Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 9(4): e95733, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24760018

RESUMEN

Alzheimer's disease is an irreversible neurodegenerative disorder that is characterized by the abnormal aggregation of amyloid-ß into neurotoxic oligomers and plaques. Although many disease-modifying molecules are currently in Alzheimer clinical trials, a small molecule that inhibits amyloid-ß aggregation and ameliorates the disorder has not been approved to date. Herein, we report the effects of a potent small molecule, 6-methoxy-2-(4-dimethylaminostyryl) benzofuran (KMS88009), that directly disrupts amyloid-ß oligomerization, preserving cognitive behavior when used prophylactically and reversing declines in cognitive behavior when used therapeutically. KMS88009 exhibited excellent pharmacokinetic profiles with extensive brain uptake and a high level of safety. When orally administered before and after the onset of Alzheimer's disease symptoms, KMS88009 significantly reduced assembly of amyloid-ß oligomers and improved cognitive behaviors in the APP/PS1 double transgenic mouse model. The unique dual mode of action indicates that KMS88009 may be a powerful therapeutic candidate for the treatment of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Benzofuranos/uso terapéutico , Trastornos del Conocimiento/tratamiento farmacológico , Animales , Benzofuranos/química , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos
2.
Bioorg Med Chem Lett ; 23(11): 3467-9, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23601707

RESUMEN

Alzheimer's disease drug discovery regarding exploration into the molecules and processes has focused on the intrinsic causes of the brain disorder correlated with the accumulation of amyloid-ß. An anti-amyloidogenic bis-styrylbenzene derivative, KMS80013, showed excellent oral bioavailability (F=46.2%), facilitated brain penetration (26%, iv) in mouse and target specific in vivo efficacy in acute AD mouse model attenuating the cognitive deficiency in Y-maze test. Acute toxicity (LD50 >2000 mg/kg) and hERG channel inhibition (14% at 10 µM) results indicated safety of KMS80013.


Asunto(s)
Compuestos de Anilina/química , Derivados del Benceno/química , Estilbenos/química , Administración Oral , Enfermedad de Alzheimer/tratamiento farmacológico , Compuestos de Anilina/farmacocinética , Compuestos de Anilina/uso terapéutico , Animales , Derivados del Benceno/farmacocinética , Derivados del Benceno/uso terapéutico , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Semivida , Masculino , Ratones , Ratones Endogámicos ICR , Estilbenos/farmacocinética , Estilbenos/uso terapéutico
3.
J Med Chem ; 55(6): 2846-57, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22372864

RESUMEN

A novel series of N(4)-(3-chlorophenyl)-5-(oxazol-2-yl)pyrimidine-4,6-diamines were synthesized and evaluated as dual inhibitors of HER-1/HER-2 tyrosine kinases. In contrast to the currently approved HER-2-targeted agent (lapatinib, 1), our irreversible HER-1/HER-2 inhibitors have the potential to overcome the clinically relevant and mutation-induced drug resistance. The selected compound (19a) showed excellent inhibitory activity toward HER-1/HER-2 tyrosine kinases with selectivity over 20 other kinases and inhibited the proliferation of both cancer cell types: lapatinib-sensitive cell lines (SK-Br3, MDA-MB-175, and N87) and lapatinib-resistant cell lines (MDA-MB-453, H1781, and H1975). The excellent pharmacokinetic profiles of 19a in mice and rats led us to further investigation of a novel therapeutic agent for HER-2-targeting treatment of solid tumors, especially HER-2-positive breast/gastric cancer and HER-2-mutated lung cancer.


Asunto(s)
Acrilamidas/síntesis química , Antineoplásicos/síntesis química , Receptores ErbB/antagonistas & inhibidores , Oxazoles/síntesis química , Pirimidinas/síntesis química , Receptor ErbB-2/antagonistas & inhibidores , Acrilamidas/farmacocinética , Acrilamidas/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Lapatinib , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Ratones , Ratones Endogámicos ICR , Modelos Moleculares , Mutación , Oxazoles/farmacocinética , Oxazoles/farmacología , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Relación Estructura-Actividad
4.
Int J Cancer ; 130(10): 2445-54, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21732342

RESUMEN

The epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases has been implicated in a variety of cancers. In particular, activating mutations such as the L858R point mutation in exon 21 and the small in-frame deletions in exon 19 of the EGFR tyrosine kinase domain are correlated with sensitivity to EGFR tyrosine kinase inhibitors in non-small cell lung cancer (NSCLC) patients. Clinical treatment of patients is limited by the development of drug resistance resulting mainly from a gatekeeper mutation (T790M). In this study, we evaluated the therapeutic potential of a novel, irreversible pan-HER inhibitor, HM781-36B. The results from this study show that HM781-36B is a potent inhibitor of EGFR in vitro, including the EGFR-acquired resistance mutation (T790M), as well as HER-2 and HER-4, compared with other EGFR tyrosine kinases inhibitors (erlotinib, lapatinib and BIBW2992). HM781-36B treatment of EGFR DelE746_A750-harboring erlotinib-sensitive HCC827 and EGFR L858R/T790M-harboring erlotinib-resistant NCI-H1975 NSCLC cells results in the inhibition of EGFR phosphorylation and the subsequent deactivation of downstream signaling proteins. Additionally, HM781-36B shows an excellent efficacy in a variety of EGFR- and HER-2-dependent tumor xenograft models, including erlotinib-sensitive HCC827 NSCLC cells, erlotinib-resistant NCI-H1975 NSCLC cells, HER-2 overexpressing Calu-3 NSCLC cells, NCI-N87 gastric cancer cells, SK-Ov3 ovarian cancer cells and EGFR-overexpressing A431 epidermoid carcinoma cancer cells. On the basis of these preclinical results, HM781-36B is the most potent pan-HER inhibitor, which will be advantageous for the treatment of patients with NSCLC including clinical limitation caused by acquired mutation (EGFR T790M), breast cancer and gastric cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Quinazolinas/uso terapéutico , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Clorhidrato de Erlotinib , Humanos , Ratones , Ratones Desnudos , Fragmentos de Péptidos/antagonistas & inhibidores , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor ErbB-2/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Anticancer Drugs ; 23(3): 288-97, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23422737

RESUMEN

HM781-36B is an orally administered pan-human epidermal growth factor receptor (HER) inhibitor. To explore the role of pan-HER inhibitor in breast cancer, we investigated the antitumor effect and mechanisms of HM781-36B in breast cancer cell lines. Six breast cancer cell lines (BT474, MDA-MB-453, SK-BR-3, T47D, MCF-7, and MDA-MB-231) were tested. The growth inhibitory effect was assessed using the tetrazolium bromide [3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl-tetrazolium bromide] assay. The cell cycle at various concentrations of HM781-36B was analyzed by flow cytometry, and analysis of downstream molecules was performed by western blot analysis. Interaction of HM781-36B with cytotoxic chemotherapeutic agents was analyzed by combination index using CalcuSyn. The HER2-amplified cells (SK-BR-3, BT474, and MDA-MB-453) were sensitive to HM781-36B (IC50=0.001 µmol/l, 0.0012 µmol/l, and 0.0095 µmol/l, respectively). HM781-36B induced G1 arrest and resulted in apoptosis. It reduced the level of p-HER2, p-AKT, p-ERK, and p-STAT3. HM781-36B combined with 5-fluorouracil, cisplatin, paclitaxel, or gemcitabine showed a synergistic inhibitory effect on the HER2-amplified and on some of the HER2-nonamplified breast cancer cells. HM781-36B could be a promising treatment for HER2-amplified breast cancer as a single agent or in combination with cytotoxic agents and can be a candidate for treatment of HER2-nonamplified breast cancer in combination with cytotoxic agents.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/administración & dosificación , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Relación Dosis-Respuesta a Droga , Femenino , Fluorouracilo/administración & dosificación , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Paclitaxel/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/administración & dosificación , Receptor ErbB-2/metabolismo , Factor de Transcripción STAT3/metabolismo , Gemcitabina
6.
Cancer Lett ; 302(2): 155-65, 2011 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-21306821

RESUMEN

Trastuzumab, a HER2 directed treatment has shown clinical benefit in HER2 amplified gastric cancer. This study demonstrated the potent antitumor activity of HM781-36B, a quinazoline-based irreversible pan-HER inhibitor, in HER2 amplified gastric cancer cells (SNU216 and N87) in vitro and in vivo. HM781-36B inhibited phosphorylation of HER family and downstream signaling molecules, and induced apoptosis and G1 arrest. Furthermore, HM781-36B exerted synergistic effects with chemotherapeutic agents in both HER2 amplified and HER2 non-amplified gastric cancer cells. Therefore, HM781-36B may be useful for the treatment of HER2 amplified gastric cancer alone or in combination with chemotherapeutic agents.


Asunto(s)
Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Concentración 50 Inhibidora , Ratones , Fosforilación/efectos de los fármacos , Quinazolinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico
7.
Eur J Med Chem ; 45(11): 5420-7, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20869793

RESUMEN

Virtual screening was performed to determine potent vascular endothelial growth factor receptor (VEGFR)-2 kinase inhibitors. A database of approximately 820,000 commercial compounds was used for screening, and 100 compounds were chosen as candidate VEGFR-2 inhibitors through pharmacophore modeling and docking studies. These 100 compounds were purchased to test their biological activities: 10 compounds were found to inhibit the enzyme, with IC(50) values ranging from 10 to 1 µM. Compound 1, which has a triazinoindole ring, inhibited the enzymatic activity of VEGFR-2, with an IC(50) value of about 1.6 µM, making it the most potent inhibitor of this enzyme. The triazinoindole derivative may therefore serve as the starting point in the design of new VEGFR-2 kinase inhibitors.


Asunto(s)
Modelos Moleculares , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos
8.
Eur J Pharmacol ; 627(1-3): 92-8, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-19903471

RESUMEN

Multi-drug resistance 1 (MDR1, ABCB1), also known as P-glycoprotein (P-gp), restricts intestinal uptake of many drugs, and contributes to cellular resistance to cancer chemotherapy. In this study, we examined the pharmacologic characteristics of HM30181, a newly developed MDR1 inhibitor, and tested its capacity to increase the oral bioavailability and efficacy of paclitaxel, an anti-cancer drug usually given by intravenous injection. In the ATPase assay using MDR1-enriched vesicles, HM30181 showed the highest potency (IC(50)=0.63nM) among several MDR1 inhibitors, including cycloporin A, XR9576, and GF120918, and effectively blocked transepithelial transport of paclitaxel in MDCK monolayers (IC(50)=35.4nM). The ATPase inhibitory activity of HM30181 was highly selective to MDR1. HM30181 did not inhibit MRP1 (ABCC1), MRP2 (ABCC2), and MRP3 (ABCC3), and partially inhibited BCRP (ABCG2) only at very high concentrations. Importantly, co-administration of HM30181 (10mg/kg) greatly increased oral bioavailability of paclitaxel from 3.4% to 41.3% in rats. Moreover, oral co-administration of paclitaxel and HM30181 showed a tumor-inhibitory strength equal or superior to that of intravenous paclitaxel in the xenograft model in nude mice. These results identify HM30181 as a highly selective and potent inhibitor of MDR1, which in combination with paclitaxel, may provide an orally effective anti-tumor regimen.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Benzopiranos/farmacología , Isoquinolinas/farmacología , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Tetrazoles/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Benzopiranos/administración & dosificación , Disponibilidad Biológica , Línea Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/terapia , Perros , Humanos , Isoquinolinas/administración & dosificación , Masculino , Ratones , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Paclitaxel/administración & dosificación , Ratas , Ratas Sprague-Dawley , Especificidad por Sustrato , Tetrazoles/administración & dosificación , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Med Chem ; 52(21): 6880-8, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19888761

RESUMEN

A novel series of (S)-1-acryloyl-N-[4-(arylamino)-7-(alkoxy)quinazolin-6-yl]pyrrolidine-2-carboxamides were synthesized and evaluated as Her-1/Her-2 dual inhibitors. In contrast to the Her-1 selective inhibitors, our novel compounds are irreversible inhibitors of Her-1 and Her-2 tyrosine kinases with the potential to overcome clinically relevant, mutation-induced drug resistance. The selected compounds (19c, 19d) showed excellent EGFR inhibition activity even toward the T790M mutation of Her-1 tyrosine kinase with excellent selectivity. The excellent pharmacokinetic profiles of these compounds in rats and their robust in vivo efficacy in an A431 xenograft model clearly demonstrate that they merit further investigation as novel therapeutic agents for EGFR-targeting treatment of solid tumors, especially Her-1 selective inhibitor-resistant non-small cell lung cancer.


Asunto(s)
Compuestos de Anilina/síntesis química , Antineoplásicos/síntesis química , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Pirrolidinas/síntesis química , Quinazolinas/síntesis química , Receptor ErbB-2/antagonistas & inhibidores , Compuestos de Anilina/farmacocinética , Compuestos de Anilina/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/genética , Clorhidrato de Erlotinib , Femenino , Gefitinib , Humanos , Neoplasias Pulmonares/enzimología , Masculino , Ratones , Ratones Desnudos , Conformación Molecular , Trasplante de Neoplasias , Pirrolidinas/farmacocinética , Pirrolidinas/farmacología , Quinazolinas/farmacocinética , Quinazolinas/farmacología , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Trasplante Heterólogo
10.
Bioorg Med Chem ; 17(8): 3152-61, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19321350

RESUMEN

Src is an important target in multiple processes associated with tumor growth and development, including proliferation, neovascularization, and metastasis. In this study, hit identification was performed by virtual screening of commercial and in-house compound libraries. Docking studies for the hits were performed, and scoring functions were used to evaluate the docking results and to rank ligand-binding affinities. Subsequently, hit optimization for potent and selective candidate Src inhibitors was performed through focused library design and docking analyses. Consequently, we report that a novel compound '43' with an IC(50) value of 89 nM, representing (S)-N-(4-(5-chlorobenzo[d][1,3]dioxol-4-ylamino)-7-(2-methoxyethoxy)quinazolin-6-yl)pyrrolidine-2-carboxamide, is highly selective for Src in comparison to EGFR (IC(50) ratio>80-fold) and VEGFR-2 (IC(50) ratio>110-fold). Compound 43 exerted anti-proliferative effects on Src-expressing PC3 human prostate cancer and A431 human epidermoid carcinoma cells, with calculated IC(50) values of 1.52 and 0.78 microM, respectively. Moreover, compound 43 (0.1 microM) suppressed the phosphorylation of extracellular signal-regulated kinases and p90 ribosomal S6 kinase, downstream molecules of Src, in a time-dependent manner, in both PC3 and A431 cell lines. The docking structure of compound 43 with Src disclosed that the chlorobenzodioxole moiety and pyrrolidine ring of C-6 quinazoline appeared to fit tightly into the hydrophobic pocket of Src. Additionally, the pyrrolidine NH forms a hydrogen bond with the carboxyl group of Asp348. These results confirm the successful application of virtual screening studies in the lead discovery process, and suggest that our novel compound 43 can be an effective Src inhibitor candidate for further lead optimization.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/química , Animales , Sitios de Unión , Línea Celular Tumoral , Técnicas Químicas Combinatorias/métodos , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ligandos , Masculino , Modelos Moleculares , Ratas , Ratas Sprague-Dawley , Programas Informáticos
11.
Int J Oncol ; 33(4): 705-12, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18813783

RESUMEN

P-glycoprotein (P-gp), a factor responsible for the multidrug resistance of tumors, is specifically expressed in brain microenvironment. To test its roles in brain metastatic tumor chemoresistance, we implanted the paclitaxel-sensitive melanoma cell line, K1735, into the skin or brain of mice and examined its paclitaxel resistances. When implanted into the skin, paclitaxel inhibited tumor growth, however, it had no inhibitory effect on cells implanted into the brain. The paclitaxel resistance of the brain K1735 tumors was eliminated by combined treatment with a P-gp inhibitor, HM30181A, and paclitaxel. Previously we found that there is a defined therapeutic window for combined treatment of brain tumors with HM30181A and paclitaxel. To determine whether it is due to responses of the brain microenvironment we measured changes in P-gp expression and function of brain endothelial cells in response to HM30181A treatment in vitro and in vivo. They were significantly increased by high-dose HM30181A treatment and it was related with the therapeutic effect loss of high-dose HM30181A treatment. Therefore, P-gp in the brain microenvironment has crucial roles in the brain metastatic tumor chemoresistance and brain microenvironment responses to P-gp inhibitor treatment should be considered in the development of brain endothelial cell-targeted chemotherapy using P-gp inhibitor.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/metabolismo , Regulación Neoplásica de la Expresión Génica , Animales , Antineoplásicos/farmacología , Benzopiranos/farmacología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Isoquinolinas/farmacología , Masculino , Ratones , Ratones Endogámicos C3H , Metástasis de la Neoplasia , Trasplante de Neoplasias , Paclitaxel/farmacología , Tetrazoles/farmacología
12.
Oncol Rep ; 19(1): 17-23, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18097571

RESUMEN

Oral chemotherapy has many advantages over parenteral chemotherapeutics administration. To use the advantages of the oral chemotherapy and maximize anti-tumor effects of the chemotherapeutic agent, we designed HM30181A (a P-glycoprotein inhibitor) and a paclitaxel oral co-administration chemotherapeutic method. HM30181A is used to aid paclitaxel absorption from gut lumen into blood and to inhibit paclitaxel exclusion out of the brain tumor mass by endothelial cells, which inhibits paclitaxel access to tumor cells in the brain parenchyma. We applied HM30181A and paclitaxel oral co-administration methods to the treatment of tumors in the brain using the K1735 melanoma brain metastasis animal model and the U-87 MG glioblastoma animal model. Administrations were performed twice per week for 28 days and the therapeutic effect was examined using tumor volume change. We observed that 32 mg/kg HM30181A and 16 mg/kg of paclitaxel (dose ratio 2:1) oral co-administration showed significant therapeutic effects in both animal models, but when the doses or dose ratio was changed, the effects could not be observed. Therefore, adjustments of doses and dose ratio of the agents seems to be essential in realizing oral HM30181A and paclitaxel treatment in brain tumors. These results suggest that if the doses and dose ratio can be successfully adjusted, the oral co-administration of HM30181A and paclitaxel can be used to treat tumors in the brain.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Paclitaxel/uso terapéutico , Administración Oral , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias
13.
Mol Med Rep ; 1(3): 343-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-21479415

RESUMEN

The objective of this study was to examine the antitumor effect of ZD6474, an orally available inhibitor of the vascular endothelial growth factor receptor-2 (VEGFR-2) and the epidermal growth factor receptor (EGFR), on tumor growth in an orthotopic metastatic brain tumor model. In order to determine the antitumor mechanism of ZD6474 treatment, in vitro and in vivo studies were performed. Human breast carcinoma cells (MDA-MB-435) were injected using direct intracranial (IC) inoculation (5x105 cells/100 µl) and internal carotid artery (ICA) injection (5x104 cells/100 µl) in Balb/c-nu female mice. Daily oral treatment with ZD6474 (50 mg/kg) was initiated on day 14 after the establishment of micrometastasis. Mice (n=12 per group) were sacrificed on day 28. Western blot analysis revealed that the autophosphorylation of EGFR and Akt was increasingly decreased with ZD6474 treatment in lung and brain endothelial cells and the MDA-MB-435 cell line. MTT assay also showed that the in vitro antitumor activity of ZD6474 was dependent on EGFR tyrosine kinase inhibition at a higher dose. Daily oral treatment with ZD6474 led to marked inhibition of metastatic tumor growth in the ICA injection and the direct IC inoculation models (median size 3.5 mm3, range 1.6-13.9 mm3) as compared to the control group (median size 62.4 mm3, range 11.5-206.9 mm3). These results suggest that simultaneous inhibition of both the EGFR and VEGFR-2 signaling pathways has a valuable therapeutic effect through its inhibition of the growth of metastatic brain tumors.

14.
J Toxicol Environ Health A ; 70(15-16): 1356-64, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17654255

RESUMEN

HM-30181, 4-oxo-4H-chromene-2-carboxylic acid [2-(2-{4-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl]-phenyl}-2H-tetrazol-5-yl)-4,5-dimethoxy-phenyl]-amide, is a new P-glycoprotein inhibitor with the potential to increase the cytotoxic activity of orally coadministered paclitaxel. This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. CYP3A4 was identified as the major isozyme responsible for the O-demethylation of HM-30181 to M2 and M3 based on the correlation analysis, chemical inhibition and immuno-inhibition study and metabolism in cDNA-expressed human CYP isozymes. HM-30181 itself had no inhibitory effects on CYPs 1A2, 2A6, 2C8, 2C9, 2C19, 2D6, and 3A4 in human liver microsomes, suggesting the possibility that the pharmacokinetics of HM-30181 could be changed with coadministration of known CYP3A4 inducers or inhibitors.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Benzopiranos/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Isoquinolinas/metabolismo , Oxidorreductasas O-Demetilantes , Tetrazoles/metabolismo , Benzopiranos/farmacocinética , Citocromo P-450 CYP3A , ADN Complementario/metabolismo , Humanos , Isoquinolinas/farmacocinética , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Tetrazoles/farmacocinética
15.
Rapid Commun Mass Spectrom ; 20(9): 1457-62, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16586459

RESUMEN

HM-30181, 4-oxo-4H-chromene-2-carboxylic acid, [2-(2-{4-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl]-phenyl}-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl]amide, is a new P-glycoprotein inhibitor. This study was performed to identify the in vitro and in vivo metabolic pathway of HM-30181 in rats. Rat liver microsomal incubation of HM-30181 in the presence of NADPH resulted in the formation of four metabolites, M1-M4. M1 and M2 were identified as 2-(2-{4-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl]-phenyl}-2H-tetrazol-5-yl)-4,5-dimethoxyaniline and 4- or 5-O-desmethyl-HM-30181, respectively, on the basis of liquid chromatography/tandem mass spectrometry (LC/MS/MS) analysis with the synthesized authentic standards. M3 and M4 were suggested to be 6- or 7-O-desmethyl-HM-30181 and hydroxy-HM-30181, respectively. These in vitro metabolites were also detected in feces and urine samples after an intravenous administration of HM-30181 to male rats. The metabolic routes for HM-30181 were O-demethylation of the methoxy group to M2 and M3, hydrolysis of the amide group to M1, and hydroxylation to M4.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Benzopiranos/farmacología , Isoquinolinas/farmacología , Tetrazoles/farmacología , Animales , Benzopiranos/análisis , Biotransformación , Cromatografía Líquida de Alta Presión , Remoción de Radical Alquila , Heces/química , Técnicas In Vitro , Indicadores y Reactivos , Inyecciones Intravenosas , Isoquinolinas/análisis , Microsomas Hepáticos/metabolismo , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray , Tetrazoles/análisis
16.
J Vet Med Sci ; 66(11): 1339-45, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15585946

RESUMEN

Toxicogenomics, the subdiscipline that merges genomics with toxicology, hold the promise to contributing toward the goal of elucidating mechanism by studying genomic profiling related with various drugs. The application of gene expression profiling technology to examine multiple genes and signaling pathways promises a significant advance in understanding the toxic mechanisms of various drugs and prediction of new drug candidate. Toxicogenomics is emerging field combining genomics and bioinformatics to identify and characterize mechanisms of toxicity of drug and various compounds. The principal hypothesis underlying on this field is that chemical-specific pattern of altered gene expression is related with each chemicals properties, especially toxicological property, and it will be revealed using high-density microarray analysis of sample from exposed organisms. So, in this study we compare the gene expression pattern of two anticancer drugs paclitaxel and orally absorbable paclitaxel, using the cDNA microarray. And from the result of this study, it is possible to provide the new possibility for genome-wide insight into mechanism of their anticancer activity and toxicological phenotype.


Asunto(s)
Antineoplásicos/toxicidad , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Paclitaxel/toxicidad , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Femenino , Inyecciones Intraperitoneales , Masculino , Paclitaxel/administración & dosificación , Fenotipo , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Toxicogenética
17.
Circulation ; 109(12): 1558-63, 2004 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-15006998

RESUMEN

BACKGROUND: Paclitaxel has been shown to inhibit vascular smooth muscle cell migration and proliferation contributing to neointimal formation. This study tested whether novel oral formulations of paclitaxel can prevent neointimal formation in a rat carotid artery injury model. METHODS AND RESULTS: Oral formulations of paclitaxel (0, 5, 7.5, or 10 mg/kg) were administered to 40 rats by gavage for 5 days after injury. The peak plasma levels of paclitaxel administered at 5, 7.5, and 10 mg/kg were 61+/-16, 89+/-22, and 108+/-28 nmol/L, respectively. Treatment effects were assessed 11 days after injury. The angiographic minimum luminal diameters of the oral paclitaxel groups treated at 5, 7.5, and 10 mg/kg were 6.28+/-2.09, 6.97+/-1.79, and 7.97+/-1.57 AU, and these were significantly larger than that of the control group (4.67+/-1.45 AU). The oral paclitaxel groups (5, 7.5, 10 mg/kg; 0.05+/-0.05, 0.04+/-0.03, 0.05+/-0.03 mm2) showed significant neointimal formation reductions versus the control group (0.13+/-0.05 mm2). All rats survived to study completion. Only 2 animals in the 10 mg/kg group experienced weight loss ( approximately 10%) and loose stools between 4 and 6 days after injury. All other animals appeared healthy during the study. For comparison purposes, intraperitoneal formulations of paclitaxel (0 or 2 mg/kg) were administered by injection to 15 rats. We confirmed that the intraperitoneal administration of paclitaxel also effectively inhibited neointimal formation. CONCLUSIONS: Oral formulations of paclitaxel provide an effective means of inhibiting proliferative response to vascular injury in the rat. Thus, oral formulations of paclitaxel may prevent human restenosis without significant toxicity.


Asunto(s)
Traumatismos de las Arterias Carótidas/patología , Estenosis Carotídea/prevención & control , Glicerol/análogos & derivados , Paclitaxel/administración & dosificación , Túnica Íntima/efectos de los fármacos , Verapamilo/análogos & derivados , alfa-Tocoferol/análogos & derivados , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Administración Oral , Animales , Disponibilidad Biológica , Traumatismos de las Arterias Carótidas/diagnóstico por imagen , Traumatismos de las Arterias Carótidas/etiología , Estenosis Carotídea/etiología , Estenosis Carotídea/patología , Cateterismo/efectos adversos , División Celular , Glicoles de Etileno/administración & dosificación , Glicerol/administración & dosificación , Hiperplasia , Infusiones Parenterales , Intestinos/efectos de los fármacos , Ácidos Linoleicos/administración & dosificación , Masculino , Modelos Animales , Miocitos del Músculo Liso/efectos de los fármacos , Paclitaxel/sangre , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Vehículos Farmacéuticos , Polisorbatos/administración & dosificación , Radiografía , Ratas , Ratas Sprague-Dawley , Recurrencia , Tensoactivos/administración & dosificación , Tocoferoles , Túnica Íntima/patología , Verapamilo/farmacología , alfa-Tocoferol/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA