cytokines. One patient treated with 4 mg without escalation developed mild cytokine release symptoms associated with elevation of interleukin 10. Transient T cell depletion occurred following treatment with the maximum dose of 4 mg, which resolved by Day 30. Antiidiotypic antibodies developed in 2 patients; however, there was no concurrent decrease in efficacy. CONCLUSION: These data indicate that huOKT3gamma1(ala-ala) may be useful in treating PsA.


Asunto(s)
Artritis Psoriásica/tratamiento farmacológico , Dosis Máxima Tolerada , Muromonab-CD3/uso terapéutico , Adolescente , Adulto , Anciano , Artritis Psoriásica/diagnóstico , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
16.
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