Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 546
Filtrar
2.
J Chromatogr Sci ; 59(3): 289-296, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33333557

RESUMO

In this study, we developed a new method for simultaneous determination of verapamil hydrochloride (VerHCl) and its metabolite norverapamil hydrochloride (NorHCl) by using the capillary electrophoresis-electrochemiluminescence. Under optimized experimental conditions, the linear ranges of the VerHCl and NorHCl concentrations were 0.015-10.0 and 0.060-10.0 µg/mL, respectively. The linearity relations were determined using the respective regression equations y = 581.2x + 19.94 and y = 339.4x + 29.16. The respective limits of detection (S/N = 3) were 0.006 and 0.024 µg/mL. The proposed method was used to study the pharmacokinetics of both agents in rat plasma. The maximum concentration (Cmax), half-life time (T1/2) and time to peak (Tmax) were 683.21 ± 74.81 ng/mL, 0.52 ± 0.21 h and 2.49 ± 0.32 h for VerHCl and 698.42 ± 71.45 ng/mL, 1.14 ± 0.26 h and 2.83 ± 0.23 h for NorHCl, respectively, following oral administration of 10 mg/kg VerHCl.


Assuntos
Eletroforese Capilar/métodos , Verapamil/análogos & derivados , Verapamil/sangue , Verapamil/farmacocinética , Animais , Limite de Detecção , Modelos Lineares , Medições Luminescentes , Ratos , Ratos Wistar , Reprodutibilidade dos Testes , Verapamil/química
3.
ACS Chem Neurosci ; 11(10): 1413-1423, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32286791

RESUMO

Amiodarone is prescribed for the treatment and prevention of irregular heartbeats. Although effective in clinical practice, the long-term use of amiodarone has many unwanted side effects, including cardiac, pulmonary, hepatic, and neurological toxicities. Our objective was to elucidate effects of amiodarone exposure on the cholesterol metabolism in cultured neuronal and non-neuronal cells and in individuals taking amiodarone. We observed that amiodarone increases distinct cholesterol precursors in different cell types in a dose-dependent manner. In liver and kidney cell lines, amiodarone causes increase in desmosterol levels, and in primary cortical neurons and astrocytes, amiodarone increases zymosterol, zymostenol, and 8-dehydrocholesterol (8-DHC). We conclude that amiodarone inhibits two enzymes in the pathway, emopamil binding protein (EBP) and dehydrocholesterol reductase 24 (DHCR24). Cortical neurons and astrocytes are more sensitive to amiodarone than liver and kidney cell lines. We confirmed the inhibition of EBP enzyme by analyzing the sterol intermediates in EBP-deficient Neuro2a cells versus amiodarone-treated control Neuro2a cells. To determine if the cell culture experiments have clinical relevance, we analyzed serum samples from amiodarone users. We found that in patient serum samples containing detectable amount of amiodarone there are elevated levels of the sterol precursors zymosterol, 8-DHC, and desmosterol. This study illustrates the need for close monitoring of blood biochemistry during prolonged amiodarone use to minimize the risk of side effects.


Assuntos
Amiodarona , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Amiodarona/farmacologia , Proteínas de Transporte , Colesterol , Desidrocolesteróis , Desmosterol , Humanos , Proteínas do Tecido Nervoso/metabolismo , Oxirredutases , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Verapamil/análogos & derivados
4.
Xenobiotica ; 50(6): 713-721, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31633443

RESUMO

In this study, UC rat model was established by administration of 5% (w/v) dextran sulfate sodium, and the pharmacokinetics of verapamil and norverapamil were evaluated in normal and UC rats using UPLC-MS/MS after oral administration of 5 mg/kg and 50 mg/kg verapamil.The peak concentration (Cmax) and the area under plasma concentration-time curves (AUC) of verapamil in UC rats after oral administration of 5 mg/kg were significantly greater (2.5 times and 2 times, respectively) than those in normal rats, but the clearance rate (Cl) was significantly lower (by 50%). For norverapamil, Cmax and AUC were significantly greater (2.8 times and 2.5 times, respectively), and Cl was significantly lower (by 45%). But, pharmacokinetic parameters of verapamil and norverapamil after oral administration of 50 mg/kg were no significant differences between UC and normal rats.The better absorption and poor excretion for low-dose verapamil may be attributed to down-regulation of P-gp expression in the intestine and kidney. No significant differences of pharmacokinetic parameters for high-dose verapamil may be explained as the saturation of an efflux mechanism.The findings of this study suggested that in UC patients, doses of verapamil should be decreased when low-dose verapamil was orally administrated.


Assuntos
Colite Ulcerativa/metabolismo , Verapamil/análogos & derivados , Verapamil/farmacocinética , Administração Oral , Animais , Área Sob a Curva , Bloqueadores dos Canais de Cálcio/farmacocinética , Cromatografia Líquida , Humanos , Masculino , Taxa de Depuração Metabólica/fisiologia , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem
5.
Drug Metab Dispos ; 47(11): 1291-1306, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31506301

RESUMO

Rivaroxaban is indicated for stroke prevention in nonvalvular atrial fibrillation (AF). Its elimination is mediated by both hepatic metabolism and renal excretion. Consequently, its clearance is susceptible to both intrinsic (pathophysiological) and extrinsic (concomitant drugs) variabilities that in turn implicate bleeding risks. Upon systematic model verification, physiologically based pharmacokinetic (PBPK) models are qualified for the quantitative rationalization of complex drug-drug-disease interactions (DDDIs). Hence, this study aimed to develop and verify a PBPK model of rivaroxaban systematically. Key parameters required to define rivaroxaban's disposition were either obtained from in vivo data or generated via in vitro metabolism and transport kinetic assays. Our developed PBPK model successfully predicted rivaroxaban's clinical pharmacokinetic parameters within predefined success metrics. Consideration of basolateral organic anion transporter 3 (OAT3)-mediated proximal tubular uptake in tandem with apical P-glycoprotein (P-gp)-mediated efflux facilitated mechanistic characterization of the renal elimination of rivaroxaban in both healthy and renal impaired patients. Retrospective drug-drug interaction (DDI) simulations, incorporating in vitro metabolic inhibitory parameters, accurately recapitulated clinically observed attenuation of rivaroxaban's hepatic clearance due to enzyme-mediated DDIs with CYP3A4/2J2 inhibitors (verapamil and ketoconazole). Notably, transporter-mediated DDI simulations between rivaroxaban and the P-gp inhibitor ketoconazole yielded minimal increases in rivaroxaban's systemic exposure when P-gp-mediated efflux was solely inhibited, but were successfully characterized when concomitant basolateral uptake inhibition was incorporated in the simulation. In conclusion, our developed PBPK model of rivaroxaban is systematically verified for prospective interrogation and management of untested yet clinically relevant DDDIs pertinent to AF management using rivaroxaban. SIGNIFICANCE STATEMENT: Rivaroxaban is susceptible to DDDIs comprising renal impairment and P-gp and CYP3A4/2J2 inhibition. Here, systematic construction and verification of a PBPK model of rivaroxaban, with the inclusion of a mechanistic kidney component, provided insight into the previously arcane role of OAT3-mediated basolateral uptake in influencing both clinically observed renal elimination of rivaroxaban and differential extents of transporter-mediated DDIs. The verified model holds potential for investigating clinically relevant DDDIs involving rivaroxaban and designing dosing adjustments to optimize its pharmacotherapy in atrial fibrillation.


Assuntos
Rivaroxabana/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Fibrilação Atrial/tratamento farmacológico , Simulação por Computador , Interações Medicamentosas , Humanos , Cetoconazol/farmacocinética , Rim/metabolismo , Modelos Biológicos , Transportadores de Ânions Orgânicos Sódio-Independentes/fisiologia , Rivaroxabana/uso terapêutico , Verapamil/análogos & derivados , Verapamil/farmacocinética
6.
Toxicol In Vitro ; 46: 1-8, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28986290

RESUMO

Drug-induced nephrotoxicity is one of the most frequent adverse events in pharmacotherapy. It has resulted in numerous clinical trial failures and high drug development costs. The predictive capabilities of existing in vitro models are limited by their inability to recapitulate the complex process of drug metabolism at the multi-organ level in vivo. We present a novel integrated liver-kidney chip that allows the evaluation of drug-induced nephrotoxicity following liver metabolism in vitro. The liver-kidney chip consists of two polydimethylsiloxane layers with compartmentalized micro-channels separated by a porous membrane. Hepatic and renal cells were co-cultured in separate micro-chambers on a single chip. Ifosfamide and verapamil were used as model drugs, and their metabolites produced by hepatic metabolism were identified using mass spectrometry, respectively. The metabolites triggered significantly distinct nephrotoxic effects as assessed by cell viability, lactate dehydrogenase leakage and permeability of renal cells. This in vitro liver-kidney model facilitates the characterization of drug metabolism in the liver as well as the assessment of subsequent nephrotoxicity in a single assay. Obviously, this multi-organ platform is simple and scalable, and maybe widely applicable to the evaluation of drug metabolism and safety during the early phases of drug development.


Assuntos
Técnicas de Cocultura/métodos , Células Epiteliais/efeitos dos fármacos , Rim/citologia , Dispositivos Lab-On-A-Chip , Fígado/citologia , Acroleína/toxicidade , Animais , Antineoplásicos Alquilantes/toxicidade , Bloqueadores dos Canais de Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Ifosfamida/toxicidade , Mostardas de Fosforamida/toxicidade , Ratos , Verapamil/análogos & derivados , Verapamil/metabolismo , Verapamil/toxicidade
7.
Biochemistry ; 56(19): 2506-2517, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28441502

RESUMO

The ATP binding cassette transporter P-glycoprotein (ABCB1 or P-gp) plays a major role in cellular resistance to drugs and drug interactions. Experimental studies support a mechanism with nucleotide-dependent fluctuation between inward-facing and outward-facing conformations, which are coupled to nucleotide hydrolysis. However, detailed insight into drug-dependent modulation of these conformational ensembles is lacking. Different drugs likely occupy partially overlapping but distinct sites and are therefore variably coupled to nucleotide binding and hydrolysis. Many fluorescent drug analogues are used in cell-based transport models; however, their specific interactions with P-gp have not been studied, and this limits interpretation of transport assays in terms of molecular models. Here we monitor binding of the fluorescent probe substrates BODIPY-verapamil, BODIPY-vinblastine, and Flutax-2 at low occupancy to murine P-gp in lipid nanodiscs via fluorescence correlation spectroscopy, in variable nucleotide-bound states. Changes in affinity for the different nucleotide-dependent conformations are probe-dependent. For BODIPY-verapamil and BODIPY-vinblastine, there are 2-10-fold increases in KD in the nucleotide-bound or vanadate-trapped state, compared to that in the nucleotide-free state. In contrast, the affinity of Flutax-2 is unaffected by nucleotide or vanadate trapping. In further contrast to BODIPY-verapamil and BODIPY-vinblastine, Flutax-2 does not cause stimulation of ATP hydrolysis despite the fact that it is transported in vesicle-based transport assays. Whereas the established substrates verapamil, paclitaxel, and vinblastine displace BODIPY-verapamil or BODIPY-vinblastine from their high-affinity sites, the transport substrate Flutax-2 is not displaced by any of these substrates. The results demonstrate a unique binding site for Flutax-2 that allows for transport without stimulation of ATP hydrolysis.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Bicamadas Lipídicas/química , Modelos Moleculares , Subfamília B de Transportador de Cassetes de Ligação de ATP/química , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Transporte Biológico , Compostos de Boro/metabolismo , Dimiristoilfosfatidilcolina/química , Corantes Fluorescentes/metabolismo , Humanos , Hidrólise , Cinética , Ligantes , Camundongos , Nanoestruturas/química , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Taxoides/metabolismo , Verapamil/análogos & derivados , Verapamil/metabolismo , Vimblastina/análogos & derivados , Vimblastina/metabolismo
8.
Nucl Med Biol ; 43(1): 52-62, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26429767

RESUMO

INTRODUCTION: 2-Isopropyl-5-[methyl-(2-phenylethyl)amino]-2-phenylpentanenitrile (emopamil; EMP) is a calcium channel blocker of the phenylalkylamine class, with weak substrate properties for P-glycoprotein (P-gp). A weak substrate for P-gp would be suitable for measuring enhanced P-gp function. This study was performed to synthesise (R)- and (S)-[(11)C]EMP and characterise their properties as P-gp tracers. METHODS: We synthesised (R)- and (S)-[(11)C]EMP and compared their biodistribution, peripheral metabolism, and effects of the P-gp inhibitor cyclosporine A (CsA, 50 mg/kg). We compared the brain pharmacokinetics of (R)-[(11)C]EMP and (R)-[(11)C]verapamil [(R)-[(11)C]VER] at baseline and CsA pretreatment with small animal positron emission tomography (PET). RESULTS: (R)- and (S)-[(11)C]EMP were synthesised from (R)- and (S)-noremopamil, respectively, by methylation with [(11)C]methyl triflate in the presence of NaOH at room temperature. (R)- and (S)-[(11)C]EMP yields were ~30%, with specific activity>74 GBq/µmol and radiochemical purity>99%. (R)-[(11)C]EMP showed significantly greater uptake in the mouse brain than (S)-[(11)C]EMP. Both showed homogeneous non-stereoselective regional brain distributions. (R)- and (S)-[(11)C]EMP were rapidly metabolised to hydrophilic metabolites. Unchanged plasma (S)-[(11)C]EMP level was significantly lower than that of (R)-[(11)C]EMP 15 minutes post-injection, whilst>88% of radioactivity in the brain was intact at 15 minutes post-injection and was non-stereoselective. CsA pretreatment increased brain activity ~3-fold in mice, but was non-stereoselective. The baseline area-under-the-curve (AUC) of brain radioactivity (0-60 minutes) of (R)-[(11)C]EMP was 2-fold higher than that of (R)-[(11)C]VER, but their AUCs after CsA pretreatment were comparable. CONCLUSIONS: (R)-[(11)C]EMP is a novel tracer for imaging P-gp function with higher baseline uptake than (R)-[(11)C]VER.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Radioisótopos de Carbono , Tomografia por Emissão de Pósitrons/métodos , Verapamil/análogos & derivados , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ciclosporina/farmacologia , Masculino , Camundongos , Ratos , Estereoisomerismo , Distribuição Tecidual/efeitos dos fármacos , Verapamil/química , Verapamil/farmacocinética
9.
Antimicrob Agents Chemother ; 60(3): 1216-25, 2015 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-26643325

RESUMO

There is a growing interest in repurposing mycobacterial efflux pump inhibitors, such as verapamil, for tuberculosis (TB) treatment. To aid in the design of better analogs, we studied the effects of verapamil on macrophages and Mycobacterium tuberculosis-specific T cells. Macrophage activation was evaluated by measuring levels of nitric oxide, tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1ß), and gamma interferon (IFN-γ). Since verapamil is a known autophagy inducer, the roles of autophagy induction in the antimycobacterial activities of verapamil and norverapamil were studied using bone marrow-derived macrophages from ATG5(flox/flox) (control) and ATG5(flox/flox) Lyz-Cre mice. Our results showed that despite the well-recognized effects of verapamil on calcium channels and autophagy, its action on intracellular M. tuberculosis does not involve macrophage activation or autophagy induction. Next, the effects of verapamil and norverapamil on M. tuberculosis-specific T cells were assessed using flow cytometry following the stimulation of peripheral blood mononuclear cells from TB-skin-test-positive donors with M. tuberculosis whole-cell lysate for 7 days in the presence or absence of drugs. We found that verapamil and norverapamil inhibit the expansion of M. tuberculosis-specific T cells. Additionally, three new verapamil analogs were found to inhibit intracellular Mycobacterium bovis BCG, and one of the three analogs (KSV21) inhibited intracellular M. tuberculosis replication at concentrations that did not inhibit M. tuberculosis-specific T cell expansion. KSV21 also inhibited mycobacterial efflux pumps to the same degree as verapamil. More interestingly, the new analog enhances the inhibitory activities of isoniazid and rifampin on intracellular M. tuberculosis. In conclusion, KSV21 is a promising verapamil analog on which to base structure-activity relationship studies aimed at identifying more effective analogs.


Assuntos
Mycobacterium tuberculosis/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Verapamil/análogos & derivados , Animais , Autofagia/efeitos dos fármacos , Humanos , Isoniazida/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/microbiologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos Transgênicos , Mycobacterium bovis/efeitos dos fármacos , Rifampina/análogos & derivados , Rifampina/farmacologia , Linfócitos T/microbiologia , Verapamil/farmacologia
10.
Food Chem Toxicol ; 84: 99-105, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26265240

RESUMO

Occupational toxicology and clinical pharmacology integration will be useful to understand potential exposure-drug interaction and to shape risk assessment strategies in order to improve occupational health. The aim of the present study was to evaluate the effect of exposure to ethanol fuel on in vivo activities of cytochrome P450 (CYP) isoenzymes CYP3A, CYP2C and CYP2D by the oral administration of the probe drugs verapamil, ibuprofen and fluoxetine. Male Wistar rats exposed to filtered air or to 2000 ppm ethanol in a nose-only inhalation chamber during (6 h/day, 5 days/week, 6 weeks) received single oral doses of 10 mg/kg verapamil or 25 mg/kg ibuprofen or 10 mg/kg fluoxetine. The enantiomers of verapamil, norverapamil, ibuprofen and fluoxetine in plasma were analyzed by LC-MS/MS. The area under the curve plasma concentration versus time extrapolated to infinity (AUC(0-∞)) was calculated using the Gauss-Laguerre quadrature. Inhalation exposure to ethanol reduces the AUC of both verapamil (approximately 2.7 fold) and norverapamil enantiomers (>2.5 fold), reduces the AUC(0-∞) of (+)-(S)-IBU (approximately 2 fold) and inhibits preferentially the metabolism of (-)-(R)-FLU. In conclusion, inhalation exposure of ethanol at a concentration of 2 TLV-STEL (6 h/day for 6 weeks) induces CYP3A and CYP2C but inhibits CYP2D in rats.


Assuntos
Biocombustíveis/toxicidade , Indutores das Enzimas do Citocromo P-450/toxicidade , Inibidores das Enzimas do Citocromo P-450/toxicidade , Sistema Enzimático do Citocromo P-450/metabolismo , Etanol/toxicidade , Exposição por Inalação/efeitos adversos , Testes de Toxicidade Crônica/métodos , Poluentes Ocupacionais do Ar/toxicidade , Animais , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/metabolismo , Câmaras de Exposição Atmosférica , Biomarcadores/sangue , Biotransformação/efeitos dos fármacos , Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/metabolismo , Inibidores das Enzimas do Citocromo P-450/sangue , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Sistema Enzimático do Citocromo P-450/química , Indução Enzimática/efeitos dos fármacos , Fluoxetina/sangue , Fluoxetina/farmacocinética , Ibuprofeno/sangue , Ibuprofeno/farmacocinética , Limoneno Hidroxilases/antagonistas & inibidores , Limoneno Hidroxilases/metabolismo , Masculino , Ratos Wistar , Verapamil/análogos & derivados , Verapamil/sangue , Verapamil/química , Verapamil/farmacocinética
13.
Drug Metab Dispos ; 43(4): 631-45, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25650380

RESUMO

We applied physiologically based pharmacokinetic (PBPK) modeling to study the dose-dependent metabolism and excretion of verapamil and its preformed metabolite, norverapamil, to unravel the kinetics of norverapamil formation via N-demethylation. Various initial verapamil (1, 50, and 100 µM) and preformed norverapamil (1.5 and 5 µM) concentrations, perfused at 12 ml/min, were investigated in the perfused rat liver preparation. Perfusate and bile were collected over 90 minutes, and livers were harvested at the end of perfusion for high-performance liquid chromatography analysis. After correction for the adsorption of 10%-25% dose verapamil and norverapamil onto Tygon tubing and binding to albumin and red blood cell, fitting of verapamil and formed and preformed norverapamil data with ADAPT5 revealed nonlinearity for protein binding, N-demethylation (V(max,met1)(VER --> NOR) = 96.6 ± 33.4 nmol/min; K(m,met1)(VER --> NOR) = 10.4 ± 4.1 µM), formation of other metabolites (V(max,met2(VER -->others) 288 ± 51 nmol/min; K(m.met2)(VER -->others )= 14.1 ± 4.9 µM), as well as biliary excretion (V(max,sec)(VER)= 0.911 ± 0.505 nmol/min; K(m,sec)(VER) = 4.75 ± 2.29 µM). The hepatic clearance of verapamil (CL(L)(VER) decreased with the dose (8.16-10.2 ml/min), with values remaining high relative to perfusate blood flow rate among the doses. The hepatic clearance of preformed norverapamil (11 ml/min) remained unchanged for the concentrations studied and approximated perfusate blood flow rate, suggesting a high norverapamil extraction ratio. The fractional formation of norverapamil and biliary excretion of verapamil based on fitted constants were 31.1% and 0.64% of CL(L)(VER), respectively. Enantiomeric disposition and auto-inhibition of verapamil failed to perturb these estimaties according to PBPK modeling, due to the low values of the Michaelis-Menten constant, Km, and inhibition parameter, kI.


Assuntos
Eritrócitos/metabolismo , Fígado/metabolismo , Modelos Biológicos , Verapamil/análogos & derivados , Animais , Proteínas Sanguíneas/metabolismo , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Masculino , Taxa de Depuração Metabólica , Dinâmica não Linear , Perfusão , Ligação Proteica , Ratos Sprague-Dawley , Estereoisomerismo , Fatores de Tempo , Distribuição Tecidual , Verapamil/sangue , Verapamil/química , Verapamil/metabolismo , Verapamil/farmacocinética
14.
Bioorg Med Chem Lett ; 24(14): 2985-90, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24894561

RESUMO

New verapamil analogues were synthesized and their inhibitory activities against Mycobacterium tuberculosis H37Rv determined in vitro alone and in combination with rifampicin (RIF). Some analogues showed comparable activity to verapamil and exhibited better synergies with RIF. Molecular docking studies of the binding sites of Rv1258c, a M. tuberculosis efflux protein previously implicated in intrinsic resistance to RIF, suggested a potential rationale for the superior synergistic interactions observed with some analogues.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/química , Mycobacterium tuberculosis/efeitos dos fármacos , Rifampina/farmacologia , Verapamil/análogos & derivados , Verapamil/farmacologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antibacterianos/síntese química , Proteínas de Bactérias/metabolismo , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Estrutura Molecular , Rifampina/química , Relação Estrutura-Atividade , Verapamil/síntese química , Verapamil/química
15.
J Infect Dis ; 210(3): 456-66, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24532601

RESUMO

Drug tolerance likely represents an important barrier to tuberculosis treatment shortening. We previously implicated the Mycobacterium tuberculosis efflux pump Rv1258c as mediating macrophage-induced tolerance to rifampicin and intracellular growth. In this study, we infected the human macrophage-like cell line THP-1 with drug-sensitive and drug-resistant M. tuberculosis strains and found that tolerance developed to most antituberculosis drugs, including the newer agents moxifloxacin, PA-824, linezolid, and bedaquiline. Multiple efflux pump inhibitors in clinical use for other indications reversed tolerance to isoniazid and rifampicin and slowed intracellular growth. Moreover, verapamil reduced tolerance to bedaquiline and moxifloxacin. Verapamil's R isomer and its metabolite norverapamil have substantially less calcium channel blocking activity yet were similarly active as verapamil at inhibiting macrophage-induced drug tolerance. Our finding that verapamil inhibits intracellular M. tuberculosis growth and tolerance suggests its potential for treatment shortening. Norverapamil, R-verapamil, and potentially other derivatives present attractive alternatives that may have improved tolerability.


Assuntos
Antituberculosos/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Macrófagos/fisiologia , Mycobacterium tuberculosis/efeitos dos fármacos , Verapamil/análogos & derivados , Verapamil/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Bloqueadores dos Canais de Cálcio/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Linhagem Celular , Humanos , Testes de Sensibilidade Microbiana
16.
Artigo em Inglês | MEDLINE | ID: mdl-25571292

RESUMO

BACKGROUND: Verapamil is a calcium channel blocker commonly used in treatments of hypertension. Verapamil and its active metabolite, norverapamil, are known to be CYP3A4 inhibitors. Co-administration of verapamil with CYP3A4 substrates can alter the pharmacokinetics of the substrates. Simvastatin, a commonly used HMG-CoA reductase inhibitor for the treatment of hypercholesterolemia is extensively metabolized by CYP3A4. Therefore, concomitant use of simvastatin and verapamil can increase simvastatin plasma concentration levels, resulting in a higher risk of rhabdomyolysis, a serious adverse drug reaction. Even though, pharmacokinetic data regarding the interaction between both drugs have been published, their use is limited to semiquantitative applications. Therefore, we aimed to develop a mathematical model describing drug-drug interaction between simvastatin and verapamil in humans. METHODS: Eligible pharmacokinetic interaction study between simvastatin and verapamil in humans was selected from PubMed database. The concentration-time courses from this study were digitally extracted and used for model development. RESULTS: The drug-drug interaction between simvastatin and verapamil was modeled simultaneously with a two compartment model for verapamil with its active metabolite, norverapamil and a one compartment model for simvastatin with its active form, simvastatin hydroxy acid. The effects of verapamil and norverapamil on pharmacokinetics of simvastatin and its active form, simvastatin hydroxy acid were described by Michaelis-Menten equation. Simulated simvastatin and simvastatin hydroxy acid concentrations obtained from the final model produced a good fit to the dataset from a literature. The final model adequately describes pharmacokinetic interaction between simvastatin and verapamil which can be helpful in prediction of rhabdomyolysis in patients with concurrent use of these drugs.


Assuntos
Anti-Hipertensivos/farmacocinética , Bloqueadores dos Canais de Cálcio/farmacocinética , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacocinética , Sinvastatina/análogos & derivados , Verapamil/análogos & derivados , Adulto , Área Sob a Curva , Estudos Cross-Over , Método Duplo-Cego , Incompatibilidade de Medicamentos , Interações Medicamentosas , Humanos , Hipertensão/induzido quimicamente , Modelos Biológicos , Sinvastatina/farmacocinética , Verapamil/farmacocinética , Adulto Jovem
17.
Mol Pharm ; 10(11): 4038-45, 2013 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-24044638

RESUMO

The Biopharmaceutics Drug Disposition Classification System (BDDCS) predicts intestinal transporter effects to be clinically insignificant following oral dosing for highly soluble and highly permeable/metabolized drugs (class 1 drugs). We investigated the effect of inhibiting P-glycoprotein (P-gp) on the in vitro rat intestinal permeability (Papp) and metabolism of the class 1 drug verapamil. Jejunal segments from Sprague-Dawley rats fasted overnight were mounted in Ussing chambers filled with 10 mL of Krebs-Ringer buffer (KRB). For P-gp inhibition studies, GG918 0.5 µM was added to the KRB solution. The experiment started by the addition of verapamil (1 or 10 µM) to either apical or basolateral sides. Samples from verapamil donor and receiver compartments were collected at 30 s and 0.166, 0.5, 1, 1.83 and 3 h after the start of the experiment. Analysis of verapamil and its major metabolite, norverapamil, in the samples and intracellularly at 3 h was performed by HPLC. The same experiment was repeated with norverapamil 10 µM (verapamil metabolite), digoxin 100 nM (positive control for P-gp activity) and atorvastatin 1 and 10 µM (example of a class 2 drug). For 1 µM verapamil, efflux ratio (B to A Papp/A to B Papp) was 4.6 and markedly decreased by GG918 (efflux ratio = 1.1). For 10 µM verapamil efflux ratio was 4.1 (control) vs 1.8 (GG918), comparable to the change seen for digoxin 100 nM with an efflux ratio of 3.6 (control) vs 1.6 (with GG918) and atorvastatin (efflux ratio of 5.2 and 3.0 for atorvastatin 1.0 and 10 µM, respectively, changed to 1.0 and 0.65 with GG918). The changes observed in the norverapamil 10 µM experiment were also significant, where efflux ratio decreased from 13.5 (control) to 1.5 (GG918). The extraction ratio (ER) of 10 µM verapamil to norverapamil decreased from 0.41 after an apical dose to 0.21 after a basolateral dose, but was unaffected by the incubation with GG918. The results suggest that P-gp inhibition has an effect on class 1 drug verapamil and class 2 drug atorvastatin Papp in the rat intestine. Moreover, a stronger P-gp effect on the Papp of the more polar norverapamil metabolite was observed. Papp changes caused by the P-gp inhibitor GG918 do not affect the extent of verapamil metabolism.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Verapamil/metabolismo , Verapamil/farmacocinética , Animais , Atorvastatina , Digoxina/metabolismo , Digoxina/farmacocinética , Ácidos Heptanoicos/metabolismo , Ácidos Heptanoicos/farmacocinética , Absorção Intestinal , Pirróis/metabolismo , Pirróis/farmacocinética , Ratos , Ratos Sprague-Dawley , Verapamil/análogos & derivados
18.
Eur J Pharm Sci ; 50(3-4): 290-302, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23916407

RESUMO

Verapamil and its major metabolite norverapamil were identified to be both mechanism-based inhibitors and substrates of CYP3A and reported to have non-linear pharmacokinetics in clinic. Metabolic clearances of verapamil and norverapmil as well as their effects on CYP3A activity were firstly measured in pooled human liver microsomes. The results showed that S-isomers were more preferential to be metabolized than R-isomers for both verapamil and norverapamil, and their inhibitory effects on CYP3A activity were also stereoselective with S-isomers more potent than R-isomers. A semi-physiologically based pharmacokinetic model (semi-PBPK) characterizing mechanism-based auto-inhibition was developed to predict the stereoselective pharmacokinetic profiles of verapamil and norverapamil following single or multiple oral doses. Good simulation was obtained, which indicated that the developed semi-PBPK model can simultaneously predict pharmacokinetic profiles of S-verapamil, R-verapamil, S-norverapamil and R-norverapamil. Contributions of auto-inhibition to verapamil and norverapamil accumulation were also investigated following the 38th oral dose of verapamil sustained-release tablet (240mg once daily). The predicted accumulation ratio was about 1.3-1.5 fold, which was close to the observed data of 1.4-2.1-fold. Finally, the developed semi-PBPK model was further applied to predict drug-drug interactions (DDI) between verapamil and other three CYP3A substrates including midazolam, simvastatin, and cyclosporine A. Successful prediction was also obtained, which indicated that the developed semi-PBPK model incorporating auto-inhibition also showed great advantage on DDI prediction with CYP3A substrates.


Assuntos
Modelos Biológicos , Verapamil/análogos & derivados , Verapamil/química , Verapamil/farmacocinética , Bloqueadores dos Canais de Cálcio/administração & dosagem , Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/farmacocinética , Ciclosporina/administração & dosagem , Ciclosporina/sangue , Ciclosporina/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Interações Medicamentosas , Mucosa Gástrica/metabolismo , Humanos , Intestino Delgado/metabolismo , Fígado/metabolismo , Microssomos Hepáticos/metabolismo , Midazolam/administração & dosagem , Midazolam/sangue , Midazolam/farmacocinética , Sinvastatina/administração & dosagem , Sinvastatina/sangue , Sinvastatina/farmacocinética , Estereoisomerismo , Verapamil/administração & dosagem , Verapamil/metabolismo
19.
Acta Pol Pharm ; 70(3): 395-401, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23757929

RESUMO

A stereospecific capillary zone electrophoresis (CZE) method was developed for the determination of verapamil (VER) and its main metabolite norverapamil (NOR) in human plasma. Optimal temperature, cyclodextrin selectors (CDs), pH of background electrolyte (BGE) and voltage were established to obtain complete separation of the chiral analytes and clopidogrel internal standard (I.S.) during one analytical run. Successful resolution of analytes was obtained in silica capillary filled with BGE consisting of heptakis 2,3,6-tri-O-methyl-beta-CD in phosphate buffer, pH 2.5 at 15 degrees C of capillary temperature. The calculated electrophoretic parameters of the analytes were as follows: apparent electrophoretic mobility, for VER enantiomers: micro(ap(+)-R) = 1.1 x 10(-4), micro(ap(-)s) = 1.06 x 10(-4) cm2/Vs and for NOR enantiomers: micro(ap(+)-R) = 1.09 x 10(-4), micro(ap(-)s) = 1.04 x 10(-4) cm2/Vs, resolution factors, R(s) = 5.4-6.6. Liquid extraction was applied for isolation of the analytes. The calibration curves ware linear in the range 0.25-10 microg/mL for VER and NOR enantiomer concentrations. The validation parameters were also established. The precision and accuracy of intra- and inter-day analysis were less than 15%. The lower limit of detection and limit of quantification for single enantiomers were 0.1 and 0.2 microg/mL, respectively. Recovery of the enantiomers from plasma was in the 91-103% range. To evaluate analytical applicability of the proposed method, plasma sample from patient suffering from arterial hypertension treated with 80 mg of commercial tablets was analyzed.


Assuntos
Bloqueadores dos Canais de Cálcio/sangue , Eletroforese Capilar/métodos , Verapamil/análogos & derivados , Verapamil/sangue , Humanos , Limite de Detecção , Estereoisomerismo
20.
J Pharm Sci ; 102(9): 3161-73, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23686764

RESUMO

Digoxin is the recommended substrate for assessment of P-glycoprotein (P-gp)-mediated drug-drug interactions (DDIs) in vivo. The overall aim of our study was to investigate the inhibitory potential of both verapamil and norverapamil on the P-gp-mediated efflux of digoxin in both gut and liver. Therefore, a physiologically-based pharmacokinetic (PBPK) model for verapamil and its primary metabolite was developed and validated through the recovery of observed clinical plasma concentration data for both moieties and the reported interaction with midazolam, albeit a cytochrome P450 3A4-mediated DDI. The validated inhibitor model was then used in conjunction with the model developed previously for digoxin. The range of values obtained for the 10 trials indicated that increases in area under the plasma concentration-time curve (AUC) profiles and maximum plasma concentration observed (Cmax ) values of digoxin following administration of verapamil were more comparable with in vivo observations, when P-gp inhibition by the metabolite, norverapamil, was considered as well. The predicted decrease in AUC and Cmax values of digoxin following administration of rifampicin because of P-gp induction was 1.57- (range: 1.42-1.77) and 1.62-fold (range: 1.53-1.70), which were reasonably consistent with observed values of 1.4- and 2.2-fold, respectively. This study demonstrates the application of permeability-limited models of absorption and distribution within a PBPK framework together with relevant in vitro data on transporters to assess the clinical impact of modulated P-gp-mediated efflux by drugs in development.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Antiarrítmicos/farmacologia , Antiarrítmicos/farmacocinética , Digoxina/farmacologia , Digoxina/farmacocinética , Verapamil/análogos & derivados , Verapamil/farmacologia , Adjuvantes Anestésicos/metabolismo , Adjuvantes Anestésicos/farmacologia , Antiarrítmicos/metabolismo , Simulação por Computador , Digoxina/metabolismo , Interações Medicamentosas , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Midazolam/metabolismo , Midazolam/farmacologia , Modelos Biológicos , Permeabilidade/efeitos dos fármacos , Rifampina/metabolismo , Rifampina/farmacologia , Verapamil/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...