Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
ACS Pharmacol Transl Sci ; 7(3): 888-898, 2024 Mar 08.
Article En | MEDLINE | ID: mdl-38481698

Resveratrol (RES) is a biopharmaceutical classification system (BCS) class II compound with low solubility and high permeability. Several strategies have been explored to overcome the low bioavailability of RES, making the formation of solid dispersions (SDs) one of the most promising. This study aimed at the development of a RES third-generation SD prepared by lyophilization as a strategy to improve RES solubility, dissolution, and oral bioavailability. Eudragit E PO was selected as the hydrophilic carrier in a 1:2 (RES:carrier) ratio, and Gelucire 44/14 as the surfactant, at 16% (w/w) of RES. Differential scanning calorimetry (DSC), scanning electron microscopy (SEM), Fourier-transform infrared spectroscopy (FTIR), polarized light microscopy (PLM), X-ray powder diffraction (XRPD), and particle size distribution (Morphologi 4 Malvern) were used for solid-state characterization and to confirm the conversion of RES to the amorphous state in the SD. Third-generation SD presented an 8-, 12-, and 8-fold increase of RES solubilized compared to pure RES at pH 1.2, 4.5, and 6.8, respectively, and a 10-fold increase compared to the physical mixture (PM), at pH 6.8, after 24 h. In the gastric environment, the dissolution rate of third-generation SD and PM was similar, and 2-fold higher than pure RES after 30 min, while at pH 6.8, third-generation SD presented approximately a 5-fold increase in comparison to pure RES and PM. Third-generation SD presented higher in vitro intestinal permeability compared to its PM and second-generation SD (without Gelucire 44/14). A 2.4 and 1.7-fold increase of RES permeated, respectively in Caco-2 and Caco-2/HT2-MTX models, was obtained with third-generation SD compared to PM, after 3 h. Third-generation SD allowed a 3-fold increase of RES bioavailability compared to second-generation SD, after oral administration of 200 mg/kg of RES to Wistar rats. Enhanced RES oral bioavailability was obtained not only by solubility and dissolution improvement, but also by the interference of Gelucire 44/14, with RES metabolism, and inhibition of P-gp-mediated efflux. The presence of excipients like Gelucire 44/14 in the SD allows for greater bioavailability of orally administered RES, making it easier to obtain some of the physiological benefits demonstrated by this molecule.

2.
Pharmacol Rep ; 74(5): 956-968, 2022 Oct.
Article En | MEDLINE | ID: mdl-36097257

BACKGROUND: Anxiolytic benzodiazepines, due to their clinical effectiveness, are one of the most prescribed drugs worldwide, despite being associated with sedative effects and impaired psychomotor and cognitive performance. Not every GABAA receptor functions in the same manner. Those containing α1 subunits are associated with sleep regulation and have a greater effect on the sedative-hypnotic benzodiazepines, whereas those containing α2 and/or α3 subunits are associated with anxiety phenomena and have a greater effect on the anxiolytic benzodiazepines. Therefore, characterization of the selectivity profile of anxiolytic drugs could translate into a significant clinical impact. METHODS: The present study pharmacodynamically evaluated chlornordiazepam, the main active metabolite of mexazolam, upon GABAA receptors containing α2 and/or α3, anxiety-related, and those containing an α1 subunit, associated with sleep modulation. RESULTS: As shown by whole-cell patch-clamp data, chlornordiazepam potentiated GABA-evoked current amplitude in α2 and α3 containing receptors without changing the current amplitude in α1 containing receptors. However, current decay time increased, particularly in GABAA receptors containing α1 subunits. In contrast, other anxiolytic benzodiazepines such as alprazolam, bromazepam, and zolpidem, all increased currents associated with GABAA receptors containing the α1 subunit. CONCLUSIONS: This novel evidence demonstrates that mexazolam (through its main metabolite chlornordiazepam) has a "pharmacodynamic fingerprint" that correlates better with an anxiolytic profile and fewer sedative effects, when compared to alprazolam, bromazepam and zolpidem, explaining clinical trial outcomes with these drugs. This also highlights the relevance of the pharmacological selectivity over GABAA receptor subtypes in the selection of benzodiazepines, in addition to their clinical performance and pharmacokinetic characteristics.


Anti-Anxiety Agents , Bromazepam , Receptors, GABA-A/metabolism , Zolpidem , Alprazolam/pharmacology , Anti-Anxiety Agents/pharmacology , Bromazepam/pharmacology , Benzodiazepines/pharmacology , Hypnotics and Sedatives/pharmacology , gamma-Aminobutyric Acid
3.
Br J Clin Pharmacol ; 88(10): 4540-4551, 2022 10.
Article En | MEDLINE | ID: mdl-35508762

AIMS: The absorption, metabolism and excretion of opicapone (2,5-dichloro-3-(5-[3,4-dihydroxy-5-nitrophenyl]-1,2,4-oxadiazol-3-yl)-4,6-dimethylpyridine 1-oxide), a selective catechol-O-methyltransferase inhibitor, were investigated. METHODS: Plasma, urine and faeces were collected from healthy male subjects following a single oral dose of 100 mg [14 C]-opicapone. The mass balance of [14 C]-opicapone and metabolic profile were evaluated. RESULTS: The recovery of total administered radioactivity averaged >90% after 144 hours. Faeces were the major route of elimination, representing 70% of the administered dose; 5% and 20% were excreted in urine and expired air, respectively. The Cmax of total radioactivity matched that of unchanged opicapone, whereas the total radioactivity remained quantifiable for a longer period, attributed to the contribution of opicapone metabolites, involving primarily 3-O-sulfate conjugation (58.6% of total circulating radioactivity) at the nitrocatechol ring. Other circulating metabolites, accounting for <10% of the radioactivity exposure, were formed by glucuronidation, methylation, N-oxide reduction and gluthatione conjugation. Additionally, various other metabolites resulting from combinations with the opicapone N-oxide reduced form at the 2,5-dichloro-4,6-dimethylpyridine 1-oxide moiety, including nitro reduction and N-acetylation, reductive opening and cleavage of the 1,2,4-oxadiazole ring and the subsequent hydrolysis products were identified, but only in faeces, suggesting the involvement of gut bacteria. CONCLUSION: [14 C]-opicapone was fully excreted through multiple metabolic pathways. The main route of excretion was in faeces, where opicapone may be further metabolized via reductive metabolism involving the 1,2,4-oxadiazole ring-opening and subsequent hydrolysis.


Catechol O-Methyltransferase Inhibitors , Oxadiazoles , Administration, Oral , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Feces , Healthy Volunteers , Humans , Male , Oxadiazoles/pharmacokinetics
4.
Pharmacol Res Perspect ; 10(1): e00891, 2022 02.
Article En | MEDLINE | ID: mdl-34939338

Opicapone (2,5-dichloro-3-(5-(3,4-dihydroxy-5-nitrophenyl)-1,2,4-oxadiazol-3-yl)-4,6-dimethylpyridine 1-oxide) is a selective catechol-O-methyltransferase inhibitor that has been granted marketing authorization in Europe, Japan, and United States. The present work describes the metabolism and disposition of opicapone in the rat obtained in support to its development and regulatory filling. Plasma levels and elimination of total radioactivity were determined after oral and intravenous administration of [14 C]-opicapone. The maximum plasma concentrations of opicapone-related radioactivity were reached at early time points followed by a gradual return to baseline with a biphasic elimination. Fecal excretion was the primary route of elimination of total radioactivity. Quantitative distribution of drug-related radioactivity demonstrated that opicapone and related metabolites did not distribute to the central nervous system. Opicapone was extensively metabolized in rats resulting in more than 20 phase I and phase II metabolites. Although O-glucuronidation, -sulfation, and -methylation of the nitrocatechol moiety were the principal metabolic pathways, small amount of the N-acetyl derivative was detected, as a result of reduction of the nitro group and subsequent conjugation. Other metabolic transformations included N-oxide reduction to the pyridine derivative and reductive cleavage of 1,2,4-oxadiazole ring followed by further conjugative reactions. Reaction phenotyping studies suggested that SULT 1A1*1 and *2 and UGT1A7, UGT1A8, UGT1A9, and UGT1A10 may be involved in opicapone sulfation and glucuronidation, respectively. However, the reductive metabolic pathways mediated by gut microflora cannot be excluded. Opicapone, in the rat, was found to be rapidly absorbed, widely distributed to peripheric tissues, metabolized mainly via conjugative pathways at the nitro catechol ring, and primarily excreted via feces.


Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Oxadiazoles/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Arylsulfotransferase/metabolism , Catechol O-Methyltransferase Inhibitors/administration & dosage , Glucuronosyltransferase/metabolism , Male , Oxadiazoles/administration & dosage , Phenotype , Rats , Rats, Wistar , Tissue Distribution
5.
Eur J Pharmacol ; 892: 173742, 2021 Feb 05.
Article En | MEDLINE | ID: mdl-33220276

Opicapone is a third generation nitrocatechol catechol-O-methyltransferase inhibitor that has received regional market approval for use as adjunctive therapy to levodopa in Parkinson's disease patients with motor fluctuations. This study evaluated the effects of opicapone as adjunct to levodopa in reversing a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced Parkinson's-like syndrome in cynomolgus monkeys in during opicapone preclinical development program. A Parkinson's-like syndrome was induced in cynomolgus monkeys by daily administrations of MPTP. Evaluation of the animals included scoring with the Primate Parkinsonism Motor Rating Scale (PPMRS) and assessment of locomotor activity. MPTP produced a stable Parkinson's-like behavioural syndrome as evidenced by tremor, postural changes, rigidity, impaired movements and balance, (PPMRS scores of 10-15) and decreased locomotor activity (13% of pre-MPTP values). Opicapone treatment alone, for 14 days, did not change Parkinson's-like symptoms nor decreased subject's locomotor behaviour. Ascending combinations of levodopa/benserazide dose-dependently decreased PPMRS and improved locomotor behaviour reaching statistical significance for levodopa/benserazide doses of 18/4.5 mg/kg and those effects were enhanced in opicapone treated subjects. Opicapone treated subjects as compared vehicle-treated, had markedly reduced erythrocyte catechol-O-methyltransferase activity, significantly increased plasma levodopa levels (1.8-fold higher AUC) with no statistically significant changes in Cmax and significantly reduced 3-OMD AUC and Cmax values (7.8- and 6.8-fold respectively). Opicapone potentiated the improvements in Parkinson's-like symptoms produced by levodopa/benserazide combinations with concomitant increase in plasma levodopa exposure, reduction of plasma 3-O-methyldopa levels and erythrocyte catechol-O-methyltransferase activity, results that were later demonstrated in 2 large Phase 3 studies in Parkinson's disease patients.


Antiparkinson Agents/pharmacology , Behavior, Animal/drug effects , Catechol O-Methyltransferase Inhibitors/pharmacology , Erythrocytes/drug effects , Levodopa/pharmacology , Locomotion/drug effects , Oxadiazoles/pharmacology , Parkinsonian Disorders/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Disease Models, Animal , Drug Therapy, Combination , Erythrocytes/enzymology , Female , Macaca fascicularis , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/physiopathology , Time Factors
6.
Br J Pharmacol ; 177(9): 2123-2142, 2020 05.
Article En | MEDLINE | ID: mdl-31901141

BACKGROUND AND PURPOSE: In 2016, one person died and four others had mild-to-severe neurological symptoms during a phase I trial of the fatty acid amide hydrolase (FAAH) inhibitor BIA 10-2474. EXPERIMENTAL APPROACH: Pharmacodynamic and pharmacokinetic studies were performed with BIA 10-2474, PF-04457845 and JNJ-42165279 using mice, rats and human FAAH expressed in COS cells. Selectivity was evaluated by activity-based protein profiling (APBB) in rats. BIA 10-2474 effect in stroke-prone spontaneously hypertensive rats (SHRSP) was investigated. KEY RESULTS: BIA 10-2474 was 10-fold less potent than PF-04457845 in inhibiting human FAAH in situ but inhibited mouse brain and liver FAAH with ED50 values of 13.5 and 6.2 µg·kg-1 , respectively. Plasma and brain BIA 10-2474 levels were consistent with in situ potency and neither BIA 10-2474 nor its metabolites accumulated following repeat administration. FAAH and α/ß-hydrolase domain containing 6 were the primary targets of BIA 10-2474 and, at higher exposure levels, ABHD11, PNPLA6, PLA2G15, PLA2G6 and androgen-induced protein 1. At 100 mg·kg-1 for 28 days, the level of several lipid species containing arachidonic acid increased. Daily treatment of SHRSP with BIA 10-2474 did not affect mortality rate or increased the incidence of haemorrhage or oedema in surviving animals. CONCLUSIONS AND IMPLICATIONS: BIA 10-2474 potently inhibits FAAH in vivo, similarly to PF-04457845 and interacts with a number of lipid processing enzymes, some previously identified in human cells as off-targets particularly at high levels of exposure. These interactions occurred at doses used in toxicology studies, but the implication of these off-targets in the clinical trial accident remains unclear.


Amidohydrolases , Pyridines , Animals , Cyclic N-Oxides , Endocannabinoids , Enzyme Inhibitors/pharmacology , Group VI Phospholipases A2 , Mice , Pyridines/pharmacology , Rats
7.
Neuropharmacology ; 143: 282-288, 2018 12.
Article En | MEDLINE | ID: mdl-30290160

INTRODUCTION: The aim of the study was to clarify the dose response for inhibition of catechol-O-methyltransferase (COMT) by opicapone, a third generation COMT inhibitor, after acute and repeated administration to the cynomolgus monkey with pharmacokinetic evaluation at the higher dose. METHODS: Three cynomolgus monkeys were used in the study. In the first experiment, COMT inhibition was evaluated over 24 h after the first and at 24 h after the last of 14 daily oral administrations of vehicle, 1, 10 and 100 mg/kg opicapone using a crossover design. In the second experiment, the effect of the maximally effective dose, 100 mg/kg, was retested under the same conditions with additional monitoring of plasma opicapone levels to explore the relationship between pharmacokinetics and pharmacodynamics. RESULTS: Opicapone dose-dependently inhibited COMT activity, significantly so at 10 and 100 mg/kg. Maximal inhibition was 13.1%, 76.4% and 93.2% at 1, 10 and 100 mg/kg respectively, and COMT remained significantly inhibited at 24 h after 10 and 100 mg/kg (42.6% and 60.2% respectively). Following repeated administration of opicapone residual COMT inhibition at 24 h was 15-25% greater at all doses. In contrast to its pharmacodynamic effect, opicapone was rapidly absorbed and eliminated, with no accumulation in plasma following repeated administration. CONCLUSION: Opicapone showed sustained and dose-dependent COMT inhibition despite being rapidly eliminated from plasma and with no evidence for accumulation in plasma after 14 days administration. Opicapone fills the unmet need for a compound with sustained COMT inhibition which will improve levodopa bioavailability in patients with Parkinson's disease.


Catechol O-Methyltransferase Inhibitors/administration & dosage , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Catechol O-Methyltransferase/metabolism , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Erythrocytes/drug effects , Erythrocytes/enzymology , Female , Macaca fascicularis , Random Allocation , Time Factors
8.
ChemMedChem ; 13(20): 2177-2188, 2018 10 22.
Article En | MEDLINE | ID: mdl-30113139

Fatty acid amide hydrolase (FAAH) can be targeted for the treatment of pain associated with various medical conditions. Herein we report the design and synthesis of a novel series of heterocyclic-N-carboxamide FAAH inhibitors that have a good alignment of potency, metabolic stability and selectivity for FAAH over monoacylglycerol lipase (MAGL) and carboxylesterases (CEs). Lead optimization efforts carried out with benzotriazolyl- and imidazolyl-N-carboxamide series led to the discovery of clinical candidate 8 l (3-(1-(cyclohexyl(methyl)carbamoyl)-1H-imidazol-4-yl)pyridine 1-oxide; BIA 10-2474) as a potent and long-acting inhibitor of FAAH. However, during a Phase I clinical trial with compound 8 l, unexpected and unpredictable serious neurological adverse events occurred, affecting five healthy volunteers, including the death of one subject.


Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Cyclic N-Oxides/pharmacology , Enzyme Inhibitors/pharmacology , Pyridines/pharmacology , Administration, Oral , Analgesics/administration & dosage , Analgesics/adverse effects , Analgesics/chemistry , Animals , Brain/drug effects , Brain/metabolism , Clinical Trials, Phase I as Topic , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/adverse effects , Cyclic N-Oxides/chemistry , Drug Design , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/chemistry , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/chemistry , Rats , Structure-Activity Relationship
9.
Neuropharmacology ; 125: 146-155, 2017 Oct.
Article En | MEDLINE | ID: mdl-28734868

Opicapone is a novel catechol-O-methyltransferase (COMT) inhibitor that emerged to fulfil the need of a safer and more efficacious COMT inhibitor. The present study was carried out in order to assess and compare the pharmacokinetics and pharmacodynamics (COMT inhibition) of opicapone after single and multiple oral administrations (30 mg/kg) to Wistar rats. For this purpose, at predefined time points up to 72 h post-dosing, blood, liver and kidneys were collected and, then, the concentrations of opicapone and its active metabolite (BIA 9-1079) were determined in plasma and in liver and kidney tissues, as well as the erythrocyte, liver and kidney COMT activity. No systemic, renal or hepatic accumulation of opicapone was observed following repeated administration. Furthermore, the tissue-systemic exposure relationships to opicapone suggested a low drug exposure in the liver and kidneys. After single-dosing, COMT inhibition profiles were reasonably comparable in all the studied matrices; although similar results were found after multiple-dosing, a higher degree of inhibition was observed, indicating a continuous peripheral COMT inhibition when opicapone is administered once-daily. Despite having a short elimination half-life (≤2.94 h), opicapone showed a strong and long-lasting COMT inhibition in both studies, since more than 50% of the COMT activity was still inhibited at 24 h post-dosing.


Catechol O-Methyltransferase Inhibitors/pharmacology , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Oxadiazoles/pharmacology , Oxadiazoles/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Catechol O-Methyltransferase/metabolism , Catechol O-Methyltransferase Inhibitors/blood , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Erythrocytes/drug effects , Erythrocytes/enzymology , Kidney/drug effects , Kidney/enzymology , Liver/drug effects , Liver/enzymology , Male , Oxadiazoles/blood , Rats, Wistar
10.
Br J Clin Pharmacol ; 83(3): 540-553, 2017 03.
Article En | MEDLINE | ID: mdl-27763682

AIMS: To compare the levodopa/carbidopa (LC) and levodopa/benserazide (LB) pharmacokinetic profiles following repeated doses of opicapone (OPC) administered apart from levodopa. METHODS: Two randomized, double blind, sex-balanced, placebo-controlled studies in four groups of 12 or 18 healthy subjects each. In each group, enrolled subjects received a once-daily morning (5, 15 and 30 mg) or evening (5, 15 and 50 mg) administration of OPC or placebo for up to 28 days. On the morning of Day 11, 12 h after the OPC or placebo evening dose, or the morning of Day 21, 1 h after the OPC or placebo dose, a single dose of immediate-release 100/25 mg LC was administered. Similarly, on Day 18 morning, 12 h after the OPC or placebo evening dose, or Day 28 morning, 1 h after the OPC or placebo dose, a single dose of immediate-release 100/25 mg LB was administered. RESULTS: All OPC treatments, in relation to the placebo group, presented a higher extent of exposure (AUC) to levodopa following either LC or LB doses. A relevant but not dose-dependent increase in the levodopa AUC occurred with all OPC dose groups in relation to placebo. All active treatments significantly inhibited both peak (Emax ) and extent (AUEC) of the catechol-O-methyltransferase activity in relation to placebo. The tolerability profile was favourable. CONCLUSION: Opicapone, as once-daily oral evening regimen and/or 1 h apart from levodopa therapy, increases the bioavailability of levodopa associated with its pronounced, long-lasting and sustained catechol-O-methyltransferase inhibition. The tolerability profile was favourable and similar between OPC and placebo.


Benserazide/pharmacokinetics , Levodopa/pharmacokinetics , Oxadiazoles/pharmacology , Oxadiazoles/pharmacokinetics , Adult , Antiparkinson Agents/pharmacokinetics , Benserazide/adverse effects , Benserazide/blood , Benserazide/pharmacology , Biological Availability , Carbidopa/adverse effects , Carbidopa/pharmacology , Catechol O-Methyltransferase Inhibitors/adverse effects , Catechol O-Methyltransferase Inhibitors/blood , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Catechol O-Methyltransferase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Combinations , Female , Humans , Levodopa/adverse effects , Levodopa/blood , Levodopa/pharmacology , Male , Middle Aged , Oxadiazoles/adverse effects , Oxadiazoles/blood
11.
Pharmacol Rep ; 67(5): 986-95, 2015 Oct.
Article En | MEDLINE | ID: mdl-26398395

BACKGROUND: The aim of this study was to evaluate the effect of carbamazepine (CBZ) upon chemically induced absence seizures and in a genetic absence seizures model in the mouse. METHODS: The γ-butyrolactone (GBL)-induced acute absence seizures and the stargazer spontaneous absence seizures mice models were used to characterize the aggravation of absence seizures induced by oral CBZ treatment. The effect of CBZ upon GABA inward-currents in Ltk cells expressing human recombinant α1ß2γ2, α2ß2γ2, α3ß2γ2 and α5ß2γ2 GABAA receptors was evaluated by means of patch clamp. RESULTS: GBL administration induced motor impairment in NMRI mice. High dose CBZ (25mg/kg body weight) had no effect on motor performance but exacerbated the behavioral incoordination observed for GBL. Also, coadministration of a high dose CBZ and GBL impaired spontaneous locomotion. Moreover, CBZ was investigated after oral administration to evaluate the potential to aggravate GBL-induced acute spike-and-wave discharges (SWD) in the electroencephalogram. High dose CBZ significantly aggravated SWD induced by GBL. Likewise, in the stargazer mouse model of genetic spontaneous absence seizures, CBZ significantly aggravated SWD frequency and duration. Pre-treatment with the T-type Ca(2+) channel blocker ethosuximide (200mg/kg body weight) prevented the CBZ aggravation of SWD induced by GBL and in the stargazer mouse. CBZ increased in a concentration dependent manner sub-maximal α1ß2γ2 and α3ß2γ2 GABA currents. CONCLUSION: CBZ aggravates absence seizures as assessed in two dedicated mouse models of absence seizures. Facilitation of sub-maximal α1ß2γ2, and α3ß2γ2 GABA currents by CBZ may play a role in CBZ-induced GABA-mediated aggravation of absence seizures.


Anticonvulsants/toxicity , Carbamazepine/toxicity , Epilepsy, Absence/chemically induced , 4-Butyrolactone , Animals , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/metabolism , Dose-Response Relationship, Drug , Electroencephalography , Epilepsy, Absence/genetics , Epilepsy, Absence/physiopathology , Ethosuximide/pharmacology , Male , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Patch-Clamp Techniques , Psychomotor Performance/drug effects , Receptors, GABA-A/drug effects , Receptors, GABA-A/genetics
12.
Pharmacol Res Perspect ; 3(2): e00124, 2015 Mar.
Article En | MEDLINE | ID: mdl-26038700

Eslicarbazepine acetate (ESL) is a once daily antiepileptic drug (AED) approved by the European Medicines Agency (EMA), the Food and Drug Administration (FDA) and Health Canada as an adjunctive therapy in adults with partial-onset seizures (POS). In humans and in relevant animal laboratory species, ESL undergoes extensive first pass hydrolysis to its major active metabolite eslicarbazepine that represents ∼95% of circulating active moieties. ESL and eslicarbazepine showed anticonvulsant activity in animal models. ESL may not only suppress seizure activity but may also inhibit the generation of a hyperexcitable network. Data reviewed here suggest that ESL and eslicarbazepine demonstrated the following in animal models: (1) the selectivity of interaction with the inactive state of the voltage-gated sodium channel (VGSC), (2) reduction in VGSC availability through enhancement of slow inactivation, instead of alteration of fast inactivation of VGSC, (3) the failure to cause a paradoxical upregulation of persistent Na(+) current (I NaP), and (4) the reduction in firing frequencies of excitatory neurons in dissociated hippocampal cells from patients with epilepsy who were pharmacoresistant to carbamazepine (CBZ). In addition, eslicarbazepine effectively inhibited high- and low-affinity hCaV3.2 inward currents with greater affinity than CBZ. These preclinical findings may suggest the potential for antiepileptogenic effects; furthermore, the lack of effect upon KV7.2 outward currents may translate into a reduced potential for eslicarbazepine to facilitate repetitive firing.

13.
Xenobiotica ; 45(9): 828-39, 2015.
Article En | MEDLINE | ID: mdl-25915108

1. This study explores the impact of permeability and P-glycoprotein (P-gp) efflux, upon brain exposure to etamicastat, a new dopamine-ß-hydroxylase (DBH) inhibitor and consequently brain levels of catecholamines. 2. Brain exposure to etamicastat (10 mg/kg), following intravenous administration to mice, was residual and upon oral administration of the same dose no compound was detected, concurring with the absence of effects upon brain catecholamines. The intravenous co-administration of elacridar (1.0 mg/kg), a known P-gp/BCRP dual modulator, significantly increased brain etamicastat exposure, but the levels attained were very low when compared to those of nepicastat, a centrally active DBH inhibitor. 3. In vitro permeability studies from apical-to-basal direction conducted in Caco-2 cells and MDCK-II cells showed that etamicastat apparent permeability was 1.2 × 10(-5) and 1.1 × 10(-6 )cm/s, respectively, 5- and 50-fold lower as compared to nepicastat. The secretory efflux ratio in MDCK-II cells overexpressing human P-gp showed an efflux ratio greater than 2, for both compounds, which was significantly decreased by elacridar. Despite its lower bioavailability and higher clearance, as compared to nepicastat, etamicastat showed preferential distribution to peripheral tissues and high plasma free fraction (15.5%), which may explain its effects upon peripheral DBH and catecholamine levels. 4. Though P-gp-mediated efflux may contribute to the limited brain penetration of etamicastat, the low permeability along with the pharmacokinetic properties of etamicastat may be perceived as the main contributors for its peripheral selectivity, which is advantageous for a cardiovascular drug candidate.


ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Benzopyrans/pharmacology , Brain/metabolism , Cell Membrane Permeability/drug effects , Imidazoles/pharmacology , Thiones/pharmacology , Acridines/administration & dosage , Acridines/pharmacology , Animals , Atenolol/pharmacology , Benzopyrans/blood , Benzopyrans/chemistry , Benzopyrans/pharmacokinetics , Biological Transport/drug effects , Blood Proteins/metabolism , Caco-2 Cells , Catecholamines/metabolism , Dogs , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dopamine beta-Hydroxylase/metabolism , Humans , Imidazoles/blood , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Liver/drug effects , Liver/metabolism , Madin Darby Canine Kidney Cells , Male , Mice , Propranolol/pharmacology , Protein Binding/drug effects , Tetrahydroisoquinolines/administration & dosage , Tetrahydroisoquinolines/pharmacology , Thiones/blood , Thiones/chemistry , Thiones/pharmacokinetics , Tissue Distribution/drug effects
14.
Eur J Pharmacol ; 751: 50-8, 2015 Mar 15.
Article En | MEDLINE | ID: mdl-25641750

The interaction of etamicastat, a novel peripherally acting dopamine-ß-hydroxylase (DBH) inhibitor, with the enzyme was studied using a classical kinetic approach and the pharmacodynamics effect of the compound upon administration to rats was also evaluated. SK-N-SH cell homogenates convert tyramine into octopamine with a Km value of 9 mM, and a Vmax of 1747 nmol/mg protein/h. The K(m) value for ascorbate was 3 mM. The inhibition of DBH by etamicastat and nepicastat, a known centrally acting DBH inhibitor, with IC50 values of 107 and 40 nM, respectively, was fully reversed by dilution. Non-linear fitting of the velocities, determined at various concentrations of substrate (tyramine) and co-substrate (ascorbic acid), and of etamicastat and nepicastat, indicated that the inhibition of DBH by both compounds follows a mixed-model inhibition mechanism, approaching competitive behavior with regards to the substrate tyramine, with K(i) values of 34 and 11 nM, respectively. Relatively to ascorbate, both compounds followed a mixed-model inhibition mechanism, approaching uncompetitive behavior. Oral administration of both compounds (at 30 mg/kg) inhibited adrenal DBH activity over time and significantly decreased noradrenaline levels in the heart. Nepicastat also decreased noradrenaline levels in the parietal cortex, but not etamicastat. Both compounds significantly decreased systolic and diastolic blood pressure in spontaneously hypertensive rats. In conclusion, etamicastat and nepicastat behave as multisubstrate DBH inhibitors, binding reversibly and preferentially to the reduced form of the enzyme, and simultaneously at the substrate and oxygen binding sites. Etamicastat, in contrast to nepicastat, offers the advantage of peripheral selectivity without central effects.


Benzopyrans/metabolism , Benzopyrans/pharmacology , Dopamine beta-Hydroxylase/metabolism , Imidazoles/metabolism , Imidazoles/pharmacology , Thiones/metabolism , Adrenal Glands/drug effects , Adrenal Glands/enzymology , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/metabolism , Antihypertensive Agents/pharmacology , Benzopyrans/chemistry , Cell Line , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dopamine beta-Hydroxylase/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/chemistry , Kinetics , Male , Models, Molecular , Protein Binding , Protein Conformation , Rats , Rats, Inbred SHR , Rats, Wistar
15.
Brain ; 138(Pt 2): 371-87, 2015 Feb.
Article En | MEDLINE | ID: mdl-25472797

In human epilepsy, pharmacoresistance to antiepileptic drug therapy is a major problem affecting a substantial fraction of patients. Many of the currently available antiepileptic drugs target voltage-gated sodium channels, leading to a rate-dependent suppression of neuronal discharge. A loss of use-dependent block has emerged as a potential cellular mechanism of pharmacoresistance for anticonvulsants acting on voltage-gated sodium channels. There is a need both for compounds that overcome this resistance mechanism and for novel drugs that inhibit the process of epileptogenesis. We show that eslicarbazepine acetate, a once-daily antiepileptic drug, may constitute a candidate compound that addresses both issues. Eslicarbazepine acetate is converted extensively to eslicarbazepine after oral administration. We have first tested using patch-clamp recording in human and rat hippocampal slices if eslicarbazepine, the major active metabolite of eslicarbazepine acetate, shows maintained activity in chronically epileptic tissue. We show that eslicarbazepine exhibits maintained use-dependent blocking effects both in human and experimental epilepsy with significant add-on effects to carbamazepine in human epilepsy. Second, we show that eslicarbazepine acetate also inhibits Cav3.2 T-type Ca(2+) channels, which have been shown to be key mediators of epileptogenesis. We then examined if transitory administration of eslicarbazepine acetate (once daily for 6 weeks, 150 mg/kg or 300 mg/kg) after induction of epilepsy in mice has an effect on the development of chronic seizures and neuropathological correlates of chronic epilepsy. We found that eslicarbazepine acetate exhibits strong antiepileptogenic effects in experimental epilepsy. EEG monitoring showed that transitory eslicarbazepine acetate treatment resulted in a significant decrease in seizure activity at the chronic state, 8 weeks after the end of treatment. Moreover, eslicarbazepine acetate treatment resulted in a significant decrease in mossy fibre sprouting into the inner molecular layer of pilocarpine-injected mice, as detected by Timm staining. In addition, epileptic animals treated with 150 mg/kg, but not those that received 300 mg/kg eslicarbazepine acetate showed an attenuated neuronal loss. These results indicate that eslicarbazepine potentially overcomes a cellular resistance mechanism to conventional antiepileptic drugs and at the same time constitutes a potent antiepileptogenic agent.


Anticonvulsants/therapeutic use , Dibenzazepines/therapeutic use , Epilepsy/drug therapy , Epilepsy/physiopathology , Adolescent , Adult , Animals , Anticonvulsants/pharmacokinetics , CHO Cells , Child , Child, Preschool , Convulsants , Cricetulus , Dibenzazepines/pharmacokinetics , Epilepsy/chemically induced , Hippocampus/drug effects , Humans , In Vitro Techniques , Male , Mice , Middle Aged , Pilocarpine , Postural Balance/drug effects , Rats , Rats, Wistar , Scopolamine , Young Adult
16.
Neuropharmacology ; 89: 122-35, 2015 Feb.
Article En | MEDLINE | ID: mdl-25242737

This study aimed at evaluating the effects of eslicarbazepine, carbamazepine (CBZ), oxcarbazepine (OXC) and lacosamide (LCM) on the fast and slow inactivated states of voltage-gated sodium channels (VGSC). The anti-epileptiform activity was evaluated in mouse isolated hippocampal slices. The anticonvulsant effects were evaluated in MES and the 6-Hz psychomotor tests. The whole-cell patch-clamp technique was used to investigate the effects of eslicarbazepine, CBZ, OXC and LCM on sodium channels endogenously expressed in N1E-115 mouse neuroblastoma cells. CBZ and eslicarbazepine exhibit similar concentration dependent suppression of epileptiform activity in hippocampal slices. In N1E-115 mouse neuroblastoma cells, at a concentration of 250 µM, the voltage dependence of the fast inactivation was not influenced by eslicarbazepine, whereas LCM, CBZ and OXC shifted the V0.5 value (mV) by -4.8, -12.0 and -16.6, respectively. Eslicarbazepine- and LCM-treated fast-inactivated channels recovered similarly to control conditions, whereas CBZ- and OXC-treated channels required longer pulses to recover. CBZ, eslicarbazepine and LCM shifted the voltage dependence of the slow inactivation (V0.5, mV) by -4.6, -31.2 and -53.3, respectively. For eslicarbazepine, LCM, CBZ and OXC, the affinity to the slow inactivated state was 5.9, 10.4, 1.7 and 1.8 times higher than to the channels in the resting state, respectively. In conclusion, eslicarbazepine did not share with CBZ and OXC the ability to alter fast inactivation of VGSC. Both eslicarbazepine and LCM reduce VGSC availability through enhancement of slow inactivation, but LCM demonstrated higher interaction with VGSC in the resting state and with fast inactivation gating.


Acetamides/pharmacology , Carbamazepine/analogs & derivatives , Carbamazepine/pharmacology , Dibenzazepines/pharmacology , Voltage-Gated Sodium Channels/physiology , Animals , Anticonvulsants/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/physiology , Lacosamide , Male , Mice , Organ Culture Techniques , Oxcarbazepine , Time Factors
17.
Hypertens Res ; 38(1): 30-8, 2015 Jan.
Article En | MEDLINE | ID: mdl-25298210

Hyperactivation of the sympathetic nervous system has an important role in the development and progression of arterial hypertension. This study evaluated the efficacy of etamicastat, a dopamine-ß-hydroxylase (DßH) inhibitor, in controlling high blood pressure in the spontaneously hypertensive rat (SHR), either alone or in combination with other classes of antihypertensives. SHRs were administered with etamicastat by gavage, and its pharmacodynamic and pharmacokinetic properties were evaluated. Etamicastat induced a time-dependent decrease in noradrenaline-to-dopamine ratios in the heart and kidney, and had no effect on catecholamine levels in the frontal cortex of SHRs. Cardiovascular pharmacodynamic effects following administration of etamicastat alone or in combination with other classes of antihypertensive drugs were assessed by telemetry. Etamicastat was evaluated in combination with captopril, losartan, hydrochlorothiazide, metoprolol, prazosin and/or diltiazem. Etamicastat monotherapy induced a dose-dependent reduction in blood pressure without reflex tachycardia. Combination therapy amplified the antihypertensive effects of all tested drugs. In conclusion, inhibition of peripheral DßH with etamicastat, as a monotherapy or combination therapy, may constitute a valid alternative treatment for high blood pressure.


Antihypertensive Agents/therapeutic use , Benzopyrans/therapeutic use , Blood Pressure/drug effects , Hypertension/drug therapy , Imidazoles/therapeutic use , Animals , Antihypertensive Agents/pharmacokinetics , Benzopyrans/pharmacokinetics , Catecholamines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Therapy, Combination , Hypertension/blood , Imidazoles/pharmacokinetics , Kidney/drug effects , Male , Rats, Inbred SHR
18.
Eur J Pharmacol ; 740: 285-94, 2014 Oct 05.
Article En | MEDLINE | ID: mdl-25058908

Despite the importance of sympathetic nervous system in pathophysiological mechanisms of cardiac heart failure and essential hypertension, therapy specifically targeting the sympathetic nervous system is currently underutilized. Etamicastat is a novel dopamine-ß-hydroxylase (DBH) inhibitor that is oxidized into BIA 5-965 and deaminated followed by oxidation to BIA 5-998, which represents 13% of total etamicastat and quantified metabolites. However, the primary metabolic pathway of etamicastat in rats was found to be the N-acetylation (BIA 5-961), which represents 44% of total etamicastat and quantified metabolites. Trace amounts of BIA 5-961 de-sulfated and S-glucuronide were also detected. All the main metabolites of etamicastat inhibited DBH with IC50 values of 306 (228, 409), 629 (534, 741), 427 (350, 522) nM for BIA 5-965, BIA 5-998 and BIA 5-961, respectively. However, only etamicastat (IC50 of 107 (94; 121) nM) was able to reduce catecholamine levels in sympathetic nervous system innervated peripheral tissues, without effect upon brain catecholamines. Quantitative whole body autoradiography revealed a limited transfer of etamicastat related radioactivity to brain tissues and the mean recovery of radioactivity was ~90% of the administered radioactive dose, eliminated primarily via renal excretion over 5 days. The absolute oral bioavailability of etamicastat was 64% of the administered dose. In conclusion, etamicastat is a peripheral selective DBH inhibitor mainly N-acetylated in the aminoethyl moiety and excreted in urine. Etamicastat main metabolites inhibit DBH, but only etamicastat demonstrated unequivocal pharmacological effects as a DBH inhibitor with impact upon the activity of the sympathetic nervous system under in vivo conditions.


Benzopyrans/pharmacology , Dopamine beta-Hydroxylase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Acetylation , Adrenal Glands/drug effects , Adrenal Glands/enzymology , Animals , Benzopyrans/blood , Benzopyrans/pharmacokinetics , Benzopyrans/urine , Cell Line, Tumor , Dopamine/metabolism , Dopamine beta-Hydroxylase/metabolism , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/urine , Feces/chemistry , Glucuronosyltransferase/metabolism , Humans , Imidazoles/blood , Imidazoles/pharmacokinetics , Imidazoles/urine , Male , Mice , Myocardium/metabolism , Norepinephrine/metabolism , Rats, Wistar , Recombinant Proteins/metabolism
19.
Drug Metab Dispos ; 41(12): 2081-6, 2013 Dec.
Article En | MEDLINE | ID: mdl-24013186

Etamicastat [(R)-5-(2-aminoethyl)-1-(6,8-difluorochroman-3-yl)-1H-imidazole-2(3H)-thione hydrochloride] is a reversible dopamine-ß-hydroxylase inhibitor that decreases norepinephrine levels in sympathetically innervated tissues. After in vivo administration, N-acetylation of etamicastat was found to be a main metabolic pathway. The purpose of the current study was to characterize the N-acetylation of etamicastat by N-acetyltransferases (NAT1 and NAT2) and evaluate potential species differences in etamicastat N-acetylation using a sensitive and specific liquid chromatography-mass spectrometry assay. Marked differences in etamicastat N-acetylation were observed among the laboratory species and humans. After oral administration, the rat, hamster, and human subjects presented the highest rates of etamicastat N-acetylation, whereas almost no acetylation was observed in the mouse, rabbit, minipig, and monkey and no acetylation was observed in the dog. In in vitro studies, rats and humans showed similar acetylation rates, whereas no acetylation was detected in the dog. Studies performed with human recombinant NAT1 4 and NAT2 4 enzymes revealed that both were able to conjugate etamicastat, although at different rates. NAT1 had lower affinity compared with NAT2 (Km, 124.8 ± 9.031 µM and 17.14 ± 3.577 µM, respectively). A significant correlation (r(2) = 0.65, P < 0.05) was observed in a comparison of etamicastat N-acetylation by human single-donor enzymes and sulfamethazine, a selective substrate to NAT2. No correlation was observed with p-aminosalicylic acid, a NAT1 selective substrate. In conclusion, these results suggest that NAT2 and, to a lesser extent, NAT1 contribute to etamicastat N-acetylation. Furthermore, the high interspecies and intraspecies differences in N-acetylation should be taken into consideration when evaluating the in vivo bioavailability of etamicastat.


Benzopyrans/metabolism , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dopamine beta-Hydroxylase/metabolism , Enzyme Inhibitors/metabolism , Imidazoles/metabolism , Acetylation , Animals , Arylamine N-Acetyltransferase/metabolism , Cricetinae , Cytosol/metabolism , Dogs , Female , Humans , Kinetics , Macaca fascicularis , Male , Mice , Rabbits , Rats , Swine , Swine, Miniature
20.
J Sep Sci ; 36(11): 1693-702, 2013 Jun.
Article En | MEDLINE | ID: mdl-23495043

Despite of membrane catechol-O-methyltransferase (MBCOMT, EC 2.1.1.6) physiological importance on catecholamines' O-methylation, no studies allowed their total isolation. Therefore, for the first time, we compare the performance of three hydrophobic adsorbents (butyl-, epoxy-, and octyl-Sepharose) in purification of recombinant human COMT (hMBCOMT) from crude Brevibacillus choshinensis cell lysates to develop a sustainable chromatographic process. Hydrophobic matrices were evaluated in terms of selectivity and hMBCOMT's binding and elution conditions. Results show that hMBCOMT's adsorption was promoted on octyl and butyl at ≤375 mM NaH2 PO4, while on epoxy higher concentrations (>850 mM) were required. Additionally, hMBCOMT's elution was promoted on epoxy, butyl, and octyl using respectively 0.1-0.5, 0.25-1, and 1% of Triton X-100. On butyl media, a stepwise strategy using 375 and 0 mM NaH2PO4, followed by three elution steps at 0.25, 0.7 and 1% Triton X-100, allowed selective hMBCOMT isolation. In conclusion, significant amounts of MBCOMT were purified with high selectivity on a single chromatography procedure, despite its elution occurs on multiple peaks. Although successful applications of hydrophobic interaction chromatography in purification of membrane proteins are uncommon, we proved that traditional hydrophobic matrices can open a promising unexplored field to fulfill specific requirements for kinetic and pharmacological trials.


Catechol O-Methyltransferase/isolation & purification , Chromatography, Liquid/methods , Adsorption , Brevibacillus/genetics , Brevibacillus/metabolism , Catechol O-Methyltransferase/chemistry , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
...