Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Epilepsy Res ; 40(1): 41-52, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10771257

ABSTRACT

The purpose of this study was to assess the concentration-anti-convulsant effect relationships of a number of anti-convulsant drugs in the direct cortical stimulation model, to obtain more insight in the properties and predictive value of this model. The time course of the effect of lamotrigine, loreclezole, flunarizine, CGP40116 and CGP39551 was determined after iv. administration in conjunction with their pharmacokinetics. Convulsive activity was induced by stimulation of the motor cortex with a ramp-shaped pulse train. This technique allows consecutive measurements of the treshold for localized (TLS) and for generalized (TGS) seizure activity. Increase in threshold was used as measure of the anti-convulsant effect. After administration of lamotrigine, pronounced elevation of the TGS, with little change in the TLS, was observed. Flunarizine caused a similar effect, but much less intense. Loreclezole strongly elevated the TGS and to a lesser extent the TLS, also. The concentration-anti-convulsant effect relationship of the three compounds could be fitted by an exponential model. The NMDA antagonists, CGP40116 and CGP39551, induced minor changes in the TLS and a slight increase in the TGS. The onset of this effect was marked by a delay relative to blood concentrations. The biophase equilibration kinetics was estimated and a linear model was applied to describe the concentration-effect relationship of both NMDA antagonists. The present results show that the cortical stimulation model is a suitable technique for integrated pharmacokinetic-pharmacodynamic modelling and for assessing anti-convulsant efficacy. The results show that the model is rather insensitive to calcium channel blockers and NMDA antagonists.


Subject(s)
Anticonvulsants/pharmacology , Anticonvulsants/pharmacokinetics , Motor Cortex/physiology , Animals , Dose-Response Relationship, Drug , Drug Evaluation , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacology , Female , Injections, Intravenous , Osmolar Concentration , Rats , Rats, Wistar , Seizures/etiology , Seizures/physiopathology
2.
Braz J Med Biol Res ; 33(2): 199-204, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10657059

ABSTRACT

R,S-sotalol, a ss-blocker drug with class III antiarrhythmic properties, is prescribed to patients with ventricular, atrial and supraventricular arrhythmias. A simple and sensitive method based on HPLC-fluorescence is described for the quantification of R,S-sotalol racemate in 500 microl of plasma. R,S-sotalol and its internal standard (atenolol) were eluted after 5.9 and 8.5 min, respectively, from a 4-micron C18 reverse-phase column using a mobile phase consisting of 80 mM KH2PO4, pH 4.6, and acetonitrile (95:5, v/v) at a flow rate of 0.5 ml/min with detection at lambdaex = 235 nm and lambdaem = 310 nm, respectively. This method, validated on the basis of R,S-sotalol measurements in spiked blank plasma, presented 20 ng/ml sensitivity, 20-10,000 ng/ml linearity, and 2.9 and 4.8% intra- and interassay precision, respectively. Plasma sotalol concentrations were determined by applying this method to investigate five high-risk patients with atrial fibrillation admitted to the Emergency Service of the Medical School Hospital, who received sotalol, 160 mg po, as loading dose. Blood samples were collected from a peripheral vein at zero, 0.5, 1.0, 1.5, 2.0, 3.0, 4. 0, 6.0, 8.0, 12.0 and 24.0 h after drug administration. A two-compartment open model was applied. Data obtained, expressed as mean, were: C MAX = 1230 ng/ml, T MAX = 1.8 h, AUC T = 10645 ng h-1 ml-1, Kab = 1.23 h-1, alpha = 0.95 h-1, ss = 0.09 h-1, t((1/2))ss = 7.8 h, ClT/F = 3.94 ml min-1 kg-1, and Vd/F = 2.53 l/kg. A good systemic availability and a fast absorption were obtained. Drug distribution was reduced to the same extent in terms of total body clearance when patients and healthy volunteers were compared, and consequently elimination half-life remained unchanged. Thus, the method described in the present study is useful for therapeutic drug monitoring purposes, pharmacokinetic investigation and pharmacokinetic-pharmacodynamic sotalol studies in patients with tachyarrhythmias.


Subject(s)
Anti-Arrhythmia Agents/blood , Atrial Fibrillation/blood , Chromatography, High Pressure Liquid/methods , Sotalol/blood , Anti-Arrhythmia Agents/pharmacokinetics , Anti-Arrhythmia Agents/therapeutic use , Area Under Curve , Atrial Fibrillation/drug therapy , Female , Humans , Male , Middle Aged , Risk Factors , Sotalol/pharmacokinetics , Sotalol/therapeutic use
3.
Br J Pharmacol ; 125(5): 997-1004, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9846637

ABSTRACT

1. In this study we used cortical stimulation to assess the effects of phenytoin (PHT), sodium valproate (VPA), and their interaction on total motor seizure and on the constituent elements of the seizure. 2. PHT (40 mg kg(-1)) was administered as an intravenous bolus infusion to animals receiving either a continuous infusion of VPA or saline. VPA plasma concentration was maintained at levels that produced no detectable anticonvulsant effect. 3. Analysis of ictal components (eyes closure, jerk, gasp, forelimb, clonus, and hindlimb tonus) and their durations revealed both qualitative and quantitative differences in drug effects. 4. The anticonvulsant effect is represented by the increase in the duration of the stimulation required to reach a given seizure threshold. PHT significantly increased the duration of the stimulation and of the motor seizure. This increase was greatly enhanced by VPA. In addition, ictal component analysis revealed that the combination of PHT and VPA causes the reduction of a specific seizure component (JERK). 5. Neither the free fraction of PHT nor the biophase equilibration kinetics changes in the presence of VPA. It is concluded that the synergism may be due to a pharmacodynamic rather than a pharmacokinetic interaction.


Subject(s)
Anticonvulsants/therapeutic use , Phenytoin/therapeutic use , Seizures/prevention & control , Valproic Acid/therapeutic use , Analysis of Variance , Animals , Anticonvulsants/pharmacokinetics , Drug Interactions , Male , Phenytoin/pharmacokinetics , Rats , Rats, Wistar , Seizures/metabolism , Valproic Acid/pharmacokinetics
4.
Brain Res ; 814(1-2): 199-208, 1998 Dec 14.
Article in English | MEDLINE | ID: mdl-9838114

ABSTRACT

Individual variability and numerous interactions between pharmacokinetics, pharmacodynamics, and homeostatic factors complicate the study of the anticonvulsant effect in animal models of seizure activity. In theory, both individual variability and the contribution of these factors to the anticonvulsant effect can be determined by following the time course of the pharmacological response and the corresponding plasma concentrations in individual animals. Currently, there are several formal pharmacokinetic-pharmacodynamic models available for the analysis of such data, which yield accurate estimates of drug intrinsic activity and potency. However, most models of seizure activity are not suited for such an approach, either because they can be applied only once, or because the expression of seizures is not constant over time. In addition, the induction of seizures constitutes repeated jeopardy to the animals, which may profoundly change behavior and interfere in the anticonvulsant response as well as in different physiological processes. In this paper, we compare ictal, post-ictal, and interictal behavior in three different models of seizure activity in rats, namely, the electroconvulsive shock, amygdala kindling and the cortical stimulation model (CSM). The methods were compared in the same way as they are currently in use for the assessment of antiepileptic drug effect. Our results show that repeated seizure activity induced by cortical stimulation does not exacerbate ictal activity (eye closure, jerk, gasp, forelimb clonus, and hind-limb tonus) nor post-ictal behavior (chewing and freezing), while producing less serious changes in interictal behavior (walk, lean, upright rearing, exploratory, grooming, and rest) than kindling or electroconvulsive shock. We conclude that seizures induced by cortical stimulation are reproducible and qualitatively similar to kindling seizures. Our results also suggest that the electroconvulsive shock model is not suited for pharmacokinetic-pharmacodynamic studies and that the assessment of interictal behavior may contribute to the evaluation of overall antiepileptic drug effect in seizure disorders.


Subject(s)
Anticonvulsants/therapeutic use , Kindling, Neurologic , Seizures/etiology , Analysis of Variance , Animals , Disease Models, Animal , Electroshock , Male , Rats , Rats, Wistar , Seizures/drug therapy , Video Recording
5.
J Pharmacol Exp Ther ; 284(2): 460-6, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9454785

ABSTRACT

In this study a pharmacokinetic-pharmacodynamic model is proposed for drugs with nonlinear elimination kinetics. We applied such an integrated approach to characterize the pharmacokinetic-pharmacodynamic relationship of phenytoin. In parallel, the anticonvulsant effect and the electroencephalogram (EEG) effect were used to determine the pharmacodynamics. Male Wistar-derived rats received a single intravenous dose of 40 mg . kg-1 phenytoin. The increase in the threshold for generalized seizure activity (TGS) was used as the anticonvulsant effect and the increase in the total number of waves in the 11.5 to 30 Hz frequency band was taken as the EEG effect measure. Phenytoin pharmacokinetics was described by a saturation kinetics model with Michaelis-Menten elimination. Vmax and Km values were, respectively, 386 +/- 31 microg . min-1 and 15.4 +/- 2.2 microg . ml-1 for the anticonvulsant effect in the cortical stimulation model and 272 +/- 31 microg . min-1 and 5.9 +/- 0.7 microg . ml-1 for the EEG effect. In both groups, a delay to the onset of the effect was observed relative to plasma concentrations. The relationship between phenytoin plasma concentrations and effect site was estimated by an equilibration kinetics routine, yielding mean ke0 values of 0.108 and 0.077 min-1 for the anticonvulsant and EEG effects, respectively. The EEG changes in the total number of waves could be fitted by the sigmoid Emax model, but Emax values could not be estimated for the nonlinear relationship between concentration and the increase in TGS. An exponential equation (E = E0 + Bn . Cn) derived from the sigmoid Emax model was applied to describe the concentration-anticonvulsant effect relationship, under the assumption that Emax values cannot be reached within acceptable electric stimulation levels. This approach yielded a coefficient (B) of 2.0 +/- 0.4 microA . ml . microg-1 and an exponent (n) of 2.7 +/- 0.9. The derived EC50 value of 12.5 +/- 1. 3 microg . ml-1 for the EEG effect coincides with the "therapeutic range" in humans.


Subject(s)
Anticonvulsants/pharmacology , Phenytoin/pharmacology , Animals , Anticonvulsants/pharmacokinetics , Blood Proteins/metabolism , Dose-Response Relationship, Drug , Electroencephalography , Male , Models, Biological , Phenytoin/blood , Phenytoin/pharmacokinetics , Protein Binding , Rats , Rats, Wistar
6.
Br J Pharmacol ; 125(7): 1610-6, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9884091

ABSTRACT

Treatment of epilepsy with a combination of antiepileptic drugs remains the therapeutic choice when monotherapy fails. In this study, we apply pharmacokinetic-pharmacodynamic modelling to characterize the interaction between phenytoin (PHT) and sodium valproate (VPA). Male Wistar rats received a 40 mg kg(-1) intravenous dose of PHT over 5 min either alone or in combination with an infusion of VPA resulting in a steady-state concentration of 115.5+/-4.9 microg ml(-1). A control group received only the infusion of VPA. The increase in the threshold for generalized seizure activity (ATGS) was used as measure of the anticonvulsant effect. PHT pharmacokinetics was described by a pharmacokinetic model with Michaelis-Menten elimination. The concentration-time course and plasma protein binding of PHT were not altered by VPA. The pharmacokinetic parameters Vmax and Km were, respectively, 294+/-63 microg min(-1) and 7.8+/-2.4 microg ml(-1) in the absence of VPA and 562+/-40 microg min(-1) and 15.6+/-0.9 microg ml(-1) upon administration in combination with VPA. A delay of the onset of the effect relative to plasma concentrations of PHT was observed. The assessment of PHT concentrations at the effect site was based on the effect-compartment model, yielding mean ke0 values of 0.128 and 0.107 min(-1) in the presence and absence of VPA, respectively. A nonlinear relationship between effect-site concentration and the increase in the TGS was observed. The concentration that causes an increase of 50% in the baseline TGS (EC50%TGS) was used to compare drug potency. A shift of EC50%TGS from 13.27+3.55 to 4.32+/-0.52 microg ml(-1) was observed upon combination with VPA (P<0.01). It is concluded that there is a synergistic pharmacodynamic interaction between PHT and VPA in vivo.


Subject(s)
Anticonvulsants/pharmacology , Phenytoin/pharmacology , Valproic Acid/pharmacology , Animals , Anticonvulsants/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Male , Phenytoin/pharmacokinetics , Phenytoin/therapeutic use , Rats , Rats, Wistar , Valproic Acid/therapeutic use
7.
Neurosci Biobehav Rev ; 23(2): 229-36, 1998.
Article in English | MEDLINE | ID: mdl-9884115

ABSTRACT

Drug concentrations at the site of action in studies on behavioural pharmacology, are seldom constant. Therefore, observed changes in behaviour can be due to the natural time course of behavioural processes, but equally to changes in drug concentration, and it is therefore crucial to separate the former from the latter. One solution is keeping drug concentrations constant. However, one can also exploit the variation in drug concentration caused by absorption, distribution and elimination of a drug. This is done by simultaneous measurement of drug effect and concentration, while the drug enters and leaves a biologically relevant compartment, such as blood or cerebrospinal fluid. The concept of determining concentration-effect curves in individual animals, by monitoring in parallel drug effect and changes in concentration in one single experiment, has not yet found wide application in behavioural studies. The fact that behavioural processes, like any other physiological process, change over time, may have contributed to the scarcity of pharmacokinetic-pharmacodynamic (PK/PD) studies in behavioural pharmacology. However, there are now mathematical techniques that allow PK/PD modelling even if the effect parameter changes over time or cannot be properly assessed in every instance. Here we use PK/PD modelling to characterize fear-induced ultrasonic vocalizations and the anxiolytic effect of buspirone. This approach reduces the number of animals required to assess concentration-effect relationships. More importantly, it allows the identification of differences in individual drug response over a wide range of concentrations. Consequently, we suggest that PK/PD modelling can be used as a tool to study drug-induced changes in behavioural response. An introduction in PK/PD modelling is presented.


Subject(s)
Behavior, Animal/drug effects , Models, Psychological , Pharmacokinetics , Animals , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/pharmacology , Mice , Rats , Vocalization, Animal/drug effects
8.
Brain Res ; 770(1-2): 221-7, 1997 Oct 03.
Article in English | MEDLINE | ID: mdl-9372222

ABSTRACT

A large number of animal models has been proposed for the evaluation of the anticonvulsant effect of antiepileptic drugs. Various seizure patterns are produced and differences are frequently observed in anticonvulsant effect estimates obtained for the same drug in different models. The incidence of seizures and the threshold for the induction are usually the only measures used for the determination of the anticonvulsant effect. However, behavioural components expressed during seizures induced by different means are likely to differ considerably. The aim of this study was to provide a detailed behavioural description of ictal and post-ictal components in two models of electrically induced seizure activity: kindling and cortical stimulation model (CSM). Seizure activity was induced in two groups of 6 Wistar-derived rats. Ictal and post-ictal behaviours were recorded on video tape and quantified using a computer supported frame-by-frame encoding of the behavioural components. We encoded the duration and rate of occurrence of the following behavioural items: whisker movements, eye closure, myoclonic jerk, facial gasping, forelimb clonus, forelimb tonus, hindlimb tonus, immobility and chewing. It appears that both models are, in many respects, qualitatively similar. However, the models differ quantitatively. Behavioural expression of seizure activity differs in the following respects: (1) the total duration of the seizure induced by cortical stimulation is shorter than by kindling; (2) seizure activity in the CSM occurs mainly during stimulation, while in amygdala kindling, it occurs thereafter; and (3) seizures evoked in the CSM comprise relatively less violent behavioural items than in the amygdala kindling. The evaluation of the ictal and post-ictal behavioural components suggests that behavioural analysis could assist in the detection of differences in the mechanisms of action of antiepileptic drugs. In addition, observational measures can also be used to assess animal distress inflicted by different experimental procedures.


Subject(s)
Cerebral Cortex/physiopathology , Epilepsy/physiopathology , Kindling, Neurologic/physiology , Animals , Behavior, Animal/physiology , Disease Models, Animal , Electric Stimulation , Male , Movement/physiology , Rats , Rats, Wistar
9.
Eur J Clin Pharmacol ; 48(1): 45-9, 1995.
Article in English | MEDLINE | ID: mdl-7621847

ABSTRACT

We studied the use of atrioventricular (AV) conduction time to assess the therapeutic equivalence of two diltiazem formulations in 20 volunteers in a double-blind, cross-over trial. ECG recording was carried out before and at several intervals after drug administration, and prolongation of the PQ interval (delta PQ) was taken as a pharmacodynamic response. In addition, diltiazem plasma concentrations were determined in 8 subjects. The effect of diltiazem increased proportionally with the plasma concentration and could be detected up to 10 h after administration. The area under the effect-time curve (AUEC(0-10)), the peak effect (Emax), and the effect mean residence time (MRTE) showed significant differences. In contrast to the pharmacodynamics, the pharmacokinetic profiles of diltiazem do not vary to the same extent. We conclude that the formulations are therapeutically different. Furthermore, at the administered dose, delta PQ appears to be a sensitive measure for assessing the electrophysiological properties of diltiazem.


Subject(s)
Diltiazem/pharmacokinetics , Electrocardiography/drug effects , Heart Rate/drug effects , Adult , Atrioventricular Node/drug effects , Cross-Over Studies , Delayed-Action Preparations , Diltiazem/administration & dosage , Diltiazem/therapeutic use , Double-Blind Method , Heart Conduction System/drug effects , Humans , Male , Therapeutic Equivalency
SELECTION OF CITATIONS
SEARCH DETAIL
...