Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Med ; 7(4): 471-7, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283675

ABSTRACT

During ischemic stroke, neurons at risk are exposed to pathologically high levels of intracellular calcium (Ca++), initiating a fatal biochemical cascade. To protect these neurons, we have developed openers of large-conductance, Ca++-activated (maxi-K or BK) potassium channels, thereby augmenting an endogenous mechanism for regulating Ca++ entry and membrane potential. The novel fluoro-oxindoles BMS-204352 and racemic compound 1 are potent, effective and uniquely Ca++-sensitive openers of maxi-K channels. In rat models of permanent large-vessel stroke, BMS-204352 provided significant levels of cortical neuroprotection when administered two hours after the onset of occlusion, but had no effects on blood pressure or cerebral blood flow. This novel approach may restrict Ca++ entry in neurons at risk while having minimal side effects.


Subject(s)
Indoles/pharmacology , Potassium Channels, Calcium-Activated , Potassium Channels/drug effects , Stroke/drug therapy , Animals , Brain/metabolism , CHO Cells , Calcium/metabolism , Cell Line , Cricetinae , Disease Models, Animal , Dogs , Glutamic Acid/metabolism , Humans , In Vitro Techniques , Indoles/pharmacokinetics , Indoles/toxicity , Large-Conductance Calcium-Activated Potassium Channels , Male , Patch-Clamp Techniques , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Safety , Stroke/metabolism , Synaptic Transmission/drug effects
4.
J Pineal Res ; 22(1): 45-51, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9062870

ABSTRACT

A review of the literature indicates that the absolute oral bioavailability of exogenous melatonin in humans or in preclinical animal models has not been adequately characterized; hence, this study was undertaken. Pharmacokinetics of melatonin was studied in rats, dogs, and monkeys following intravenous and oral administrations, and the absolute oral bioavailability of melatonin was calculated from the area under the plasma concentration-time curve. The apparent elimination half-life of melatonin following an intravenous dose of 3 mg/kg (5 mg/kg in rats) was 19.8, 18.6, and 34.2 minutes, respectively, in rats, dogs, and monkeys. The dose normalized oral bioavailability of melatonin following a 10 mg/kg oral dose was 53.5% in rats, while it was in excess of 100% in dogs and monkeys. Further, bioavailability of melatonin following a 10 mg/kg intraperitoneal administration in rats was 74.0%, suggesting the lack of substantial first-pass hepatic extraction of melatonin in rats. However, the oral bioavailability of melatonin in dogs decreased to 16.9% following a 1 mg/kg oral dose, indicating dose-dependent bioavailability in dogs. In vitro permeability studies with CACO-2 cells suggest that melatonin is likely to be well absorbed in humans. In vitro metabolism studies with fresh liver slices from rats as well as human donors were conducted to compare the initial rates of metabolism of melatonin between the two species and the results suggest that the intrinsic clearance of melatonin in humans may be lower than that in rats.


Subject(s)
Melatonin/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Chromatography, High Pressure Liquid , Disease Models, Animal , Dogs , Humans , Injections, Intravenous , Liver/chemistry , Liver/drug effects , Macaca fascicularis , Male , Melatonin/administration & dosage , Melatonin/blood , Rats , Rats, Sprague-Dawley , Spectrophotometry, Ultraviolet
5.
Res Commun Mol Pathol Pharmacol ; 89(1): 27-44, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7582859

ABSTRACT

Plasma concentrations of BMS-184111, an anxiolytic, were determined as a function of time following single intravenous, intraperitoneal and oral administrations. In order to assess the brain penetration of this compound, concentrations in whole brain samples were also determined in the intravenous leg of the study. Concentrations of BMS-184111 in plasma and brain homogenate samples were determined using an HPLC assay following liquid/liquid extraction. After intravenous administration, BMS-184111 was eliminated from plasma with a half-life of about 3.6 hours. The brain/plasma AUC ratio for BMS-184111 concentration was 5.5, indicating effective penetration of the compound into the brain. Comparison of the plasma AUC values obtained following intravenous and intraperitoneal doses indicated that BMS-184111 was only 33% bioavailable after intraperitoneal administration, suggesting that the compound undergoes significant first-pass hepatic extraction. The oral bioavailability of BMS-184111 was found to be 10% after administration of the free base and 23% after administration of the hydrochloride salt. These results suggest that BMS-184111 undergoes incomplete GI absorption and/or intestinal metabolism in addition to first-pass hepatic extraction. The in vitro metabolism of BMS-184111 was studied using rat liver homogenate preparation (the 9000 g supernatant; S-9). Several of the metabolites thus generated were profiled using LC/MS and LC/MS/MS. Metabolism of BMS-184111 in rat liver S-9 occurs through hydroxylation, O-demethylation, and demethylenation.


Subject(s)
Anti-Anxiety Agents/pharmacokinetics , Dioxoles/pharmacokinetics , Piperidines/pharmacokinetics , Animals , Anti-Anxiety Agents/analysis , Chromatography, High Pressure Liquid , Dioxoles/analysis , Male , Mass Spectrometry , Piperidines/analysis , Rats , Rats, Sprague-Dawley
7.
J Pharmacol Exp Ther ; 267(1): 425-31, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8229771

ABSTRACT

Labetalol has been previously shown to cause significant maternal and fetal metabolic effects in pregnant sheep after maternal administration. To investigate these observations further, the present study describes the pharmacokinetics, metabolism and pharmacodynamics of labetalol in the fetal lamb after direct fetal i.v. bolus (4 mg) administration. The fetal total body clearance of labetalol (50.45 +/- 1.37 ml m-1 kg-1), which was significantly higher than that previously determined in the ewe, was composed of transplacental and nonplacental CLs of 23.4 +/- 8.99 ml m-1 kg-1 and 27.05 +/- 10.36 ml m-1 kg-1, respectively. The maternal to fetal plasma labetalol area under the curve ratio was 0.031 +/- 0.002 and the CLmp and CLmn were 7.27 +/- 2.11 ml m-1 kg-1 and 30.5 +/- 5.94 ml m-1 kg-1, respectively. Labetalol concentrations in fetal tracheal fluid were consistently higher than that in fetal plasma. The glucuronide conjugate of labetalol was found in the amniotic fluid at up to 20 times the free drug concentration but the oxidative metabolite, 3-amino-1-phenyl-butane, was not detected in plasma or amniotic fluid samples. The fetal effect of labetalol was characterized by an acute lactic acidosis. The calculated hind limb arteriovenous lactate flux showed a net output of lactic acid equal to 3.85 +/- 2.05 g from the hind limb over 24 h after labetalol administration. Although the fetal exposure to labetalol in this study was roughly 4 times that after a 100-mg maternal bolus administration, the magnitude of fetal lactic acidosis was not significantly different in these studies. The clinical implications of the observations made in this study remain to be investigated.


Subject(s)
Labetalol/metabolism , Placenta/metabolism , Animals , Blood Gas Analysis , Female , Hemodynamics/drug effects , Injections, Intravenous , Labetalol/pharmacokinetics , Labetalol/pharmacology , Lactates/blood , Pregnancy , Sheep
8.
J Chromatogr ; 619(1): 79-86, 1993 Sep 08.
Article in English | MEDLINE | ID: mdl-8245166

ABSTRACT

A chiral high-performance liquid chromatographic assay for the separation of the four stereoisomers of labetalol, an antihypertensive, in biological fluids has been developed. Baseline separation of the isomers was achieved using an alpha 1-acid glycoprotein stationary phase. No interference from endogenous substances was observed following extraction from various biological fluids obtained from pregnant (ewe and fetus) and non-pregnant sheep. The concentration of the individual isomers of labetalol was determined by first measuring the total concentration of racemic labetalol obtained from an achiral assay followed by reassay of each sample by the chiral method after which, by using the estimate of the percentage of each individual isomer, the individual concentration of each of the four isomers was determined. The mobile phase was 0.02 M phosphate buffer containing 0.015 M tetrabutylammonium phosphate. The pH of the mobile phase was adjusted to 7.10. The detector was set at an excitation wavelength of 230 nm and emission wavelength of 400 nm to monitor the nascent fluorescence intensity of the isomers of labetalol. The limit of detection of the individual isomers was 0.15 ng (0.6 ng of injected racemic labetalol). The assay was linear over the range 0.6-15.0 ng of labetalol (injected) with the intra- and inter-day mean coefficients of variation being less than 9.0 and 6.0%, respectively. Application of the assay in the study of pharmacokinetics of the stereoisomers of labetalol in sheep following administration of racemic labetalol has been demonstrated.


Subject(s)
Labetalol/isolation & purification , Orosomucoid/chemistry , Animals , Chromatography, High Pressure Liquid , Female , Labetalol/pharmacokinetics , Sheep , Spectrometry, Fluorescence , Stereoisomerism
9.
Drug Metab Dispos ; 21(2): 284-92, 1993.
Article in English | MEDLINE | ID: mdl-8097698

ABSTRACT

Labetalol causes significant maternal and fetal metabolic effects in pregnant sheep (Yeleswaram et al., J. Pharmacol. Exp. Ther. 262, 683-691 (1992)). This study was undertaken to investigate the contribution of skeletal muscles in the development of metabolic acidosis induced by labetalol and to explore the involvement of active metabolite(s) using conscious, chronically instrumented adult nonpregnant ewes. Following a 100 mg iv bolus, the disposition of labetalol was similar to that observed in pregnant sheep. The effects of labetalol included hypotension, reflex tachycardia, a significant increase in femoral blood flow, hyperglycemia, lactic acidosis, and increased hind limb oxygen consumption. The arteriovenous flux of labetalol, glucose, lactate, and oxygen across the hindlimb was calculated using the Fick principle. The net output of lactate from the hindquarter over 12 hr following drug administration was calculated to be 6.25 +/- 1.35 g (0.07 +/- 0.015 mol). Glucuronidation, sulfation, and oxidative metabolism of labetalol were studied using urine and bile samples. The cumulative urinary excretion of labetalol as unchanged drug, glucuronide and sulfate was found to be 1.61 +/- 0.3, 11.46 +/- 2.83, and 1.47 +/- 0.74% of the dose, respectively. Using GC-mass selective detection, the presence of 3-amino-1-phenylbutane (3-APB), a close congener of amphetamine, in urine and bile samples was established. The cumulative excretion of 3-APB in urine represents 0.044 +/- 0.016% of the dose. Pharmacokinetic analysis shows the apparent elimination half-life of the metabolite to be 13.5 +/- 3.8 min. Conjugates of 3-APB were also found in the bile and urine.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Labetalol/pharmacokinetics , Animals , Blood Gas Analysis , Blood Glucose/metabolism , Blood Pressure/drug effects , Butylamines/pharmacokinetics , Female , Glucuronates/metabolism , Heart Rate/drug effects , Labetalol/pharmacology , Lactates/metabolism , Lactic Acid , Muscles/metabolism , Oxygen Consumption/drug effects , Regional Blood Flow/drug effects , Sheep , Sulfates/metabolism , Tissue Distribution
10.
Biol Mass Spectrom ; 21(11): 534-40, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1457468

ABSTRACT

A sensitive and selective assay has been developed for the identification and quantitation of 3-amino-1-phenyl butane (3-APB), a metabolite of labetalol, in biological fluids using electron impact gas chromatography/mass-selective detection. Samples were extracted with n-hexane, derivatized with heptafluorobutyric anhydride and chromatographed on a cross-linked fused-silica capillary column. A positive EI spectrum was obtained using a mass-selective detector. Identification of the metabolite was accomplished using an authentic standard; quantitation was performed in the selected ion monitoring mode using ions m/z 345 (M+) and 132. The assay was linear over the calibration range of 0.5-1000 ng of the analyte and the intra-sample coefficients of variation were less than 12% in all cases. The absolute recovery of 3-APB following extraction from urine and bile was found to be 102.9 +/- 4.9% and 98.3 +/- 1.45% (mean +/- SEM) respectively. The minimum quantitation limit of the assay was 0.5 ng ml-1 (approximately 2 pg injected). Application of the assay in a pharmacokinetic-pharmacodynamic study of labetalol in sheep is demonstrated. The metabolite was detected in urine and bile samples obtained from adult non-pregnant sheep following labetalol administration. The cumulative amount of 3-APB excreted in urine over 24 h was found to be 71.55 micrograms in one animal following a 100 mg dose of labetalol. Evidence for biliary excretion, glucuronidation and sulfation of 3-APB was also found.


Subject(s)
Butylamines/analysis , Labetalol/metabolism , Animals , Bile/chemistry , Butylamines/urine , Female , Gas Chromatography-Mass Spectrometry/methods , Labetalol/pharmacokinetics , Sheep
11.
J Pharmacol Exp Ther ; 262(2): 683-91, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1501116

ABSTRACT

The maternal-fetal disposition of labetalol, a combined alpha-1 and beta adrenergic blocker, and its pharmacodynamics in pregnancy are not well understood. This study describes the pharmacokinetics, cardiovascular and metabolic effects of labetalol in the mother and in utero fetus after a 100-mg maternal i.v. bolus administration, in the chronically instrumented pregnant sheep. Labetalol shows a triexponential decline in the mother with a total body clearance of 30.8 +/- 3.83 ml/min/kg, an apparent steady-state volume of distribution (nonparametric) of 3.02 +/- 0.18 liters/kg and terminal elimination half-life of 2.79 +/- 0.66 hr. These estimates are similar to the reported values in pregnant women. Labetalol rapidly crosses the sheep placenta. The peak fetal plasma concentration was 33.7 +/- 5.8 ng/ml, the fetal exposure to labetalol as calculated by the fetal to maternal area under the curve ratio was 14.37 +/- 1.54% and the apparent fetal elimination half-life was 3.71 +/- 0.5 hr. Labetalol persists in the amniotic and fetal tracheal fluids up to 24 hr with concentrations reaching 2- to 4 times the fetal plasma concentration. Whereas there were no significant maternal or fetal cardiovascular effects, some very significant metabolic effects were observed, including fetal and maternal lactic acidosis and hyperglycemia. Lactic acid accumulates in the fetal blood and amniotic fluid with peak concentrations (6.0 +/- 0.31 and 5.5 +/- 0.26 mM, respectively) showing a more than 300% increase over control values. The exact mechanism by which labetalol causes these metabolic effects is not clear, but it may involve its partial beta-2 agonist activity.


Subject(s)
Labetalol/pharmacology , Labetalol/pharmacokinetics , Pregnancy, Animal/metabolism , Animals , Blood Glucose/analysis , Blood Pressure/drug effects , Carbon Dioxide/blood , Female , Lactates/blood , Lactic Acid , Oxygen/blood , Pregnancy , Sheep
12.
J Chromatogr ; 565(1-2): 383-90, 1991 Apr 19.
Article in English | MEDLINE | ID: mdl-1874882

ABSTRACT

A rapid and sensitive microbore high-performance liquid chromatographic (HPLC) assay is reported for the quantitation of labetalol, an anti-hypertensive agent, in small volumes (250 microliters) of biological fluids (viz., maternal plasma, fetal plasma, amniotic fluid and fetal tracheal fluid) obtained from the chronically instrumented pregnant sheep. Labetalol was extracted from the samples using ethyl acetate and then partitioned into dilute phosphoric acid. Chromatography was performed on a microbore HPLC system using a 2.1 mm I.D. C18 column and detection was accomplished by a low-dispersion fluorescence detector designed for trace analysis. The drug was well separated from endogenous substances in all biological fluids sampled. The calibration curves were linear for all fluids over the range of study with mean coefficients of variation consistently below 5%. Quantitation was possible down to approximately 30 pg of labetalol injected (approximately 1.6 ng/ml in plasma using 250 microliters).


Subject(s)
Amniotic Fluid/chemistry , Chromatography, High Pressure Liquid/methods , Fetal Blood/chemistry , Labetalol/analysis , Sheep/metabolism , Animals , Chromatography, High Pressure Liquid/instrumentation , Female , Fetus/metabolism , Injections, Intravenous , Labetalol/administration & dosage , Labetalol/pharmacokinetics , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...