Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anal Bioanal Chem ; 412(17): 4057-4065, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32248396

RESUMO

Graphene oxide (GO) has the ability to absorb certain compounds, and it can be modified with functional groups for different purposes; for instance, iron oxide (IO) nanoparticles can be used to concentrate analyte by a magnet. Recently, many kinds of GO have been developed, such as single-layer GO (SLGO), two-to-four layers of GO (i.e., few-layer GO, FLGO2-4), and four-to-eight layers of GO (i.e., multi-layer GO, MLGO4-8). However, the abilities of these layered GO coated with IO nanoparticles have not been investigated. In this study, we conducted a novel analysis of glimepiride by using layered GO-coated magnetic clusters of IO nanoparticles that were synthesized through a simple and facile emulsion-solvent evaporation method. The methodology is based on (i) enrichment of glimepiride using the layered GO-coated magnetic clusters of IO nanoparticles (IO@SLGO, IO@FLGO2-4, and IO@MLGO4-8), and (ii) rapid determination using magnetic cluster-based surface-assisted laser desorption/ionization time-of-flight mass spectrometry (SALDI-TOFMS). We found that IO@MLGO4-8, the magnetic cluster with the greatest number of GO layers, had the best limit of detection (28.6 pmol/µL for glimepiride). The number of GO layers played a significant role in increasing the sensitivity of the SALDI-MS, indicating that the size of GO in the magnetic clusters contributed to the desorption/ionization efficiency. To the best of our knowledge, this is the first study to enrich glimepiride using magnetic clusters of different GO types and to show that the glimepiride in HLB purified urine adsorbed by magnetic clusters can be analyzed by SALDI-TOFMS.


Assuntos
Grafite/química , Hipoglicemiantes/urina , Nanopartículas Magnéticas de Óxido de Ferro/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Compostos de Sulfonilureia/urina , Adsorção , Antiarrítmicos/isolamento & purificação , Antiarrítmicos/urina , Humanos , Hipoglicemiantes/isolamento & purificação , Limite de Detecção , Extração Líquido-Líquido/métodos , Nanopartículas Magnéticas de Óxido de Ferro/ultraestrutura , Extração em Fase Sólida/métodos , Compostos de Sulfonilureia/isolamento & purificação
2.
J Toxicol Environ Health A ; 81(12): 481-492, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29641932

RESUMO

Amiodarone is a class III anti-arrhythmic benzofuran derivative extensively utilized in treatment of life-threatening ventricular and supraventricular arrhythmias. However, amiodarone also produces adverse side effects including liver injury due to its metabolites rather than parent drug. The purpose of the present study was to identify metabolites of amiodarone in the plasma and urine of rats administered the drug by using an untargeted metabolomics approach. Drug metabolites were profiled by ultra-performance liquid chromatography-linked electrospray ionization quadrupole time-of-flight mass spectrometry (UPLC-ESI-QTOFMS) and results subjected to multivariate data analysis. A total of 49 amiodarone metabolites were identified and their structures were characterized by tandem mass spectrometry. Amiodarone metabolites are presumed to be generated via five major types of metabolic reactions including N-desethylation, hydroxylation, carboxylation (oxo/hydroxylation), de-iodination, and glucuronidation. Data demonstrated that an untargeted metabolomics approach appeared to be a reliable tool for identifying unknown metabolites in a complex biological matrix.


Assuntos
Amiodarona/metabolismo , Antiarrítmicos/metabolismo , Amiodarona/sangue , Amiodarona/urina , Animais , Antiarrítmicos/sangue , Antiarrítmicos/urina , Cromatografia Líquida de Alta Pressão , Masculino , Metabolômica , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
3.
Electrophoresis ; 37(2): 339-46, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26462723

RESUMO

Electromembrane extraction (EME) of model analytes was carried out using a virtually rotating supported liquid membrane (SLM). The virtual (nonmechanical) rotating of the SLM was achieved using a novel electrode assembly including a central electrode immersed inside the lumen of the SLM and five counter electrodes surrounding the SLM. A particular electronic circuit was designed to distribute the potential among five counter electrodes in a rotating pattern. The effect of the experimental parameters on the recovery of the extraction was investigated for verapamil (VPL), trimipramine (TRP), and clomipramine (CLP) as the model analytes and 2-ethyl hexanol as the SLM solvent. The results showed that the recovery of the extraction is a function of the angular velocity of the virtual rotation. The best results were obtained at an angular velocity of 1.83 RadS(-1) (or a rotation frequency of 0.29 Hz).The optimization of the parameters gave higher recoveries up to 50% greater than those of a conventional EME method. The rotating also allowed the extraction to be carried out at shorter time (15 min) and lower voltage (200 V) with respect to the conventional extraction. The model analytes were successfully extracted from wastewater and human urine samples with recoveries ranging from 38 to 85%. The RSD of the determinations was in the range of 12.6 to 14.8%.


Assuntos
Fracionamento Químico/instrumentação , Clomipramina/isolamento & purificação , Técnicas Eletroquímicas/instrumentação , Membranas Artificiais , Trimipramina/isolamento & purificação , Verapamil/isolamento & purificação , Antiarrítmicos/isolamento & purificação , Antiarrítmicos/urina , Antidepressivos Tricíclicos/isolamento & purificação , Antidepressivos Tricíclicos/urina , Clomipramina/urina , Eletrodos , Desenho de Equipamento , Humanos , Limite de Detecção , Rotação , Trimipramina/urina , Urinálise/instrumentação , Verapamil/urina , Águas Residuárias/análise , Poluentes Químicos da Água/isolamento & purificação , Poluentes Químicos da Água/urina , Purificação da Água/instrumentação
4.
Artigo em Inglês | MEDLINE | ID: mdl-25618251

RESUMO

The present work is based on a one-step method including derivatization and solid-based disperser liquid-liquid microextraction followed by gas chromatography-flame ionization detection (GC-FID) for the determination of four antidepressants (fluoxetine, fluvoxamine, tranylcypromine, and nortriptyline) and an antiarrhythmic agent (mexiletine) in human urine and plasma samples. In this method, a mixture of 1,1,2,2-tetrachloroethane (extraction solvent) and butylchloroformate (derivatizing reagent) is added on a sugar cube (solid disperser) and it is introduced into an aqueous sample containing the analytes and a catalyst, e.g. 3-methylpyridine (picoline). During dissolving the sugar cube by manual shaking, the extractant and derivatization agent are gradually released into the sample as very fine droplets. Then the resulted cloudy solution is centrifuged and the sedimented phase is analyzed by GC-FID. The influence of several variables on the efficiency of derivatization/microextraction procedure such as kind and volume of extraction solvent, type and amount of disperser, amount of derivatization agent, and catalyst volume are optimized. Under the optimum conditions the calibration curves are linear in the range of 8-100,000µgL(-1) (coefficient of determination ≥0.994). The relative standard deviations are obtained in the range of 3.0-6.0% for all compounds. Moreover, the detection limits and enrichment factors of the target analytes are obtained in the ranges 1-15µgL(-1) and 228-268, respectively, for both plasma and urine samples. The relative recoveries obtained for the spiked plasma and urine samples are between 70 and 91%. The results show that the proposed method is simple, reliable, low cost, and applicable to determine trace amounts of the studied drugs in biological samples.


Assuntos
Antiarrítmicos/sangue , Antiarrítmicos/urina , Antidepressivos/sangue , Antidepressivos/urina , Cromatografia Gasosa/métodos , Ionização de Chama/métodos , Microextração em Fase Líquida/métodos , Adulto , Feminino , Humanos
5.
Pharmacotherapy ; 34(5): e30-3, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24510469

RESUMO

Flecainide is recommended as a first-line antiarrhythmic drug to maintain normal sinus rhythm in patients with atrial fibrillation (AF) who have structurally normal hearts or hypertension without left ventricular hypertrophy. Flecainide is a sodium channel blocker with minimal effects expected on ventricular repolarization. We describe the case of a 32-year-old man with a structurally normal heart and persistent AF who was started on diltiazem and flecainide 50 mg twice/day approximately a year prior to presentation. Due to persistent and bothersome symptoms, his dose was increased to 150 mg twice/day, which was associated with a progressive lengthening of his corrected QT interval. On the day of presentation, he underwent an exercise test as part of his job requirements. While running, he felt lightheaded and experienced a syncopal event and cardiac arrest. An automated external defibrillator was available that displayed polymorphic ventricular tachycardia. The patient was successfully resuscitated. Although rare, this case suggests that flecainide can induce QT prolongation leading to torsades de pointes. Clinicians should be aware and consider periodic evaluations with electrocardiograms.


Assuntos
Antiarrítmicos/efeitos adversos , Flecainida/efeitos adversos , Parada Cardíaca/induzido quimicamente , Síndrome do QT Longo/induzido quimicamente , Adulto , Antiarrítmicos/administração & dosagem , Antiarrítmicos/urina , Fibrilação Atrial/tratamento farmacológico , Desfibriladores , Flecainida/administração & dosagem , Flecainida/uso terapêutico , Parada Cardíaca/terapia , Humanos , Síndrome do QT Longo/terapia , Masculino , Ressuscitação , Resultado do Tratamento
6.
Electrophoresis ; 34(20-21): 2962-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24037989

RESUMO

Optimization based on central composite design (CCD) for enantioseparation of anisodamine (AN), atenolol (AT), and metoprolol (ME) in human urine was developed using a microfluidic chip-CE device. Coupling the flexible and wide working range of microfluidic chip-CE device to CCD for chiral separation of AN, AT, and ME in human urine, a total of 15 experiments is needed for the optimization procedure as compared to 75 experiments using the normal one variable at a time optimization. The optimum conditions obtained are found to be more robust as shown by the curvature effects of the interaction factors. The developed microfluidic chip-CE-ECL system with adjustable dilution ratios has been validated by satisfactory recoveries (89.5-99% for six enanotiomers) in urine sample analysis. The working range (0.3-600 µM), repeatability (3.1-4.9% RSD for peak height and 4.0-5.2% RSD for peak area), and detection limit (0.3-0.6 µM) of the method developed are found to meet the requirements for bedside monitoring of AN, AT, and ME in patients under critical conditions. In summary, the hyphenation of CCD with the microfluidic chip-CE device is shown to offer a rapid means for optimizing the working conditions on simultaneous separation of three racemic drugs using the microfluidic chip-CE device developed.


Assuntos
Antiarrítmicos/urina , Atenolol/urina , Eletroforese em Microchip/instrumentação , Metoprolol/urina , Alcaloides de Solanáceas/urina , Antiarrítmicos/isolamento & purificação , Atenolol/isolamento & purificação , Desenho de Equipamento , Humanos , Limite de Detecção , Medições Luminescentes/instrumentação , Metoprolol/isolamento & purificação , Reprodutibilidade dos Testes , Alcaloides de Solanáceas/isolamento & purificação , Estereoisomerismo
7.
Curr Med Res Opin ; 23(7): 1531-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17559748

RESUMO

OBJECTIVE: NXY-059 is a novel free-radical trapping neuroprotectant. Digoxin treatment is common in acute ischaemic stroke, the intended patient population for NXY-059. Since both digoxin and NXY-059 are eliminated primarily renally, with a substantial contribution by active renal secretion, and because digoxin has a narrow therapeutic window, this open, randomised, crossover, two-period study investigated whether NXY-059 affects the pharmacokinetics (PK) of digoxin. RESEARCH DESIGN AND METHODS: Twenty-two healthy subjects received 0.5 mg oral digoxin 2 h after the start of 60-h intravenous infusions of NXY-059 and placebo separated by a 14-day washout. Blood and urine were collected for 60 h. Digoxin concentrations were measured by a novel liquid chromatography-mass spectrometry assay. MAIN OUTCOME MEASURES: The ratio of the geometric mean (90% confidence interval) between NXY-059 and placebo for the digoxin area under the concentration-versus-time curve was 0.91 (0.83-0.99) and was within the predefined range for no interaction (0.80-1.25). No safety concerns were raised in the study. No serious adverse events were recorded. The most common adverse event was headache with similar frequencies in the two treatments. CONCLUSIONS: NXY-059 had no clinically significant effect on the PK of digoxin.


Assuntos
Antiarrítmicos/farmacocinética , Benzenossulfonatos/administração & dosagem , Digoxina/farmacocinética , Fármacos Neuroprotetores/administração & dosagem , Adolescente , Adulto , Antiarrítmicos/sangue , Antiarrítmicos/urina , Benzenossulfonatos/sangue , Benzenossulfonatos/urina , Cromatografia Líquida/métodos , Digoxina/sangue , Digoxina/urina , Feminino , Humanos , Masculino , Espectrometria de Massas/métodos , Pessoa de Meia-Idade , Fármacos Neuroprotetores/sangue , Fármacos Neuroprotetores/urina
8.
Artigo em Inglês | MEDLINE | ID: mdl-17448738

RESUMO

A rapid, sensitive and specific liquid chromatographic-electrospray ionization (ESI) tandem ion trap mass spectrometric method has been developed for identification of dauricine and its metabolites in rat urine. Six healthy rats were administrated a single dose (100 mg/kg) of dauricine by oral gavage. The urine were sampled from 0 to 24 h and purified by using a C18 solid-phase extraction (SPE) cartridge, then the purified urine samples were separated on a reversed-phase C18 column using methanol/2 mmol/L ammonium acetate (70:30, v/v, adjusted to pH 3.5 with formic acid) as mobile phase and detected by an on-line MS detector. Identification and structural elucidation of the metabolites were performed by comparing their changes in molecular mass (Deltam) and full scan MS(n) spectra with those of the parent drug. At least eight metabolites (such as N-demethyl, dehydrogenate, demethoxyl, hydroxyl, glucuronide conjugated and sulfate conjugated metabolites) and the parent drug were found in rat urine.


Assuntos
Alcaloides/urina , Antiarrítmicos/urina , Benzilisoquinolinas/urina , Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas em Tandem/métodos , Tetra-Hidroisoquinolinas/urina , Animais , Ratos , Ratos Wistar , Sensibilidade e Especificidade
9.
Br J Clin Pharmacol ; 60(6): 659-63, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16305592

RESUMO

AIMS: We aimed to investigate effects of grapefruit juice on acebutolol pharmacokinetics. METHODS: In a randomized cross-over study, 10 healthy subjects ingested 200 mL grapefruit juice or water three times daily for 3 days and twice on day 4. On day 3, each subject ingested 400 mg acebutolol with grapefruit juice or water. The concentrations of acebutolol and its metabolite diacetolol were measured in plasma and urine up to 33 h. RESULTS: Grapefruit juice decreased the peak plasma concentration (Cmax) of acebutolol by 19% from 872 +/- 207 ng mL(-1) to 706 +/- 140 ng mL(-1) (95% CI on the difference -306, -26.4; P < 0.05), and the area under the concentration time curve (AUC(0-33 h)) by 7%, from 4498 +/- 939 ng mL(-1) h to 4182 +/- 915 ng mL(-1) h (95% CI -609, -23.0; P < 0.05). The half-life (t1/2) of acebutolol prolonged from 4.0 to 5.1 h (P < 0.05). The time to peak concentration and the amount of acebutolol excreted into urine (Ae) were unchanged. The Cmax, AUC(0-33 h), and Ae of diacetolol were decreased by 24% (P < 0.05), 18% (P < 0.05), and 20% (P < 0.01), respectively, by grapefruit juice. CONCLUSION: Grapefruit juice caused a small decrease in the plasma concentrations of acebutolol and diacetolol by interfering with gastrointestinal absorption. The interaction between the grapefruit juice and acebutolol is unlikely to be of clinical significance in most of the patients.


Assuntos
Acebutolol/sangue , Antagonistas Adrenérgicos beta/sangue , Bebidas , Citrus paradisi , Interações Alimento-Droga , Acebutolol/análogos & derivados , Acebutolol/urina , Antagonistas Adrenérgicos beta/urina , Adulto , Antiarrítmicos/sangue , Antiarrítmicos/urina , Anti-Hipertensivos/uso terapêutico , Estudos Cross-Over , Feminino , Hemodinâmica/efeitos dos fármacos , Humanos , Masculino
10.
J Pharm Sci ; 94(9): 2084-95, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16052551

RESUMO

The metabolic fate of azimilide in man is unusual as it undergoes a cleavage in vivo resulting in the formation of two classes of structurally distinct metabolites. During a metabolite profiling study conducted in human volunteers to assess the contribution of all pathways to the clearance of (14)C-azimilide, greater than 82% of radioactivity was recovered in urine (49%-58%) and feces (33%). Urine, feces, and plasma were profiled for metabolites. A cleaved metabolite, 4-chloro-2-phenyl furoic acid was present at high concentration in plasma (metabolite/parent AUC ratio approx. 4), while other plasma metabolites, azimilide N-oxide (metabolite/parent AUC ratio 0.001), and a cleaved hydantoin metabolite (metabolite/parent AUC ratio = 0.3) were present at lower concentrations than azimilide. In urine, the cleaved metabolites were the major metabolites, (> 35% of the dose) along with phenols (as conjugates, 7%-8%), azimilide N-oxide (4%-10%), a butanoic acid metabolite (2%-3%), and desmethyl azimilide (2%). A limited investigation of fecal metabolites indicated that azimilide (3%-5%), desmethyl azimilide (1%-3%), and the butanoic acid metabolite (< 1%) were present. Contributing pathways for metabolism of azimilide, identified through in vitro and in-vivo studies, were CYPs 1A1 (est. 28%), 3A4/5 (est. 20%), 2D6 (< 1%), FMO (est. 14%), and cleavage (35%). Enzyme(s) involved in the cleavage of azimilide were not identified.


Assuntos
Antiarrítmicos/farmacocinética , Imidazolidinas/farmacocinética , Microssomos Hepáticos/metabolismo , Piperazinas/farmacocinética , Adulto , Antiarrítmicos/sangue , Antiarrítmicos/urina , Área Sob a Curva , Radioisótopos de Carbono , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/metabolismo , Fezes/química , Humanos , Hidantoínas , Imidazolidinas/sangue , Imidazolidinas/urina , Técnicas In Vitro , Masculino , Microssomos Hepáticos/enzimologia , Piperazinas/sangue , Piperazinas/urina , Fatores de Tempo
11.
J Clin Pharmacol ; 45(7): 773-80, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15951467

RESUMO

The influence of coadministration on digoxin and azimilide pharmacokinetics/pharmacodynamics was assessed in a randomized, 3-way crossover study in 18 healthy men. Serial blood and urine samples were obtained for azimilide and digoxin quantitation. Treatment effects on pharmacokinetics were assessed using analysis of variance. The relationship between azimilide blood concentrations and QT(c) prolongation was characterized by an E(max) model. Effects of coadministration on pharmacodynamics were assessed using a mechanistic-based inhibition model. Azimilide pharmacokinetics was unaffected by digoxin, except for a 36% increase in CL(r) (P = .0325), with no change in CL(o). Digoxin pharmacokinetics was unaffected by azimilide, except for a 21% increase in C(max) (P = .0176) and a 10% increase in AUC(tau) (P = .0121). Digoxin coadministration increased the apparent EC(50) with no effect on E(max), consistent with competitive inhibition (K(i) = 0.899 ng/mL). The pharmacokinetic and pharmacodynamic changes observed upon coadministration were small and are not expected to be clinically important.


Assuntos
Antiarrítmicos/farmacocinética , Cardiotônicos/farmacocinética , Digoxina/farmacocinética , Imidazolidinas/farmacocinética , Piperazinas/farmacocinética , Adolescente , Adulto , Antiarrítmicos/administração & dosagem , Antiarrítmicos/sangue , Antiarrítmicos/urina , Cardiotônicos/administração & dosagem , Cardiotônicos/sangue , Cardiotônicos/urina , Estudos Cross-Over , Digoxina/administração & dosagem , Digoxina/sangue , Digoxina/urina , Combinação de Medicamentos , Interações Medicamentosas , Eletrocardiografia , Humanos , Hidantoínas , Imidazolidinas/administração & dosagem , Imidazolidinas/sangue , Imidazolidinas/urina , Masculino , Piperazinas/administração & dosagem , Piperazinas/sangue , Piperazinas/urina
12.
J Anal Toxicol ; 27(8): 595-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14670140

RESUMO

First synthesized in 1970, propafenone is a frequently used 1C antiarrhythmic drug metabolized into two major metabolites, 5-hydroxypropafenone and norpropafenone. Paradoxically, fatal intoxication is rarely described, and only six cases have been reported in the literature. We report our experience with two patients found dead of self-inflicted poisoning where the propafenone blood concentration was very high (one concentration to our knowledge is one of the highest reported in the literature). At autopsy, no evidence of significant pathological disease were found. Propafenone was detected in blood by gas chromatography-mass spectrometry and by high-performance liquid chromatography using a diode-array detector, respectively, as propafenone artifact and propafenone. Blood propafenone concentrations were 4180 ng/mL and 9123 ng/mL. The literature regarding propafenone pharmacokinetic and intoxication is reviewed, and we discuss the low death rate attributed to this drug in contrast to its frequent use.


Assuntos
Antiarrítmicos/intoxicação , Propafenona/intoxicação , Adulto , Antiarrítmicos/sangue , Antiarrítmicos/urina , Overdose de Drogas , Evolução Fatal , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Masculino , Metanol/sangue , Pessoa de Meia-Idade , Propafenona/sangue , Propafenona/urina
13.
Br J Clin Pharmacol ; 56(1): 32-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12848773

RESUMO

AIMS: A clinically important interaction between the cardiac glycoside digoxin and the antibiotic clarithromycin has been suggested in earlier reports. The aim of this study was to investigate the extent of the interaction and the relative contribution of different mechanisms. METHODS: In a randomized, placebo-controlled, double-blind cross-over design single oral doses of 0.75 mg digoxin with oral coadministration of placebo or 250 mg clarithromycin twice daily for 3 days were administered to 12 healthy men. Additionally, three of the subjects received single intravenous doses of 0.01 mg x kg(-1) digoxin with oral placebo or clarithromycin. Digoxin plasma and urine concentrations were determined by a highly sensitive radioimmunoassay. RESULTS: Oral coadministration of clarithromycin resulted in a 1.7-fold increase of the area under the digoxin plasma concentration-time curve [mean AUC(0,24) +/- SD 23 +/- 5.2 vs. 14 +/- 2.9 microg x L(-1) x h; 95% confidence interval (CI) on the difference 7.0, 12; P = 0.002] and in a reduction of the nonglomerular renal clearance of digoxin [mean ClRng(0, 24) +/- SD 34 +/- 39 vs. 57 +/- 41 mL min-1; 95% CI on the difference 7.2, 45; P = 0.03]. The ratios of mean digoxin plasma concentrations with and without clarithromycin were highest during the absorption period of clarithromycin. After intravenous administration digoxin AUC(0,24) increased only 1.2-fold during coadministration of clarithromycin. CONCLUSIONS: Increased oral bioavailability and reduced nonglomerular renal clearance of digoxin both contribute to the interaction between digoxin and clarithromycin, probably due to inhibition of intestinal and renal P-glycoprotein.


Assuntos
Antiarrítmicos/farmacocinética , Antibacterianos/farmacocinética , Claritromicina/farmacocinética , Digoxina/farmacocinética , Administração Oral , Adulto , Antiarrítmicos/sangue , Antiarrítmicos/urina , Antibacterianos/administração & dosagem , Área Sob a Curva , Disponibilidade Biológica , Claritromicina/administração & dosagem , Creatinina/sangue , Creatinina/urina , Estudos Cross-Over , Digoxina/sangue , Digoxina/urina , Método Duplo-Cego , Interações Medicamentosas , Genes MDR/genética , Genótipo , Humanos , Infusões Intravenosas , Rim/metabolismo , Masculino , Polimorfismo Genético , Estudos Prospectivos
14.
Eur J Drug Metab Pharmacokinet ; 28(4): 265-72, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14743967

RESUMO

Guanfu base A is a novel arrhythmic drug candidate isolated from the tuber of a traditional Chinese herb. Phase I and Phase II metabolites of Guanfu base A (GFA) Hydrochloride were studied in human urine by means of liquid chromatography mass spectrometry (LC/MSD) and tandem mass spectrometry (MS/MS). For phase I metabolites, Guanfu base I (GFI) was separated by HPLC and identified by comparison with authentic reference for their retention times, molecular ion peaks, fragment ions, and UV spectra. GFA oxide was also indicated to exist in human urine. For phase II metabolites, after human urine was treated either with glucuronidase or sulfatase, GFA occured in the chromatograms. It was suggested that there were GFA glucuronide and GFA sulfate in human urine. Further more, positive molecular ions, m/z 606 and m/z 510, of the two conjugates were detected in human urine by LC/MSD. In addition, characteristic ion of m/z 606 was identified as the precursor ion of m/z 177 [Glucuronic acid+H]+ by using MS/MS. Characteristic ion of m/z 430 [GFA+H]+ was also identified as a product ion of m/z 606 [GFA glucuronide+H]+. It was concluded that there were GFI. GFA oxide, GFA glucuronide and GFA sulfate in human urine.


Assuntos
Antiarrítmicos/urina , Compostos Heterocíclicos de 4 ou mais Anéis/urina , Adulto , Biotransformação , Cromatografia Líquida de Alta Pressão , Cromatografia em Camada Fina , Feminino , Glucuronidase/metabolismo , Humanos , Indicadores e Reagentes , Masculino , Espectrometria de Massas , Espectrometria de Massas por Ionização por Electrospray , Espectrofotometria Ultravioleta , Sulfatases/metabolismo
15.
Clin Pharmacol Ther ; 71(6): 457-67, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12087349

RESUMO

BACKGROUND: Cariporide (HOE642) is a recently developed inhibitor of the myocardial sodium-hydrogen exchange system. The clinical effects of sodium-hydrogen exchange inhibition in patients at high risk for myocardial cell necrosis were investigated in the GUARDIAN trial (n = 11,590 patients). Although the trial did not show a significant benefit of cariporide over placebo in the overall population, a 25% relative risk reduction in the primary end point of death or myocardial infarction (12.1% for the highest tested cariporide dose of 120 mg 3 times a day versus 16.2% for placebo; P =.03) was observed in the subpopulation of patients who underwent bypass surgery. OBJECTIVE: Our objective was to identify an optimal dosing regimen that might offer increased protection during the period of highest risk. METHODS: A population pharmacokinetic model of cariporide was developed with use of data from phase I studies. After adequate predictability was shown for the patients in the pharmacokinetic substudy of the GUARDIAN trial (n = 269 patients), the model was used to predict the individual pharmacokinetic profile in the remaining patients. These predicted concentrations were used to calculate the mean concentration during the period of coronary artery bypass graft surgery (the acute risk period in patients who receive coronary artery bypass grafts), and this mean concentration was used as predictor variable in a time-to-event analysis. RESULTS: A mixture of two Weibull functions adequately described the time course of the observed sum of the acute and chronic hazard rate. The calculated mean concentration during the period of surgery was an adequate predictor for the probability of an event in the acute risk period. The estimated minimal effective mean concentration was 0.5 mg/L. CONCLUSIONS: A dosing regimen with a loading dose of an infusion of 120 mg/h for 1 hour followed by an infusion of 20 mg/h for 47 hours should achieve stable exposure above the estimated minimal effective concentration in more than 95% of patients during and after coronary artery bypass graft surgery.


Assuntos
Antiarrítmicos/administração & dosagem , Antiarrítmicos/farmacocinética , Ponte de Artéria Coronária , Morte Súbita Cardíaca/prevenção & controle , Guanidinas/administração & dosagem , Guanidinas/farmacocinética , Infarto do Miocárdio/prevenção & controle , Sulfonas/administração & dosagem , Sulfonas/farmacocinética , Idoso , Antiarrítmicos/sangue , Antiarrítmicos/urina , Ponte de Artéria Coronária/efeitos adversos , Morte Súbita Cardíaca/etiologia , Esquema de Medicação , Feminino , Guanidinas/sangue , Guanidinas/urina , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/etiologia , Sulfonas/sangue , Sulfonas/urina
16.
J Chromatogr B Biomed Sci Appl ; 744(2): 299-306, 2000 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-10993518

RESUMO

An enantioselective high-performance liquid chromatography method was developed for the simultaneous determination of disopyramide (DP) and mono-N-dealkyldisopyramide (MND) enantiomers in plasma and urine. The drugs were extracted from plasma samples by liquid-liquid extraction with dichloromethane after protein precipitation with trichloroacetic acid; the urine samples were processed by liquid-liquid extraction with dichloromethane. The enantiomers were resolved on a Chiralpak AD column using hexane-ethanol (91:9, v/v) plus 0.1% diethylamine as the mobile phase and monitored at 270 nm. Under these conditions the enantiomeric fractions of the drug and of its metabolite were analyzed within 20 min. The extraction procedure was efficient in removing endogenous interferents and low values for the relative standard deviations were demonstrated for both within-day and between-day assays. The method described in this paper allows the determination of DP and MND enantiomers at plasma levels as low as 12.5 ng/ml and can be used in clinical pharmacokinetic studies.


Assuntos
Amilose/química , Antiarrítmicos/farmacocinética , Cromatografia Líquida de Alta Pressão/métodos , Disopiramida/farmacocinética , Antiarrítmicos/sangue , Antiarrítmicos/urina , Disopiramida/sangue , Disopiramida/urina , Humanos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Estereoisomerismo
17.
Clin Pharmacol Ther ; 68(1): 44-57, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10945315

RESUMO

BACKGROUND AND OBJECTIVE: Mexiletine and propafenone are often used concomitantly and are metabolized by the same cytochrome P450 isozymes, namely CYP2D6, CYP1A2, and probably CYP3A4. Our objective was to study the potential pharmacokinetic and electrophysiological interactions between mexiletine and propafenone. METHODS: Fifteen healthy volunteers, 8 extensive metabolizers and 7 poor metabolizers of CYP2D6, received oral doses of mexiletine 100 mg two times daily from day 1 to day 8 and oral doses of propafenone 150 mg two times daily from day 5 to day 12. Interdose studies were performed at steady-state on mexiletine alone (day 4), mexiletine plus propafenone (day 8), and propafenone alone (day 12). RESULTS: In subjects in the extensive metabolizer group, coadministration of propafenone decreased oral clearances of R-(-)-mexiletine (from 41+/-11 L/h to 28+/-7 L/h) and S-(+)-mexiletine (from 43+/-15 L/h to 29+/-11 L/h) to an extent such that these values were no longer different between the extensive and the poor metabolizer groups. Propafenone coadministration also decreased partial metabolic clearances of mexiletine to hydroxymethylmexiletine, p-hydroxymexiletine, and m-hydroxymexiletine in extensive metabolizers by 71%, 67%, and 73%, respectively. In contrast, propafenone did not alter the kinetics of mexiletine enantiomers in subjects in the poor metabolizer group except for a slight decrease in the formation of hydroxymethylmexiletine. Pharmacokinetic parameters of propafenone were not changed during concomitant administration of mexiletine in subjects of either phenotype. Finally, electrocardiographic parameters (QRS duration, QTc, RR, and PR intervals) were not modified during the combined administration of the drugs. CONCLUSION: Propafenone is a potent CYP2D6 inhibitor that may cause an increase in plasma concentrations of coadministered CYP2D6 substrates.


Assuntos
Antiarrítmicos/farmacocinética , Sistema Enzimático do Citocromo P-450/metabolismo , Mexiletina/farmacocinética , Propafenona/farmacocinética , Administração Oral , Adulto , Antiarrítmicos/administração & dosagem , Antiarrítmicos/sangue , Antiarrítmicos/farmacologia , Antiarrítmicos/urina , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A , Esquema de Medicação , Eletrocardiografia/efeitos dos fármacos , Genótipo , Humanos , Masculino , Mexiletina/administração & dosagem , Mexiletina/sangue , Mexiletina/farmacologia , Mexiletina/urina , Oxigenases de Função Mista/metabolismo , Fenótipo , Propafenona/administração & dosagem , Propafenona/sangue , Propafenona/farmacologia , Propafenona/urina , Valores de Referência
18.
Clin Pharmacol Ther ; 67(3): 222-8, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10741624

RESUMO

OBJECTIVE: To investigate the effect of cimetidine and probenecid on the renal clearance of pilsicainide in healthy subjects. METHODS: Nine healthy men (age range, 21 to 38 years) were given oral doses of 50 mg pilsicainide hydrochloride alone, with coadministration of 800 mg oral cimetidine, or with coadministration of 1,500 mg oral probenecid on three occasions in a Latin-square order. Urine and venous blood samples were collected on a timely basis. The concentration of pilsicainide in plasma and urine were determined by an HPLC method. RESULTS: Concomitant administration of cimetidine significantly increased the area under the plasma concentration-time curve of pilsicainide by a mean of 33%, prolonged elimination half-life by a mean of 24% (from 5 to 6.2 hours), reduced apparent oral clearance by a mean of 26% (from 14.7 +/- 0.1 to 10.8 +/- 0.8 L/h) and reduced renal clearance by a mean of 28% (from 196.8 +/- 53.9 to 141.8 +/- 25.9 mL/min). The net renal clearance by tubular secretion was significantly reduced by a mean value of 38%, from 151.4 +/- 62.9 to 93.0 +/- 31.1 mL/min. Coadministration of probenecid did not show any changes in plasma concentrations of pilsicainide, pharmacokinetics, or the net renal clearance by tubular secretion of pilsicainide. CONCLUSIONS: Pilsicainide appeared to be secreted by the active transport system for organic bases in the proximal tubule, and the excretion of pilsicainide was inhibited by cimetidine.


Assuntos
Antiarrítmicos/farmacocinética , Cimetidina/farmacologia , Inibidores Enzimáticos/farmacologia , Rim/metabolismo , Lidocaína/análogos & derivados , Probenecid/farmacologia , Uricosúricos/farmacologia , Adulto , Antiarrítmicos/sangue , Antiarrítmicos/urina , Cromatografia Líquida de Alta Pressão , Estudos Cross-Over , Humanos , Rim/efeitos dos fármacos , Lidocaína/sangue , Lidocaína/farmacocinética , Lidocaína/urina , Masculino , Valores de Referência
19.
Xenobiotica ; 29(9): 945-55, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10548454

RESUMO

1. The metabolism of moricizine.HCl was studied in 12 male volunteers dosed with 250 mg (300 microCi) 14C-radiolabelled drug. 2. Moricizine was biotransformed to many metabolites in humans (at least 35 plasma and 51 urine metabolites). 3. Urine and faecal combined mean (range) recovery accounted for 90.2% (73.4-101.6%) of the administered radioactivity, with most of the recovered radioactivity present in faeces (mean 58.4%; range 45.6-64.7%). Mean (range) urinary recovery was 31.8% (26.2-36.9%), with <1% of the dose recovered as intact moricizine, and no one metabolite accounting for >2.5% of the dose. 4. Total radioactivity (TR) plasma t1/2 was 85.2 h, while that for moricizine was 2.4 h. Mean half-lives for plasma metabolites ranged from 2.9 to 23.6 h. The largest portion (11%) of TR AUC (area under the plasma concentration-time curve) was attributed to 2amino-10-glucuronophenothiazine. Each of the other metabolites accounted for less of the TR AUC than parent drug except for two unidentified peaks which had comparable areas (approximately 5% of the total radioactivity area). 5. Two identified moricizine metabolites, 2-amino-10-(3-morpholinopropionyl) phenothiazine and ethyl [10-(3-aminopropionyl) phenothiazin-2-yl] carbamate, possess the structural characteristics proposed for class 1 anti-arrhythmic activity (pendant amine functionality) and have plasma half-lives 4-7-fold longer than moricizine.


Assuntos
Antiarrítmicos/sangue , Antiarrítmicos/urina , Moricizina/sangue , Moricizina/urina , Adulto , Antiarrítmicos/farmacocinética , Radioisótopos de Carbono , Meia-Vida , Humanos , Masculino , Pessoa de Meia-Idade , Moricizina/farmacocinética , Valores de Referência
20.
Clin Pharmacol Ther ; 65(4): 402-7, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10223777

RESUMO

OBJECTIVE: CYP2D6 mediates both alpha-hydroxylation and O-demethylation of metoprolol. In Chinese subjects, CYP2D6*1 (the wild-type) alleles are relatively uncommon. Subjects with P34S (C188-->T188) and S486T (G4268-->C4268) mutations (CYP2D6J or CYP2D6*10A) are more frequently seen. Recently, the CYP2D6*2 (CYP2D6L) genotype that results in R296C (C2938-->T2938) and S486T mutations was also found important. In this study, metoprolol pharmacokinetics was investigated in subjects of these 3 major genotypes. METHODS: Allele-specific polymerase chain reaction was used to differentiate CYP2D6*1 and CYP2D6*2 alleles from the common CYP2D6*10A allele in Chinese. Subjects with both CYP2D6*1 and CYP2D6*2 have homozygous C188 in the exon 1, whereas subjects with CYP2D6*10A have T188. Metoprolol pharmacokinetics was compared in 16 C188 subjects (6 homozygous CYP2D6*1 subjects and 10 heterozygous CYP2D6*1/CYP2D6*2 subjects), 12 heterozygous C/T188 subjects, and 12 homozygous T188 subjects. RESULTS: No significant difference in plasma concentration profile or urinary alpha-hydroxymetoprolol excretion could be found among subjects with R296C polymorphism (CYP2D6*1/CYP2D6*2). Therefore data from subjects with CYP2D6*1 and CYP2D6*2 were pooled to compare with data from subjects with CTP2D6*10A. The area under plasma concentration curves (AUC) of S-metoprolol was 1411+/-116 (mean +/- SEM, n = 16), 1899+/-120 (n = 12), and 3588+/-435 (n = 12) nmol x hr/L for homozygous C188, heterozygous C/T188, and homozygous T188 subjects, respectively. The urinary recovery of all 4 alpha-hydroxymetoprolol diastereomers was significantly lower in T188 subjects than in C188 subjects. CONCLUSION: The P34S polymorphism but not the R296C polymorphism resulted in higher metoprolol plasma concentrations and lower urinary metoprolol metabolite levels in Chinese subjects. This finding suggests that a lower dose of metoprolol may be used in subjects with T188 mutation (CYP2D6*10A allele).


Assuntos
Antagonistas Adrenérgicos beta/farmacocinética , Antiarrítmicos/farmacocinética , Anti-Hipertensivos/farmacocinética , Povo Asiático/genética , Citocromo P-450 CYP2D6/genética , Metoprolol/farmacocinética , Administração Oral , Antagonistas Adrenérgicos beta/sangue , Antagonistas Adrenérgicos beta/urina , Adulto , Antiarrítmicos/sangue , Antiarrítmicos/urina , Anti-Hipertensivos/sangue , Anti-Hipertensivos/urina , Área Sob a Curva , Genótipo , Humanos , Hidroxilação , Masculino , Metilação , Reação em Cadeia da Polimerase , Polimorfismo Genético , Valores de Referência , Taiwan
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...