Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur J Drug Metab Pharmacokinet ; 49(1): 7-21, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37921950

ABSTRACT

BACKGROUND AND OBJECTIVE: Cenobamate is an antiseizure medication (ASM) approved for treatment of focal epilepsy in adults. The objective of this study was to characterize the distribution, metabolism, and excretion of cenobamate in adult and pre- and postnatal rats, including pregnant and lactating females and nursing pups. METHODS: Distribution, metabolic, and excretion profiles were determined for 14C-labeled and unlabeled cenobamate using liquid scintillation counting, radiochromatography, LCMS, and LCMS/MS after oral or intravenous (IV) administration. RESULTS: Distribution of 14C-cenobamate-related material in adult male rats was widespread throughout the body, with nearly 1:1 tissue-to-plasma ratios observed for most tissues, including brain. Cenobamate administered to pregnant females was also transferred across the placental barrier into amniotic fluid and fetal plasma. Following administration to lactating F0 females, cenobamate was detected in breast milk and in plasma of nursing pups. 14C-cenobamate administered to adult male rats as a single oral dose was extensively metabolized with nine metabolites identified in urine and feces, including a principal dihydrodiol metabolite. Cenobamate was the principal drug-related material in rat plasma. Following a single dose of 14C-cenobamate to male and female rats, radioactivity was excreted equally into urine and feces, with mass balance achieved by 48 h postdose. CONCLUSIONS: Distribution of cenobamate was widespread into many rat tissues, including brain, amniotic fluid, fetal plasma, breast milk, and breastfeeding rat pups. These distribution findings, along with the results of the metabolism and excretion studies, may help inform treatment decisions for patients with epilepsy being treated with cenobamate, including pregnant or nursing mothers.


Subject(s)
Carbamates , Chlorophenols , Lactation , Placenta , Tetrazoles , Humans , Rats , Male , Animals , Pregnancy , Female , Placenta/metabolism , Milk/metabolism , Feces/chemistry
2.
Heliyon ; 9(8): e18920, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37636350

ABSTRACT

Background: Cenobamate is an antiseizure medication used to treat partial-onset (focal) seizures. It is a molecule with one chiral center and a unique dual mechanism of action: enhancement of fast and slow inactivation of sodium channels with preferential inhibition of the persistent current and positive allosteric modulation of GABAA receptor-mediated ion channels. Aims/Methods: Anticonvulsant effects of cenobamate (YKP3089; R-enantiomer), YKP3090 (S-enantiomer), and YKP1983 (racemate) were evaluated in chemically and electrically induced focal and generalized seizure models in rodents. The Genetic Absence Epilepsy Rat from Strasbourg (GAERS) model examined the effect of cenobamate on spike-wave seizures. Motor coordination was assessed with rotarod tests and minimal motor impairment exams. Results: Early in development, cenobamate was found to have activity in focal and generalized seizure models in animals and was selected for continued development. Cenobamate prevented seizures in a dose-dependent manner, prevented seizure spread, and increased seizure threshold without potentiating seizure initiation or the development of tolerance to its anticonvulsant effects. In contrast, YKP3090 and YKP1983 were only effective against generalized tonic-clonic seizures. Cenobamate also protected mice from 6 Hz psychomotor-induced seizures. Cenobamate showed significant dose-dependent reductions in the number and cumulative duration of spike-and-wave discharges in the GAERS model. Discussion: Cenobamate showed efficacy or efficacy signals in all animal models of epilepsy tested with a favorable risk-versus-benefit ratio, supporting its clinical use in the treatment of partial-onset (focal) seizures in adults and warranting further clinical research in generalized seizures and absence seizures.

3.
Eur J Pharmacol ; 879: 173117, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32325146

ABSTRACT

Cenobamate is a novel antiepileptic drug under investigation for use in patients with focal (partial-onset) seizures. To understand its potential molecular mechanism of action, the effects of cenobamate on GABAA-mediated currents and GABAA receptors in rodent hippocampal neurons were examined. Cenobamate potentiated GABA-induced currents (IGABA) in acutely isolated CA3 pyramidal cells in a concentration-dependent manner (EC50, 164 µM), which was not affected by flumazenil, a benzodiazepine receptor antagonist. Cenobamate enhanced tonic GABAA currents (Itonic), which is defined as a holding current shift by the GABAA receptor antagonist bicuculline (EC50, 36.63 µM). At therapeutically relevant concentrations, cenobamate induced minimal changes in the frequency, amplitudes, and decay time of spontaneous inhibitory postsynaptic currents in the CA1 neurons. Flumazenil failed to affect cenobamate-potentiated Itonic and Iphasic in CA1 neurons. Cenobamate showed positive allosteric modulation of GABA-induced IGABA mediated by GABAA receptors. This effect was similar for all tested hGABAA receptors containing six different alpha subunits (α1ß2γ2 or α2-6ß3γ2), with EC50 values ranging from 42 to 194 µM. Cenobamate did not displace the binding of flunitrazepam, a benzodiazepine derivative, or flumazenil to GABAA receptors. The results showed that cenobamate, a novel antiepileptic drug, acts as a positive allosteric modulator of high-affinity GABAA receptors, activated by GABA at a site independent of the benzodiazepine binding site and efficiently enhances Itonic inhibition in hippocampal neurons, which could be an underlying molecular mechanism stabilizing neural circuits of the epileptic hippocampus.


Subject(s)
Anticonvulsants/pharmacology , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/drug effects , Carbamates/pharmacology , Chlorophenols/pharmacology , Neurons/drug effects , Receptors, GABA-A/physiology , Tetrazoles/pharmacology , Animals , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Female , HEK293 Cells , Humans , Male , Neurons/physiology , Rats, Sprague-Dawley , Rats, Wistar
4.
Eur J Drug Metab Pharmacokinet ; 45(4): 513-522, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32301064

ABSTRACT

BACKGROUND AND OBJECTIVE: Cenobamate is an antiepileptic drug for the treatment of partial-onset seizures. The current study was designed to assess the mass balance and the metabolic profiling of cenobamate in humans. METHODS: Absorption, metabolism, and excretion of cenobamate were investigated in healthy male subjects after a single oral dose of 400 mg of cenobamate containing 50 µCi of [14C]-cenobamate as capsule formulation. RESULTS: Cenobamate was rapidly (median time to maximum plasma concentration of 1.25 h) and extensively (≥ 88% of dose) absorbed. The mean cenobamate plasma concentration-time profile revealed a multiphasic elimination profile whereas the mean plasma/blood concentration-time curve for total radioactivity did not appear to be multiphasic, suggesting that elimination mechanisms for cenobamate and its metabolites may be different. Blood/plasma ratios observed for the area under the concentration-time curve (AUC) and peak concentration (both ~ 0.60) suggest a limited penetration of cenobamate and metabolites into red blood cells (RBCs). Eight cenobamate metabolites were identified across plasma, urine, and feces. Cenobamate was the main plasma radioactive component and M1 was the only metabolite detected in plasma (> 98% and < 2% total radioactivity AUC, respectively). All detected metabolites were found in urine, with M1 as the major radioactive component (mean cumulative recovery 37.7% of dose); unchanged cenobamate accounted for 6%. Metabolites comprised ~ 88% of the dose recovered in urine, indicating extensive metabolism by the kidneys and/or metabolites formed in the liver were rapidly eliminated from the bloodstream. However, cenobamate metabolites appear to be formed slowly. Minor amounts of cenobamate (0.48%) and five metabolites (≤ 1.75% each; M1, M3, M6, M7, M11) were recovered in feces. CONCLUSION: This study indicates that cenobamate is primarily eliminated in urine as metabolites. Cenobamate is the major circulating component in plasma after oral administration and has a limited penetration into RBCs.


Subject(s)
Anticonvulsants/administration & dosage , Anticonvulsants/pharmacokinetics , Carbamates/administration & dosage , Carbamates/pharmacokinetics , Chlorophenols/administration & dosage , Chlorophenols/pharmacokinetics , Renal Elimination , Tetrazoles/administration & dosage , Tetrazoles/pharmacokinetics , Administration, Oral , Adult , Anticonvulsants/blood , Biotransformation , Carbamates/blood , Chlorophenols/blood , Gastrointestinal Absorption , Healthy Volunteers , Humans , Intestinal Elimination , Male , Metabolomics , Middle Aged , New Jersey , Tetrazoles/blood , Young Adult
5.
J Neurosci Methods ; 150(2): 143-9, 2006 Jan 30.
Article in English | MEDLINE | ID: mdl-16430967

ABSTRACT

In the traditional [14C] deoxyglucose (2DG) method for the measurement of local cerebral glucose utilization (LCGU), blood samples are collected from the femoral artery. However, the placement of a femoral catheter can affect locomotor activity of the animal. We wanted to develop a new technique for blood sampling that would not interfere with the ongoing behavior. Therefore, the present report establishes a method of collecting blood samples for the 2DG method through the jugular vein. To calibrate this method, catheters were inserted in both the femoral artery and jugular vein of adult male Sprague Dawley rats. The next day, rats were injected with 2DG (125 microCi/kg) through the jugular vein. To quantify 14C in plasma, the standard method of blood collection was used for the femoral artery while syringes were used to extract blood samples from the jugular vein. We calculated the integrated specific activity of the plasma and final tissue 2DG concentrations based on Sokoloff's original equation using blood samples derived from both vessels. LCGU determined in selected brain regions was equivalent using both sampling methods. In conclusion, sampling from the jugular vein is appropriate for the quantified 2DG method and does not disrupt locomotor activity of the rat.


Subject(s)
Brain/metabolism , Deoxyglucose/metabolism , Hematologic Tests/methods , Jugular Veins , Animals , Carbon Isotopes , Catheters, Indwelling , Deoxyglucose/blood , Femoral Artery , Male , Motor Activity , Rats , Rats, Sprague-Dawley
6.
Neurotoxicol Teratol ; 27(4): 599-608, 2005.
Article in English | MEDLINE | ID: mdl-15994058

ABSTRACT

The present study was designed to assess the dopaminergic and serotonergic contributions of the acoustic startle response (ASR) and the tactile startle response (TSR) in adult rats that had been perinatally exposed to AZT (azidothymidine, zidovudine; an antiretroviral agent). Each dam was randomly assigned to a treatment group: non-treated, AZT0, 100 or 150 mg/kg. Once daily gastric intubation began prenatally on gestational day (G) 19 and continued to G22 and then the pups were intubated between postnatal day (PND) 2-20. On PND60, animals were tested for responses to both acoustic and tactile stimuli following a challenge of vehicle, 0.25 or 0.5 mg/kg 8-OH-DPAT, a 5-HT(1A) agonist, or 0.75 or 2.0 mg/kg apomorphine (APO, a dopaminergic agonist) IP. Both DPAT and APO increased startle magnitude as expected. Additionally, perinatal AZT exposure enhanced startle responses following both DPAT and APO, an effect not due to perinatal handling or intubation. Similarly, perinatal AZT increased tactile responses following drug challenge in a gender-specific manner. Perinatal AZT also prolonged startle latencies, a change which may indicate that perinatal AZT alters conduction velocity. Therefore, the administration of AZT during the perinatal period results in long-term functional alterations within the startle reflex pathways.


Subject(s)
8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Apomorphine/pharmacology , Dopamine Agonists/pharmacology , Prenatal Exposure Delayed Effects , Reflex, Startle/drug effects , Serotonin Receptor Agonists/pharmacology , Zidovudine/toxicity , Analysis of Variance , Animals , Anti-HIV Agents/toxicity , Dose-Response Relationship, Drug , Drug Interactions , Female , Male , Physical Stimulation/methods , Pregnancy , Rats , Reflex, Startle/physiology , Sex Factors
7.
Brain Res Dev Brain Res ; 153(1): 127-34, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15464226

ABSTRACT

Developmental cocaine exposure produces long-term alterations in function of many neuronal circuits. This study examined glucose metabolic rates following repeated amphetamine administration in adult male and female rats pretreated with cocaine during postnatal days (PND) 11-20. PND11-20 cocaine increased the response to amphetamine in many components of the motor system and the dorsal caudate-putamen, in particular, and decreased the metabolic response in the hypothalamus. While amphetamine alone produced widespread increases in metabolism, there were no cocaine-related effects in the mesolimbic, limbic or sensory structures. These data suggest that a brief cocaine exposure during development can alter ontogeny and result in abnormal neuronal responses to repeated psychostimulant administration in adulthood.


Subject(s)
Amphetamine/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Glucose/metabolism , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain/metabolism , Female , Male , Motor Activity/drug effects , Rats , Sex Factors , Time Factors
8.
Behav Res Methods Instrum Comput ; 35(3): 452-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14587554

ABSTRACT

Many studies have shown that developmental cocaine exposure alters brain function and behavior, the present study examined the relationship between brain metabolism and behavioral responses to drug challenge. SKF 82958, a selective D1 dopamine agonist, was administered to preweaning cocaine-exposed (50 mg/kg/day) rats and controls at 60 days of age. Deoxyglucose was administered 30 min later, during the peak behavioral response, to measure brain functional activity. Pearsonproduct-moment correlations of behavior (locomotor activity and stereotypic behavior) with rates of glucose metabolism in components of the nigrostriatal and mesolimbic circuits were analyzed. The analysis revealed that under saline-challenge conditions in control animals, rates of metabolism in mesolimbic regions are positively correlated to rates of locomotor activity, whereas in cocaine-treated rats, these correlations were absent. Following SKF challenge, a different pattern was seen; locomotor activity or stereotypic behavior was not correlated with mesolimbic or nigrostriatal metabolism, respectively, in controls but was positively correlated in cocaine-treated rats. Therefore, cocaine exposure during development enhances the coupling of metabolism in components of the mesolimbic and nigrostriatal dopamine systems with the behavioral output associated with these systems under drug-challenge conditions. This may be due to loss of inhibitory influences within the mesolimbic and nigrostriatal systems. Thus, the correlation of behavior and cerebral glucose metabolism provides a unique way of examining the effect of developmental cocaine exposure.


Subject(s)
Benzazepines/pharmacology , Brain/drug effects , Dopamine Agonists/pharmacology , Motor Activity/drug effects , Stereotyped Behavior/drug effects , Animals , Brain/metabolism , Cocaine/pharmacology , Glucose/metabolism , Male , Rats , Rats, Sprague-Dawley
9.
Brain Res Dev Brain Res ; 143(2): 253-9, 2003 Jul 12.
Article in English | MEDLINE | ID: mdl-12855197

ABSTRACT

This study examined glucose metabolic rates following dopamine D(1) agonist challenge in adult male rats pretreated with cocaine during postnatal days 11-20. Water-pretreated control rats showed a reliable decrease in glucose metabolism of rostral mesolimbic structures when challenged with SKF 82958 while cocaine-pretreated males did not. These data support the notion that cocaine exposure during the preweaning period dampens D(1) receptor-mediated function and that the mesolimbic system exhibits a selective vulnerability to early cocaine exposure.


Subject(s)
Benzazepines/pharmacology , Cocaine/pharmacology , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Glucose/metabolism , Limbic System/drug effects , Animals , Body Weight/drug effects , Brain/drug effects , Male , Motor Activity/drug effects , Rats , Weaning
10.
Neurotoxicol Teratol ; 24(6): 773-81, 2002.
Article in English | MEDLINE | ID: mdl-12460660

ABSTRACT

AZT (azidothymidine, zidovudine, ZDV) has become the standard medication to prevent the transmission of the human immunodeficiency virus from mother to fetus. The present study was designed to assess the acoustic startle response (ASR) in adult rats that had been perinatally exposed to AZT. Each litter was randomly assigned to a treatment group: nontreated, AZT 0, 50, 100 or 150 mg/kg. Once daily gastric intubation began prenatally between gestational day (G) 19 and 22 and then continued postnatally between postnatal day (PND) 2 and 20. Between PND75 and PND80, animals were tested for habituation to the acoustic stimuli and prepulse inhibition following a challenge of either saline or 1.0 mg/kg amphetamine (AMP) intraperitoneally. Amphetamine increased ASR and startle latencies throughout the session. The AZT100 dose increased ASR habituation. AZT treatment did not affect prepulse inhibition. Females treated with AZT150 continued to show high ASRs at the end of the startle session. AZT-treated animals showed a dose-dependent increase in peak latency, suggesting a possible abnormal conduction velocity. These effects are independent of handling and intubation effects. Therefore, perinatal AZT treatment results in long-term changes within the primary acoustic startle pathway.


Subject(s)
Anti-HIV Agents/toxicity , Auditory Pathways/drug effects , Brain/drug effects , HIV Infections/drug therapy , Prenatal Exposure Delayed Effects , Reflex, Startle/drug effects , Zidovudine/toxicity , Acoustic Stimulation , Animals , Animals, Newborn , Auditory Pathways/physiopathology , Body Temperature/drug effects , Body Temperature/physiology , Body Weight/drug effects , Body Weight/physiology , Brain/physiopathology , Dose-Response Relationship, Drug , Estrous Cycle/drug effects , Estrous Cycle/physiology , Female , Infectious Disease Transmission, Vertical , Male , Neural Inhibition/drug effects , Neural Inhibition/physiology , Pregnancy , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Reflex, Startle/physiology
11.
Pharmacol Biochem Behav ; 72(1-2): 125-30, 2002 May.
Article in English | MEDLINE | ID: mdl-11900779

ABSTRACT

The present study describes an objective, cost- and time-efficient procedure for characterizing the ataxic effects of psychoactive drugs. Male Sprague-Dawley rats were administered an intraperitoneal injection of either saline or one of three doses (1, 5 or 10 mg/kg) of phencyclidine (PCP) 15 min prior to being placed into an empty standard operant conditioning chamber (all manipulanda were removed). The floor of the test apparatus consisted of parallel rows of metal rods spaced approximately 1.5 cm apart. During a 5-min test, a single observer counted the frequency with which each animal's paws (front or back) slipped between the rows of bars that constituted the cage floor. The data demonstrated that while saline animals exhibited no instability in their ambulation, PCP-treated animals demonstrated a highly reliable dose-dependent increase in the number of "paw slips" in a single trial. Since animals are known to develop tolerance to the ataxic response to PCP, the validity of the test as a measure of drug-induced ataxia was examined in a separate group of animals treated with the middle (5 mg/kg) dose every other day over the course of a 9-day period (i.e., resulting in five injection trials). In this experiment, each subsequent test produced a reliable reduction in the magnitude of the ataxic response, and by the fifth drug challenge, the PCP animals were performing at near-control levels. These results suggest that the "paw slip test" can serve as a simple, reliable, objective and valid measure of drug-induced ataxia. The relevance of the ataxia data for interpreting the locomotor response of animals treated with PCP is also discussed.


Subject(s)
Ataxia/chemically induced , Ataxia/diagnosis , Conditioning, Operant/drug effects , Phencyclidine/toxicity , Research Design , Animals , Conditioning, Operant/physiology , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Research Design/standards
SELECTION OF CITATIONS
SEARCH DETAIL
...