Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 6: 191, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26441648

RESUMEN

Incorporating phenotypic screening as a key strategy enhances predictivity and translatability of drug discovery efforts. Cellular imaging serves as a "phenotypic anchor" to identify important toxicologic pathology that encompasses an array of underlying mechanisms, thus provides an effective means to reduce drug development failures due to insufficient safety. This mini-review highlights the latest advances in hepatotoxicity, cardiotoxicity, and genetic toxicity tests that utilized cellular imaging as a screening strategy, and recommends path forward for further improvement.

2.
MAbs ; 4(6): 710-23, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23007574

RESUMEN

The c-Met proto-oncogene is a multifunctional receptor tyrosine kinase that is stimulated by its ligand, hepatocyte growth factor (HGF), to induce cell growth, motility and morphogenesis. Dysregulation of c-Met function, through mutational activation or overexpression, has been observed in many types of cancer and is thought to contribute to tumor growth and metastasis by affecting mitogenesis, invasion, and angiogenesis. We identified human monoclonal antibodies that bind to the extracellular domain of c-Met and inhibit tumor growth by interfering with ligand-dependent c-Met activation. We identified antibodies representing four independent epitope classes that inhibited both ligand binding and ligand-dependent activation of c-Met in A549 cells. In cells, the antibodies antagonized c-Met function by blocking receptor activation and by subsequently inducing downregulation of the receptor, translating to phenotypic effects in soft agar growth and tubular morphogenesis assays. Further characterization of the antibodies in vivo revealed significant inhibition of c-Met activity (≥ 80% lasting for 72-96 h) in excised tumors corresponded to tumor growth inhibition in multiple xenograft tumor models. Several of the antibodies identified inhibited the growth of tumors engineered to overexpress human HGF and human c-Met (S114 NIH 3T3) when grown subcutaneously in athymic mice. Furthermore, lead candidate antibody CE-355621 inhibited the growth of U87MG human glioblastoma and GTL-16 gastric xenografts by up to 98%. The findings support published pre-clinical and clinical data indicating that targeting c-Met with human monoclonal antibodies is a promising therapeutic approach for the treatment of cancer.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Proteínas Proto-Oncogénicas c-met/inmunología , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/inmunología , Procesos de Crecimiento Celular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/inmunología , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Epítopos Inmunodominantes/inmunología , Ratones , Ratones Desnudos , Morfogénesis/efectos de los fármacos , Células 3T3 NIH , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/genética , Transgenes/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Methods Mol Biol ; 795: 83-107, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21960217

RESUMEN

Kinases are members of a major protein family targeted for drug discovery and development. Given the ubiquitous nature of many kinases as well as the broad range of pathways controlled by these enzymes, early safety assessments of small molecule inhibitors of kinases are crucial in identifying new molecules with sufficient therapeutic window for clinical development. Failure or attrition of drug candidates in late-stage pipelines due to hepatotoxicity is a significant challenge in the drug development field. Herein we provide detailed methods for the hepatocyte imaging assay technology (HIAT) and the bile flux imaging assay technology (BIAT) to evaluate drug-induced liver injury (DILI) potentials for drug candidates. Optimized culturing methods for primary human hepatocytes, both freshly isolated and prequalified cryopreserved cells, are also presented. The applications of these high-content cellular imaging technologies in the evaluation of p38 and Her2 kinase inhibitors are highlighted to illustrate the usefulness of the research methodology in a compound screening as well as mechanistic investigative setting.


Asunto(s)
Bilis/metabolismo , Inhibidores Enzimáticos/toxicidad , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Imagen Molecular/métodos , Fosfotransferasas/antagonistas & inhibidores , Pruebas de Toxicidad/métodos , Bioensayo/métodos , Células Cultivadas , Colágeno , Criopreservación , Combinación de Medicamentos , Humanos , Procesamiento de Imagen Asistido por Computador , Laminina , Microscopía Fluorescente , Proteoglicanos , Coloración y Etiquetado
4.
Drug Metab Dispos ; 38(12): 2302-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20843939

RESUMEN

Drug-induced liver injury (DILI) is one of the most important reasons for drug development failure at both preapproval and postapproval stages. There has been increased interest in developing predictive in vivo, in vitro, and in silico models to identify compounds that cause idiosyncratic hepatotoxicity. In the current study, we applied machine learning, a Bayesian modeling method with extended connectivity fingerprints and other interpretable descriptors. The model that was developed and internally validated (using a training set of 295 compounds) was then applied to a large test set relative to the training set (237 compounds) for external validation. The resulting concordance of 60%, sensitivity of 56%, and specificity of 67% were comparable to results for internal validation. The Bayesian model with extended connectivity functional class fingerprints of maximum diameter 6 (ECFC_6) and interpretable descriptors suggested several substructures that are chemically reactive and may also be important for DILI-causing compounds, e.g., ketones, diols, and α-methyl styrene type structures. Using Smiles Arbitrary Target Specification (SMARTS) filters published by several pharmaceutical companies, we evaluated whether such reactive substructures could be readily detected by any of the published filters. It was apparent that the most stringent filters used in this study, such as the Abbott alerts, which captures thiol traps and other compounds, may be of use in identifying DILI-causing compounds (sensitivity 67%). A significant outcome of the present study is that we provide predictions for many compounds that cause DILI by using the knowledge we have available from previous studies. These computational models may represent cost-effective selection criteria before in vitro or in vivo experimental studies.


Asunto(s)
Teorema de Bayes , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Humanos , Ligandos
6.
Chem Biol Interact ; 182(1): 45-51, 2009 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-19560444

RESUMEN

The existence of a porphyrin uptake transporter in hepatocytes has been hypothesized in recent years, but to date it has not been identified. While the linear tetrapyrrole bilirubin has been shown to be a substrate for the organic anion transporting polypeptide 1B1 (OATP1B1), similar studies have not been conducted for the cyclic tetrapyrroles (porphyrins). The aim of this study was to determine the structural features of linear and cyclic tetrapyroles necessary for interaction with OATP1B1. The interaction was quantified using HEK cells stably expressing OATP1B1 and measuring the inhibition of OATP1B1-mediated uptake of estradiol 17beta-d-glucuronide in the presence or absence of various linear and cyclic tetrapyrroles. Ditaurine-conjugated bilirubin was the most potent inhibitor of uptake, with an IC50 of 5 nM, while the substitution of the taurine side chains with methyl ester eliminated the inhibition of estradiol 17beta-d-glucuronide uptake. Hematoporphyrin, a cyclic tetrapyrrole with carboxyalcohol side chains at positions C-3 and C-8 and carboxyethyl side chains at positions 13 and 17 had an IC50 of 60 nM, while porphyrins lacking charged side chains such as etioporphyrin I and phthalocyanine did not inhibit OATP1B1. Chlorin e6 and hematoporphyrin were shown to be competitive inhibitors of OATP1B1-mediated uptake of bromosulfophthalein with Kis of 5.8+/-0.3 and 1.6+/-0.3 microM, respectively. While these studies do not provide direct evidence, they do support the assumption that tetrapyrroles are transported by OATP1B1. Additionally, these findings offer a possible explanation for the clinical observation that patients suffering from certain porphyrietic diseases have a reduced ability to excrete organic anions.


Asunto(s)
Estradiol/análogos & derivados , Transportadores de Anión Orgánico/metabolismo , Porfirinas/farmacocinética , Sitios de Unión , Unión Competitiva , Transporte Biológico , Línea Celular , Estradiol/metabolismo , Estradiol/farmacocinética , Humanos , Concentración 50 Inhibidora , Transportador 1 de Anión Orgánico Específico del Hígado , Modelos Moleculares , Transportadores de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico/biosíntesis , Transportadores de Anión Orgánico/genética , Porfirinas/metabolismo , Sulfobromoftaleína/metabolismo , Transfección
7.
Toxicol Appl Pharmacol ; 237(3): 317-30, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19362101

RESUMEN

Idiosyncratic drug hepatotoxicity represents a major problem in drug development due to inadequacy of current preclinical screening assays, but recently established rodent models utilizing bacterial LPS co-administration to induce an inflammatory background have successfully reproduced idiosyncratic hepatotoxicity signatures for certain drugs. However, the low-throughput nature of these models renders them problematic for employment as preclinical screening assays. Here, we present an analogous, but high-throughput, in vitro approach in which drugs are administered to a variety of cell types (primary human and rat hepatocytes and the human HepG2 cell line) across a landscape of inflammatory contexts containing LPS and cytokines TNF, IFN gamma, IL-1 alpha, and IL-6. Using this assay, we observed drug-cytokine hepatotoxicity synergies for multiple idiosyncratic hepatotoxicants (ranitidine, trovafloxacin, nefazodone, nimesulide, clarithromycin, and telithromycin) but not for their corresponding non-toxic control compounds (famotidine, levofloxacin, buspirone, and aspirin). A larger compendium of drug-cytokine mix hepatotoxicity data demonstrated that hepatotoxicity synergies were largely potentiated by TNF, IL-1 alpha, and LPS within the context of multi-cytokine mixes. Then, we screened 90 drugs for cytokine synergy in human hepatocytes and found that a significantly larger fraction of the idiosyncratic hepatotoxicants (19%) synergized with a single cytokine mix than did the non-hepatotoxic drugs (3%). Finally, we used an information theoretic approach to ascertain especially informative subsets of cytokine treatments for most highly effective construction of regression models for drug- and cytokine mix-induced hepatotoxicities across these cell systems. Our results suggest that this drug-cytokine co-treatment approach could provide a useful preclinical tool for investigating inflammation-associated idiosyncratic drug hepatotoxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citocinas/toxicidad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Mediadores de Inflamación/toxicidad , Animales , Línea Celular Tumoral , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Citocinas/clasificación , Citocinas/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/métodos , Sinergismo Farmacológico , Hepatocitos/metabolismo , Humanos , Mediadores de Inflamación/clasificación , Mediadores de Inflamación/farmacocinética , Masculino , Preparaciones Farmacéuticas/clasificación , Preparaciones Farmacéuticas/metabolismo , Ratas , Ratas Endogámicas F344
8.
Toxicol Sci ; 108(2): 492-500, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19223659

RESUMEN

CP-724,714, a potent and selective orally active HER2 tyrosine kinase inhibitor, was discontinued from clinical development due to unexpected hepatotoxicity in cancer patients. Based on the clinical manifestation of the toxicity, CP-724,714 likely exerted its hepatotoxicity via both hepatocellular injury and hepatobiliary cholestatic mechanisms. The direct cytotoxic effect, hepatobiliary disposition of CP-724,714, and its inhibition of active canalicular transport of bile constituents were evaluated in established human hepatocyte models and in vitro transporter systems. CP-724,714 exhibited direct cytotoxicity using human hepatocyte imaging assay technology with mitochondria identified as a candidate organelle for its off-target toxicity. Additionally, CP-724,714 was rapidly taken up into human hepatocytes, partially via an active transport process, with an uptake clearance approximately fourfold higher than efflux clearance. The major human hepatic uptake transporter, OATP1B1, and efflux transporters, multidrug resistance protein 1 (MDR1) and breast cancer resistance protein, were involved in hepatobiliary clearance of CP-724,714. Furthermore, CP-724,714 displayed a concentration-dependent inhibition of cholyl-lysyl fluorescein and taurocholate (TC) efflux into canaliculi in cryopreserved and fresh cultured human hepatocytes, respectively. Likewise, CP-724,714 inhibited TC transport in membrane vesicles expressing human bile salt export pump with an IC(50) of 16 microM. Finally, CP-724,714 inhibited the major efflux transporter in bile canaliculi, MDR1, with an IC(50) of approximately 28 microM. These results suggest that inhibition of hepatic efflux transporters contributed to hepatic accumulation of drug and bile constituents leading to hepatocellular injury and hepatobiliary cholestasis. This study provides likely explanations for clinically observed adverse liver effects of CP-724,714.


Asunto(s)
Proteínas Portadoras/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/toxicidad , Hígado/metabolismo , Quinazolinas/farmacocinética , Quinazolinas/toxicidad , Receptor ErbB-2/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP , 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 , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Bilis/metabolismo , Ácidos y Sales Biliares/metabolismo , Calcio/metabolismo , Células Cultivadas , Fluoresceínas/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado/antagonistas & inhibidores , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo
9.
Toxicol Sci ; 105(1): 97-105, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18524759

RESUMEN

Drug-induced liver injury (DILI) is the most common adverse event causing drug nonapprovals and drug withdrawals. Using drugs as test agents and measuring a panel of cellular phenotypes that are directly linked to key mechanisms of hepatotoxicity, we have developed an in vitro testing strategy that is predictive of many clinical outcomes of DILI. Mitochondrial damage, oxidative stress, and intracellular glutathione, all measured by high content cellular imaging in primary human hepatocyte cultures, are the three most important features contributing to the hepatotoxicity prediction. When applied to over 300 drugs and chemicals including many that caused rare and idiosyncratic liver toxicity in humans, our testing strategy has a true-positive rate of 50-60% and an exceptionally low false-positive rate of 0-5%. These in vitro predictions can augment the performance of the combined traditional preclinical animal tests by identifying idiosyncratic human hepatotoxicants such as nimesulide, telithromycin, nefazodone, troglitazone, tetracycline, sulindac, zileuton, labetalol, diclofenac, chlorzoxazone, dantrolene, and many others. Our findings provide insight to key DILI mechanisms, and suggest a new approach in hepatotoxicity testing of pharmaceuticals.


Asunto(s)
Hepatocitos/efectos de los fármacos , Células Cultivadas , Bases de Datos como Asunto , Reacciones Falso Positivas , Hepatocitos/patología , Humanos , Mitocondrias Hepáticas/fisiología , Preparaciones Farmacéuticas/clasificación , Farmacocinética , Curva ROC
10.
Toxicol Sci ; 103(2): 335-45, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18344530

RESUMEN

Mitochondrial toxicity is increasingly implicated in a host of drug-induced organ toxicities, including hepatotoxicity. Nefazodone was withdrawn from the U.S. market in 2004 due to hepatotoxicity. Accordingly, we evaluated nefazodone, another triazolopyridine trazodone, plus the azaspirodecanedione buspirone, for cytotoxicity and effects on mitochondrial function. In accord with its clinical disposition, nefazodone was the most toxic compound of the three, trazodone had relatively modest effects, whereas buspirone showed the least toxicity. Nefazodone profoundly inhibited mitochondrial respiration in isolated rat liver mitochondria and in intact HepG2 cells where this was accompanied by simultaneous acceleration of glycolysis. Using immunocaptured oxidative phosphorylation (OXPHOS) complexes, we identified Complex 1, and to a lesser amount Complex IV, as the targets of nefazodone toxicity. No inhibition was found for trazodone, and buspirone showed 3.4-fold less inhibition of OXPHOS Complex 1 than nefazodone. In human hepatocytes that express cytochrome P450, isoform 3A4, after 24 h exposure, nefazodone and trazodone collapsed mitochondrial membrane potential, and imposed oxidative stress, as detected via glutathione depletion, leading to cell death. Our results suggest that the mitochondrial impairment imposed by nefazodone is profound and likely contributes to its hepatotoxicity, especially in patients cotreated with other drugs with mitochondrial liabilities.


Asunto(s)
Ansiolíticos/toxicidad , Antidepresivos de Segunda Generación/toxicidad , Buspirona/toxicidad , Hepatocitos/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Trazodona/toxicidad , Triazoles/toxicidad , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Respiración de la Célula/efectos de los fármacos , Respiración de la Célula/fisiología , Supervivencia Celular/efectos de los fármacos , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/metabolismo , Hepatocitos/enzimología , Hepatocitos/patología , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Piperazinas , Ratas , Ratas Sprague-Dawley
11.
FEBS Lett ; 582(8): 1276-82, 2008 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-18282474

RESUMEN

The majority of drug-related toxicities are idiosyncratic, with little pathophysiological insight and mechanistic understanding. Pathway toxicology is an emerging field of toxicology in the post-genomic era that studies the molecular interactions between toxicants and biological pathways as a way to bridge this knowledge gap. Using two case studies--acetaminophen and p38 MAPK inhibitors--this review illustrates how a pathway-based perspective has advanced our understanding of compound and target-based toxicities. The advancement of pathway toxicology will be dependent on integrated applications of techniques from basic sciences and a fundamental understanding of the interdependence of multiple biological pathways in living organisms.


Asunto(s)
Acetaminofén/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Acetaminofén/toxicidad , Animales , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Inhibidores de Proteínas Quinasas/toxicidad , Piel/enzimología
12.
Drug Metab Dispos ; 33(4): 537-46, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15616150

RESUMEN

The present study examined the interaction of four 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (atorvastatin, lovastatin, and simvastatin in acid and lactone forms, and pravastatin in acid form only) with multidrug resistance gene 1 (MDR1, ABCB1) P-glycoprotein, multidrug resistance-associated protein 2 (MRP2, ABCC2), and organic anion-transporting polypeptide 1B1 (OATP1B1, SLCO21A6). P-glycoprotein substrate assays were performed using Madin-Darby canine kidney (MDCK) cells expressing MDR1, and the efflux ratios [the ratio of the ratio of basolateral-to-apical apparent permeability and apical-to-basolateral permeability between MDR1 and MDCK] were 1.87, 2.32/4.46, 2.17/3.17, and 0.93/2.00 for pravastatin, atorvastatin (lactone/acid), lovastatin (lactone/acid), and simvastatin (lactone/acid), respectively, indicating that these compounds are weak or moderate substrates of P-glycoprotein. In the inhibition assays (MDR1, MRP2, Mrp2, and OATP1B1), the IC50 values for efflux transporters (MDR1, MRP2, and Mrp2) were >100 microM for all statins in acid form except lovastatin acid (>33 microM), and the IC50 values were up to 10-fold lower for the corresponding lactone forms. In contrast, the IC50 values for the uptake transporter OATP1B1 were 3- to 7-fold lower for statins in the acid form compared with the corresponding lactone form. These data demonstrate that lactone and acid forms of statins exhibit differential substrate and inhibitor activities toward efflux and uptake transporters. The interconversion between the lactone and acid forms of most statins exists in the body and will potentially influence drug-transporter interactions, and may ultimately contribute to the differences in pharmacokinetic profiles observed between statins.


Asunto(s)
Estradiol/análogos & derivados , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Proteínas de Transporte de Membrana/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Atorvastatina , Línea Celular , Perros , Estradiol/metabolismo , Fluoresceínas/metabolismo , Ácidos Heptanoicos/química , Ácidos Heptanoicos/farmacocinética , Hidrólisis , Inhibidores de Hidroximetilglutaril-CoA Reductasas/química , Lovastatina/química , Lovastatina/farmacocinética , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Permeabilidad , Pirroles/química , Pirroles/farmacocinética , Simvastatina/química , Simvastatina/farmacocinética , Relación Estructura-Actividad
13.
Chem Biol Interact ; 150(2): 179-87, 2004 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-15535988

RESUMEN

OATP1B1 (a.k.a. OATP-C, OATP2, LST-1, or SLC21A6) is a liver-specific organic anion uptake transporter and has been shown to be a higher affinity bilirubin uptake transporter than OATP1B3. Using human embryonic kidney (HEK 293) cells stably transfected with OATP1B1, we have studied the effects of indinavir, saquinavir, cyclosporin A, and rifamycin SV on human OATP1B1 transport function. These drugs are potent inhibitors of OATP1B1 transport activity in vitro. We further provide evidence that the calculated fraction of OATP1B1 inhibited at the clinical exposure level correlated very well with the observed hyperbilirubinemia outcome for these drugs in humans. Our data support the hypothesis that inhibition of OATP1B1 is an important mechanism for drug-induced unconjugated hyperbilirubinemia. Inhibition of OATPs may be an important mechanism in drug-drug and drug-endogenous substance interactions.


Asunto(s)
Hiperbilirrubinemia/enzimología , Transportador 1 de Anión Orgánico Específico del Hígado/antagonistas & inhibidores , Hígado/metabolismo , Bilirrubina/metabolismo , Línea Celular , Humanos , Hiperbilirrubinemia/inducido químicamente , Riñón , Cinética
14.
Chem Biol Interact ; 150(1): 115-28, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15522265

RESUMEN

While drug toxicity (especially hepatotoxicity) is the most frequent reason cited for withdrawal of an approved drug, no simple solution exists to adequately predict such adverse events. Simple cytotoxicity assays in HepG2 cells are relatively insensitive to human hepatotoxic drugs in a retrospective analysis of marketed pharmaceuticals. In comparison, a panel of pre-lethal mechanistic cellular assays hold the promise to deliver a more sensitive approach to detect endpoint-specific drug toxicities. The panel of assays covered by this review includes steatosis, cholestasis, phospholipidosis, reactive intermediates, mitochondria membrane function, oxidative stress, and drug interactions. In addition, the use of metabolically competent cells or the introduction of major human hepatocytes in these in vitro studies allow a more complete picture of potential drug side effect. Since inter-individual therapeutic index (TI) may differ from patient to patient, the rational use of one or more of these cellular assay and targeted in vivo exposure data may allow pharmaceutical scientists to select drug candidates with a higher TI potential in the drug discovery phase.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Hígado/patología , Toxicología/métodos , Supervivencia Celular , Interacciones Farmacológicas , Humanos , Hígado/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/patología
15.
Curr Top Med Chem ; 3(10): 1125-54, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12769713

RESUMEN

Major reasons preventing many early candidates reaching market are the inappropriate ADME (absorption, distribution, metabolism and excretion) properties and drug-induced toxicity. From a commercial perspective, it is desirable that poorly behaved compounds are removed early in the discovery phase rather than during the more costly drug development phases. As a consequence, over the past decade, ADME and toxicity (ADMET) screening studies have been incorporated earlier in the drug discovery phase. The intent of this review is to introduce the desirable attributes of a new chemical entity (NCE) to the medicinal chemist from an ADMET perspective. Fundamental concepts, key tools, reagents and experimental approaches used by the drug metabolism scientist to aid a modern project team in predicting human pharmacokinetics and assessing the "drug-like" molecule are discussed.


Asunto(s)
Absorción , Diseño de Fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Farmacocinética , Distribución Tisular , Animales , Disponibilidad Biológica , Transporte Biológico , Ensayos Clínicos como Asunto , Simulación por Computador , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Humanos , Modelos Biológicos , Relación Estructura-Actividad Cuantitativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...