Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Toxicol Sci ; 170(2): 345-356, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31020317

RESUMEN

The goal of this research consortium including Janssen, MSD, Ncardia, FNCR/LBR, and Health and Environmental Sciences Institute (HESI) was to evaluate the utility of an additional in vitro assay technology to detect potential drug-induced long QT and torsade de pointes (TdP) risk by monitoring cytosolic free Ca2+ transients in human stem-cell-derived cardiomyocytes (hSC-CMs). The potential proarrhythmic risks of the 28 comprehensive in vitro proarrhythmia assay (CiPA) drugs linked to low, intermediate, and high clinical TdP risk were evaluated in a blinded manner using Ca2+-sensitive fluorescent dye assay recorded from a kinetic plate reader system (Hamamatsu FDSS/µCell and FDSS7000) in 2D cultures of 2 commercially available hSC-CM lines (Cor.4U and CDI iCell Cardiomyocytes) at 3 different test sites. The Ca2+ transient assay, performed at the 3 sites using the 2 different hSC-CMs lines, correctly detected potential drug-induced QT prolongation among the 28 CiPA drugs and detected cellular arrhythmias-like/early afterdepolarization in 7 of 8 high TdP-risk drugs (87.5%), 6 of 11 intermediate TdP-risk drugs (54.5%), and 0 of 9 low/no TdP-risk drugs (0%). The results were comparable among the 3 sites and from 2 hSC-CM cell lines. The Ca2+ transient assay can serve as a user-friendly and higher throughput alternative to complement the microelectrode array and voltage-sensing optical action potential recording assays used in the HESI-CiPA study for in vitro assessment of drug-induced long QT and TdP risk.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Calcio/metabolismo , Síndrome de QT Prolongado/inducido químicamente , Miocitos Cardíacos/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Miocitos Cardíacos/metabolismo , Riesgo , Células Madre/citología
2.
Toxicol Appl Pharmacol ; 360: 88-98, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30268579

RESUMEN

Targeting the anti-apoptotic protein Mcl-1 holds a promise to improve therapy of multiple types of Mcl-1 dependent cancers but raises concerns of on-target cardiotoxicity due to the presence and reported role of Mcl-1 in heart. Herein, we investigated the importance of Mcl-1 in the survival and contractile function of human pluripotent stem cell-derived cardiomyocytes in culture. Effective knockdown of Mcl-1 with siRNAs reproducibly resulted in early (measured at Day 3) marginal alterations in caspase 3/7 activity, LDH leakage, ATP content and cellular impedance. After 14 days of Mcl-1 knockdown, loss of mitochondrial membrane potential, deteriorating effects on mitochondrial ultrastructure, and alterations in beat rate and amplitude were revealed. Inhibition of Bcl-xL by siRNA-mediated knockdown or selective inhibitors did not cause any overt cellular responses except for a minimal increase in caspase 3/7 activity; however, loss of Mcl-1 concomitant with down-regulated Bcl-xL activated apoptosis and caused extensive cell death as reflected by an 80% loss in cell index, activation of caspase-3 with associated PARP cleavage, and a decrease in beat amplitude and mitochondrial membrane potential after 3 days of Mcl-1/Bcl-xL knockdown., Together, these findings suggest that Mcl-1 and Bcl-xL provide duplicate safeguard measures in maintaining structural and functional integrity of cardiomyocytes. Hence, BH3-mimetic drugs targeting Mcl-1 may be well tolerated in the presence of intact Bcl-xL.


Asunto(s)
Muerte Celular/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Miocitos Cardíacos/metabolismo , Adenosina Trifosfato/metabolismo , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína bcl-X/metabolismo
3.
J Pharmacol Toxicol Methods ; 81: 201-16, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27282640

RESUMEN

INTRODUCTION: The ICH S7B guidelines recommend that all new chemical entities should be subjected to hERG repolarization screening due to its association with life-threatening "Torsades de Pointes" (TdP) arrhythmia. However, it has become evident that not all hERG channel inhibitors result in TdP and not all compounds that induce QT prolongation and TdP necessarily inhibit hERG. In order to address the limitations of the S7B/E14 guidelines, the FDA through a public/private partnership initiated the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative to examine the possible modification and refinement of the ICH E14/S7B guidelines. One of the main components of the CiPA initiative is to utilize a predictive assay system together with human cardiomyocytes for risk assessment of arrhythmia. METHOD: In this manuscript we utilize the xCELLigence® CardioECR system which simultaneously measures excitation-contraction coupling together with human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) to assess the effect of 8 reference compounds across 3 different independent sites. These 8 compounds were part of Phase I CiPA validation study. RESULTS: Our data demonstrate that hERG channel blockers, such as E4031 and moxifloxacin, prolonged field potential duration (FPD) at low concentration and induced arrhythmic beating activity as measured by field potential (FP) recording and impedance (IMP) recordings at higher concentrations. On the contrary, nifedipine, an inhibitor of calcium channel, didn't disrupt the periodicity of cell beating and weakened cell contractile activity and shortened FPD. Multichannel inhibitors, such as flecainide, quinidine and mexiletine, not only increased FPD and induced arrhythmia but also significantly reduced the amplitude of FP spike. JNJ303, an IKs inhibitor, only affected FPD. Comparison of the compound effect on FPD across the 3 different sites is consistent in terms of trend of the effect with observed 3-10 fold differences in minimal effective concentration at which a minimum of 10% response is detected. In addition, pentamidine, a hERG trafficking inhibitor which induced irregular beating activity over a more prolonged duration of time was readily flagged in this assay system. Taken together, this multi-parameter assay using hiPSC-CMs in conjunction with simultaneous measurement of ion channel activity and contractility can be a reliable approach for risk assessment of proarrhythmic compounds.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Arritmias Cardíacas/inducido químicamente , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Arritmias Cardíacas/fisiopatología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Guías como Asunto , Humanos , Células Madre Pluripotentes Inducidas , Bloqueadores de los Canales de Potasio/farmacología , Seguridad , Torsades de Pointes/inducido químicamente
4.
Toxicol Sci ; 131(2): 470-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23143926

RESUMEN

The anticancer drug (2-[4-amino-3-methylphenyl]-5-fluorobenzothiazole lysylamide dihydrochloride) (NSC 710305, Phortress) is a metabolically activated prodrug that causes DNA adduct formation and subsequent toxicity. Preclinically, it was found that hepatic, bone marrow, and pulmonary toxicity presented challenges to developing this drug. An ex vivo precision-cut lung slice (PCLS) model was used to search for concentration dependent effects of NSC 710305 (10, 25, 50, and 100 µM) on cytokine content, protein content, and immuno/histological endpoints. Preparation and culture of PCLS caused an initial spike in proinflammatory cytokine expression and therefore treatment with NSC 710305 was delayed until 48 h after initiating the slice cultures to avoid confounding the response to slicing with any drug response. PCLSs were evaluated after 24, 48, and 72 h exposures to NSC 710305. Reversibility of toxicity due to the 72-h treatment was evaluated after a 24-h recovery period. NSC 710305 caused a concentration-dependent cytokine response, and only the toxicity caused by a 72-h exposure to 25 µM reversed during the 24-h recovery period. Immuno/histological examination and quantitation of tissue protein levels indicated that tissue destruction, ED-1 (activated macrophage) staining, and protein levels were associated with the levels of proinflammatory cytokines in the tissue. In conclusion, the concentration- and time-dependent inflammatory response of PCLS to NSC 710305 preceded relevant tissue damage by a few days. The no-observable adverse effect level (NOAEL) for 24, 48, and 72 h exposures was established as 10 µM NSC 710305.


Asunto(s)
Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Lisina/análogos & derivados , Tiazoles/toxicidad , Animales , Técnicas In Vitro , Pulmón/patología , Lisina/toxicidad , Masculino , Ratas , Ratas Endogámicas F344
5.
J Med Chem ; 53(9): 3685-95, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20361799

RESUMEN

Among the known antimalarial drugs, chloroquine (CQ) and other 4-aminoquinolines have shown high potency and good bioavailability. Yet complications associated with drug resistance necessitate the discovery of effective new antimalarial agents. ADMET prediction studies were employed to evaluate a library of new molecules based on the 4-aminoquinolone-related structure of CQ. Extensive in vitro screening and in vivo pharmacokinetic studies in mice helped to identify two lead molecules, 18 and 4, with promising in vitro therapeutic efficacy, improved ADMET properties, low risk for drug-drug interactions, and desirable pharmacokinetic profiles. Both 18 and 4 are highly potent antimalarial compounds, with IC(50) values of 5.6 and 17.3 nM, respectively, against the W2 (CQ-resistant) strain of Plasmodium falciparum (for CQ, IC(50) = 382 nM). When tested in mice, these compounds were found to have biological half-lives and plasma exposure values similar to or higher than those of CQ; they are therefore desirable candidates to pursue in future clinical trials.


Asunto(s)
Aminoquinolinas/farmacología , Antimaláricos/química , Aminoquinolinas/uso terapéutico , Animales , Antimaláricos/farmacología , Evaluación Preclínica de Medicamentos , Semivida , Ratones , Farmacocinética , Plasmodium falciparum/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Toxicología
6.
Cancer Chemother Pharmacol ; 65(6): 1083-91, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19727731

RESUMEN

2-Amino-O4-benzylpteridine derivatives inactivate the human DNA repair protein O6-alkylguanine-DNA alkyltransferase and have been tested as modulators of alkylating agent chemotherapy. Recently, the therapeutic potential of O4-benzylfolate (O4BF) in modulating bis-chloroethylnitrosourea (BCNU) toxicity was demonstrated in vitro using human HT29 and KB tumor lines. The current studies replicated the previous findings in HT29 and KB cells using ATP as an endpoint. However, the effective treatment conditions were severely toxic to human neutrophil progenitors called CFU-granulocyte/macrophage (CFU-GM), which could not tolerate > or =40 microM BCNU at any O4BF concentration. A lower BCNU concentration (10 microM) in combination with O4BF (2-100 microM) was only moderately tumoricidal. To screen for conditions tolerated by CFU-GM, bone marrow (BM) cells were pre-incubated (5 h) with O4BF, co-treated with O4BF and BCNU (42 h), washed, and plated to quantify CFU-GM survival. O4BF at 2 or 5 microM progressively lowered the inhibitory concentrations (ICs) for BCNU, but further increases in O4BF concentrations did not. Increasing O4BF concentrations with constant BCNU (10 microM) under the same prolonged exposure as in the human marrow study achieved only modest tumoricidal effects. In summary, the unexpected finding that normal BM cells are impacted by an agent developed to target malignant tissue refutes speculation that normal beta-folate receptor expressing hematopoietic cells will be spared. Further, the validated IC90 endpoint from the huCFU-GM assay has provided a reference point for judging the potential therapeutic effectiveness of this investigational combination in man using in vitro assays.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Carmustina/farmacología , Ácido Fólico/análogos & derivados , Células Progenitoras de Granulocitos y Macrófagos/efectos de los fármacos , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ácido Fólico/farmacología , Células HT29 , Humanos , O(6)-Metilguanina-ADN Metiltransferasa/antagonistas & inhibidores , Factores de Tiempo
7.
J Pharm Pharmacol ; 58(4): 469-79, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16597364

RESUMEN

Resveratrol (trans-resveratrol, trans-3,5,4'-trihydroxystilbene) is a naturally occurring stilbene analogue found in high concentrations in red wine. There is considerable research interest to determine the therapeutic potential of resveratrol, as it has been shown to have tumour inhibitory and antioxidant properties. This study was performed to investigate the glucuronidation of resveratrol and possible drug interactions via glucuronidation. Two glucuronide conjugates, resveratrol 3-O-glucuronide and resveratrol 4'-O-glucuronide, were formed by human liver and intestinal microsomes. UGT1A1 and UGT1A9 were predominantly responsible for the formation of the 3-O-glucuronide (Km = 149 microM) and 4'-O-glucuronide (Km = 365 microM), respectively. The glucuronide conjugates were formed at higher levels (up to 10-fold) by intestinal rather than liver microsomes. Resveratrol was co-incubated with substrates of UGT1A1 (bilirubin and 7-ethyl-10-hydroxycamptothecin (SN-38)) and UGT1A9 (7-hydroxytrifluoromethyl coumarin (7-HFC)). No major changes were noted in bilirubin glucuronidation in the presence of resveratrol. Resveratrol significantly inhibited the glucuronidation of SN-38 (Ki = 6.2 +/- 2.1 microM) and 7-HFC (Ki = 0.6 +/- 0.2 microM). Hence, resveratrol has the potential to inhibit the glucuronidation of concomitantly administered therapeutic drugs or dietary components that are substrates of UGT1A1 and UGT1A9.


Asunto(s)
Glucurónidos/metabolismo , Glucuronosiltransferasa/metabolismo , Microsomas Hepáticos/metabolismo , Estilbenos/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Humanos , Insectos , Isoenzimas/metabolismo , Resveratrol , Estilbenos/química , UDP Glucuronosiltransferasa 1A9
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...