Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Pharmacol Res Perspect ; 9(3): e00792, 2021 05.
Article in English | MEDLINE | ID: mdl-34018344

ABSTRACT

The management of Parkinson's disease (PD) is frequently compromised by complications induced by dopaminergic treatment such as involuntary movements (dyskinesias) and psychosis. Mesdopetam (IRL790) is a novel dopamine D3 receptor antagonist developed for the management of complications of therapy in PD. This study evaluated the safety, tolerability, and pharmacokinetics of escalating single and multiple doses of mesdopetam. We conducted a prospective, single-center, randomized, double-blind, placebo-controlled phase I, and first-in-human (FIH) study with mesdopetam administered to healthy male subjects. Overall, mesdopetam was well-tolerated up to a 120 mg single dose and up to 80 mg upon multiple dosing. Adverse events (AEs) were mainly related to the nervous system and were dose-dependent. No serious adverse events occurred and no AEs led to withdrawal. The results of the single-ascending-dose and multiple-ascending-dose parts indicated dose- and time-independent pharmacokinetics with rapid absorption and maximum plasma levels that were generally reached within 2 h after dosing. No accumulation was observed upon multiple dosing. It is concluded that mesdopetam was safe and well-tolerated in healthy male volunteers. Pharmacokinetic analysis indicated rapid absorption and dose-linear pharmacokinetics of mesdopetam, with a plasma half-life of around 7 h, upon single and repeated dosing. The pharmacokinetics of mesdopetam supports twice-daily use in patients.


Subject(s)
Dopamine Antagonists/administration & dosage , Phenyl Ethers/administration & dosage , Propylamines/administration & dosage , Receptors, Dopamine D3/antagonists & inhibitors , Administration, Oral , Adolescent , Adult , Biological Availability , Dopamine Antagonists/adverse effects , Dopamine Antagonists/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Fasting/metabolism , Food-Drug Interactions , Half-Life , Healthy Volunteers , Humans , Male , Middle Aged , Phenyl Ethers/adverse effects , Phenyl Ethers/pharmacokinetics , Propylamines/adverse effects , Propylamines/pharmacokinetics , Young Adult
2.
Arch Pharm Res ; 43(12): 1356-1363, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33245517

ABSTRACT

The aim of this study was to investigate the effects of paroxetine, a potent inhibitor of CYP2D6, on the pharmacokinetics of atomoxetine and its two metabolites, 4-hydroxyatomoxetine and N-desmethylatomoxetine, in different CYP2D6 genotypes. Twenty-six healthy subjects were recruited and divided into CYP2D6*wt/*wt (*wt=*1 or *2, n = 10), CYP2D6*wt/*10 (n = 9), and CYP2D6*10/*10 groups (n = 7). In atomoxetine phase, all subjects received a single oral dose of atomoxetine (20 mg). In paroxetine phase, after administration of a single oral dose of paroxetine (20 mg) for six consecutive days, all subjects received a single oral dose of atomoxetine with paroxetine. Plasma concentrations of atomoxetine and its metabolites were determined up to 24 h after dosing. During atomoxetine phase, there were significant differences in Cmax and AUC0-24 of atomoxetine and N-desmethylatomoxetine among three genotype groups, whereas significant differences were not found in relation to CYP2D6*10 allele after administration of paroxetine. AUC ratios of 4-hydroxyatomoxetine and N-desmethylatomoxetine to atomoxetine were significantly different among three genotype groups during atomoxetine phase (all, P < 0.001), but after paroxetine treatment significant differences were not found. After paroxetine treatment, AUC0-24 of atomoxetine was increased by 2.3-, 1.7-, and 1.3-fold, in CYP2D6*wt/*wt, CYP2D6*wt/*10, and CYP2D6*10/*10 groups in comparison to atomoxetine phase, respectively. AUC ratio of 4-hydroxyatomoxetine to atomoxetine in each group was significantly decreased, whereas AUC ratio of N-desmethylatomoxetine to atomoxetine significantly increased after administration of paroxetine. In conclusion, paroxetine coadministration significantly affected pharmacokinetic parameters of atomoxetine and its two metabolites, 4-hydroxyatomoxetine and N-desmethylatomoxetine. When atomoxetine was administered alone, Cmax, AUC0-24 and CL/F of atomoxetine were significantly different among the three CYP2D6 genotype groups. However, after paroxetine coadministration, no significant differences in these pharmacokinetic parameters were observed among the CYP2D6 genotype groups.


Subject(s)
Atomoxetine Hydrochloride/pharmacokinetics , Cytochrome P-450 CYP2D6 Inhibitors/pharmacology , Cytochrome P-450 CYP2D6/metabolism , Paroxetine/pharmacology , Pharmacogenomic Variants , Phenols/pharmacokinetics , Phenyl Ethers/pharmacokinetics , Propylamines/pharmacokinetics , Administration, Oral , Adult , Atomoxetine Hydrochloride/administration & dosage , Biotransformation , Cytochrome P-450 CYP2D6/genetics , Drug Interactions , Female , Genotype , Humans , Male , Models, Biological , Pharmacogenetics , Young Adult
3.
Drug Metab Pharmacokinet ; 35(2): 191-200, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32184039

ABSTRACT

Atomoxetine is an approved medicine for attention-deficit/hyperactivity disorder and a cytochrome P450 2D6 (CYP2D6) probe substrate. Simple physiologically based pharmacokinetic (PBPK) models and compartment models were set up to account for drug monitoring results of 33 Japanese patients (6-15 years of age) to help establish the correct dosage for the evaluation of clinical outcomes. The steady-state one-point drug monitoring data for the most participants indicated the extensive biotransformation of atomoxetine to 4-hydroxyatomoxetine under individually prescribed doses of atomoxetine. However, 5 participants (with impaired CYP2D6 activity scores based on the CYP2D6 genotypes) showed high plasma concentrations of atomoxetine (0.53-1.5 µM) compared with those of total 4-hydroxyatomoxetine (0.49-1.4 µM). Results from full PBPK models using the in-built Japanese pediatric system of software Simcyp, one-compartment models, and new simple PBPK models (using parameters that reflected the subjects' small body size and normal/reduced CYP2D6-dependent clearance) could overlay one-point measured drug/metabolite plasma concentrations from almost common 28 participants within threefold ranges. Validated one-compartment or simple PBPK models can be used to predict steady-state plasma concentrations of atomoxetine and/or its primary metabolites in Japanese pediatric patients (>6 years) who took a variety of individualized doses in a clinical setting.


Subject(s)
Atomoxetine Hydrochloride/pharmacokinetics , Attention Deficit Disorder with Hyperactivity/drug therapy , Cytochrome P-450 CYP2D6/genetics , Models, Biological , Phenols/pharmacokinetics , Propylamines/pharmacokinetics , Adolescent , Asian People , Atomoxetine Hydrochloride/blood , Attention Deficit Disorder with Hyperactivity/diagnosis , Attention Deficit Disorder with Hyperactivity/metabolism , Child , Cytochrome P-450 CYP2D6/metabolism , Drug Monitoring , Female , Genotype , Humans , Male , Phenols/blood , Propylamines/blood
4.
Eur J Pharm Sci ; 143: 105184, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31846695

ABSTRACT

The present study describes synthesis of amino-decorated mesoporous silica nanoparticles (MSNs) for sustained delivery and enhanced bioavailability of sofosbuvir. Sofosbuvir is active against hepatitis C virus and pharmaceutically classified as class III drug according to biopharmaceutics classification system (BCS). MSNs were synthesized using modified sol-gel method and the surface was decorated with amino functionalization. Drug loaded MSNs were also grafted with polyvinyl alcohol in order to compare it with the amino-decorated MSNs for sustained drug release. The prepared MSNs were extensively characterized and the optimized formulation was toxicologically and pharmacokinetically evaluated. The functionalized MSNs of 196 nm size entrapped 29.13% sofosbuvir in the pores, which was also confirmed by the decrease in surface area, pore volume and pore size. The drug-loaded amino-decorated MSNs revealed an improved thermal stability as confirmed by thermal analysis. Amino-decorated MSNs exhibited Fickian diffusion controlled sofosbuvir release as compared with non-functionalized and PVA grafted MSNs. Amino-decorated MSNs were deemed safe to use in Sprague-Dawley rats after 14-days exposure as confirmed by the toxicological studies. More interestingly, we achieved a 2-fold higher bioavailability of sofosbuvir in Sprague-Dawley rats in comparison with sofosbuvir alone, and the Tmax was delayed 3-times indicating a sustained release of sofosbuvir.


Subject(s)
Antiviral Agents , Drug Carriers , Nanoparticles , Propylamines , Silanes , Silicon Dioxide , Sofosbuvir , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/toxicity , Cell Survival/drug effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/toxicity , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/toxicity , Drug Liberation , Hep G2 Cells , Humans , Male , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/toxicity , Polyvinyl Alcohol/chemistry , Porosity , Propylamines/administration & dosage , Propylamines/chemistry , Propylamines/pharmacokinetics , Propylamines/toxicity , Rats, Sprague-Dawley , Silanes/administration & dosage , Silanes/chemistry , Silanes/pharmacokinetics , Silanes/toxicity , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Silicon Dioxide/toxicity , Sofosbuvir/administration & dosage , Sofosbuvir/chemistry , Sofosbuvir/pharmacokinetics , Sofosbuvir/toxicity
5.
Colloids Surf B Biointerfaces ; 181: 48-57, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31121381

ABSTRACT

Carbon dots (CDots) for their excellent optical and other properties have been widely pursued for potential biomedical applications, in which a more comprehensive understanding on the cellular behaviors and mechanisms of CDots is required. For such a purpose, two kinds of CDots with surface passivation by 3-ethoxypropylamine (EPA-CDots) and oligomeric polyethylenimine (PEI-CDots) were selected for evaluations on their uptakes by human cervical carcinoma HeLa cells at three cell cycle phases (G0/G1, S and G2/M), and on their different internalization pathways and translocations in cells. The results show that HeLa cells could internalize both CDots by different pathways, with an overall slightly higher internalization efficiency for PEI-CDots. The presence of serum in culture media could have major effects, significantly enhancing the cellular uptake of EPA-CDots, yet markedly inhibiting that of PEI-CDots. The HeLa cells at different cell cycle phases have different behaviors in taking up the CDots, which are also affected by the different dot surface moieties and serum in culture media. Mechanistic implications of the results and the opportunities associated with an improved understanding on the cellular behaviors of CDots for potentially the manipulation and control of their cellular uptakes and translocations are discussed.


Subject(s)
Carbon/pharmacokinetics , Quantum Dots/chemistry , Carbon/chemistry , Cell Survival/drug effects , HeLa Cells , Humans , Particle Size , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacokinetics , Propylamines/chemistry , Propylamines/pharmacokinetics , Surface Properties , Tumor Cells, Cultured
6.
Neuropharmacology ; 131: 96-103, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29217282

ABSTRACT

Methamphetamine (METH) is a highly addictive drug, but no pharmacological treatment is yet available for METH use disorders. Similar to METH, the wake-promoting drug (R)-modafinil (R-MOD) binds to the dopamine transporter (DAT). Unlike METH, R-MOD is not a substrate for transport by DAT and has low abuse potential. We tested the hypothesis that the atypical DAT inhibitor R-MOD and compounds that are derived from modafinil would decrease METH intake by reducing the actions of METH at the DAT. We tested the effects of systemic injections of R-MOD and four novel modafinil-derived ligands with increased DAT affinity (JJC8-016, JJC8-088, JJC8-089, and JJC8-091) on intravenous (i.v.) METH self-administration in rats that were allowed short access (ShA; 1 h) or long access (LgA; 6 h) to the drug. ShA rats exhibited stable METH intake over sessions, whereas LgA rats exhibited an escalation of drug intake. R-MOD decreased METH self-administration in ShA and LgA rats (in the 1st hour only). JJC8-091 and JJC8-016 decreased METH self-administration in both ShA and LgA rats. JJC8-089 decreased METH self-administration in LgA rats only, whereas JJC8-088 had no effect on METH self-administration in either ShA or LgA rats. These findings support the potential of atypical DAT inhibitors for the treatment of METH use disorders and suggest several novel compounds as candidate drugs.


Subject(s)
Benzhydryl Compounds/therapeutic use , Central Nervous System Stimulants/administration & dosage , Compulsive Behavior/drug therapy , Dopamine Plasma Membrane Transport Proteins/metabolism , Methamphetamine/administration & dosage , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Dopamine Antagonists/pharmacokinetics , Dopamine Antagonists/therapeutic use , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Male , Modafinil , Propylamines/pharmacokinetics , Propylamines/therapeutic use , Rats , Rats, Wistar , Saccharin/administration & dosage , Self Administration , Time Factors , Wakefulness-Promoting Agents
7.
Eur J Drug Metab Pharmacokinet ; 42(2): 261-268, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27207674

ABSTRACT

BACKGROUND AND OBJECTIVES: Atomoxetine is the first non-stimulant drug to be approved for the treatment of ADHD, while the effect of myricetin on the pharmacokinetic of atomoxetine in rats or human is still unknown. The present work was to study the impact of myricetin on the metabolism of atomoxetine both in vivo and in vitro. METHODS: Twenty healthy male Sprague-Dawley rats were randomly divided into four groups: A (control group), B (100 mg/kg myricetin), C (50 mg/kg myricetin), and D (25 mg/kg myricetin). A single dose of atomoxetine (10 mg/kg) was administrated half an hour later. In addition, human and rat liver microsomes were performed to determine the effect of myricetin on the metabolism of atomoxetine in vitro. RESULTS: Group B, C, D all increased the C max and AUC of atomoxetine, but decreased the C max and AUC of 4-hydroxyatomoxetine. Moreover, myricetin showed inhibitory effect on human and rat microsomes, the IC50 of myricetin was 8.651 and 35.45 µmol/L, respectively. CONCLUSIONS: Our study showed that myricetin could significantly inhibit the formation of atomoxetine metabolite both in vivo and in vitro. It is recommended that the effect of myricetin on the metabolism of atomoxetine should be noted and atomoxetine plasma concentration should be monitored.


Subject(s)
Atomoxetine Hydrochloride/pharmacokinetics , Flavonoids/pharmacology , Microsomes, Liver/metabolism , Phenols/pharmacokinetics , Propylamines/pharmacokinetics , Adrenergic Uptake Inhibitors/administration & dosage , Adrenergic Uptake Inhibitors/pharmacology , Animals , Area Under Curve , Atomoxetine Hydrochloride/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions , Flavonoids/administration & dosage , Humans , Inhibitory Concentration 50 , Male , Rats , Rats, Sprague-Dawley
8.
Drug Metab Dispos ; 44(7): 1070-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27052878

ABSTRACT

Atomoxetine (ATX) is a second-line nonstimulant medication used to control symptoms of attention deficit hyperactivity disorder (ADHD). Inconsistent therapeutic efficacy has been reported with ATX, which may be related to variable CYP2D6-mediated drug clearance. We characterized ATX metabolism in a panel of human liver samples as a basis for a bottom-up PBPK model to aid in ATX exposure prediction and control. Km, Vmax, and Clint values in pooled human liver microsomes (HLMs) were 2.4 µM, 479 pmol/min/mg protein, and 202 µl/min/mg protein, respectively. Mean population values of kinetic parameters are not adequate to describe variability in a population, given that Km, Vmax, and Clint values from single-donor HLMs ranged from 0.93 to 79.2 µM, 20.0 to 1600 pmol/min/mg protein, and 0.3 to 936 µl/min/mg protein. All kinetic parameters were calculated from 4-hydroxyatomoxetine (4-OH-ATX) formation. CYP2E1 and CYP3A contributed to 4-OH-ATX formation in livers with CYP2D6 intermediate and poor metabolizer status. In HLMs with lower CYP2D6 activity levels, 2-hydroxymethylatomoxetine (2-CH2OH-ATX) formation became a more predominant pathway of metabolism, which appeared to be catalyzed by CYP2B6. ATX biotransformation at clinically relevant plasma concentrations was characterized in a panel of pediatric HLM (n = 116) samples by evaluating primary metabolites. Competing pathways of ATX metabolism [N-desmethylatomoxetine (NDM-ATX) and 2-CH2OH-ATX formation] had increasing importance in livers with lesser CYP2D6 activity, but, overall ATX clearance was still compromised. Modeling ATX exposure to individualize therapy would require comprehensive knowledge of factors that affect CYP2D6 activity as well as an understanding of competing pathways, particularly for individuals with lower CYP2D6 activity.


Subject(s)
Atomoxetine Hydrochloride/administration & dosage , Atomoxetine Hydrochloride/pharmacokinetics , Attention Deficit Disorder with Hyperactivity/drug therapy , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Drug Dosage Calculations , Models, Biological , Adolescent , Adult , Age Factors , Attention Deficit Disorder with Hyperactivity/diagnosis , Attention Deficit Disorder with Hyperactivity/psychology , Biotransformation , Child , Cytochrome P-450 Enzyme System/genetics , Genotype , Humans , Hydroxylation , Infant , Isoenzymes , Methylation , Microsomes, Liver/metabolism , Middle Aged , Phenols/pharmacokinetics , Phenotype , Propylamines/pharmacokinetics , Substrate Specificity , Young Adult
9.
Arch Pharm Res ; 38(11): 2083-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26254792

ABSTRACT

To investigate the effect of the variant CYP2D6*10 allele on the pharmacokinetics of atomoxetine and its metabolites, 4-hydroxyatomoxetine (4-HAT) and N-desmethylatomoxetine (NAT), in healthy subjects, a single oral dose of atomoxetine was administered to 62 subjects with a CYP2D6*wt/*wt (*wt = *1 or *2, n = 22), CYP2D6*wt/*10 (n = 22) or CYP2D6*10/*10 (n = 18) genotype. Plasma samples were then collected for 24 h after atomoxetine administration. The concentrations of atomoxetine and its metabolites were assayed using LC-MS/MS. For atomoxetine, the Cmax, AUC0-∞, t1/2 and CL/F showed genotype-dependent differences. The CYP2D6*10/*10 and CYP2D6*wt/*10 groups showed 1.74- and 1.15-fold higher Cmax, 3.40- and 1.33-fold higher AUC0-∞, and 69.7 and 24.6 % lower CL/F, compared to those of the CYP2D6*wt/*wt group, respectively. The Cmax and t1/2 for 4-HAT were lower and longer in the CYP2D6*10/*10 group than those in the CYP2D6*wt/*wt group, but the AUC0-∞ was not different between these groups. The Cmax, AUC0-∞ and t1/2 for NAT were profoundly greater in the CYP2D6*10/*10 group than they were in the CYP2D6*wt/*wt group. The concentration of active moieties of atomoxetine (atomoxetine + 4-HAT) in the CYP2D6*10/*10 group was 3.32-fold higher than that in the CYP2D6*wt/*wt group. The mean exposure to active moieties of atomoxetine was markedly higher in subjects with the CYP2D6*10/*10 genotype compared to that in those with the CYP2D6*wt/*wt genotype. The higher systemic exposure of the active atomoxetine moieties in CYP2D6*10/*10 individuals may increase the risk of concentration-related adverse events of atomoxetine, although this has not yet been clinically confirmed.


Subject(s)
Atomoxetine Hydrochloride/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Phenols/pharmacokinetics , Phenyl Ethers/pharmacokinetics , Propylamines/pharmacokinetics , Adrenergic Uptake Inhibitors/pharmacokinetics , Adult , Alleles , Area Under Curve , Chromatography, Liquid , Genotype , Half-Life , Humans , Male , Tandem Mass Spectrometry , Young Adult
10.
CNS Drugs ; 29(2): 131-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25698145

ABSTRACT

Atomoxetine was first licensed to treat attention-deficit/hyperactivity disorder (ADHD) in children and adolescents in the US in 2002. The aim of this paper is to comprehensively review subsequent publications addressing the efficacy of atomoxetine in 6- to 18-year-olds with ADHD. We identified 125 eligible papers using a predefined search strategy. Overall, these papers demonstrate that atomoxetine is an effective treatment for the core ADHD symptoms (effect sizes 0.6-1.3, vs. placebo, at 6-18 weeks), and improves functional outcomes and quality of life, in various pediatric populations with ADHD (i.e., males/females, patients with co-morbidities, children/adolescents, and with/without prior exposure to other ADHD medications). Initial responses to atomoxetine may be apparent within 1 week of treatment, but can take longer (median 23 days in a 6-week study; n=72). Responses often build gradually over time, and may not be robust until after 3 months. A pooled analysis of six randomized placebo-controlled trials (n=618) indicated that responses at 4 weeks may predict response at 6-9 weeks, although another pooled analysis of open-label data (n=338) suggests that the probability of a robust response to atomoxetine [≥40% decrease in ADHD-Rating Scale (ADHD-RS) scores] may continue to increase beyond 6-9 weeks. Atomoxetine may demonstrate similar efficacy to methylphenidate, particularly immediate-release methylphenidate, although randomized controlled trials are generally limited by short durations (3-12 weeks). In conclusion, notwithstanding these positive findings, before initiating treatment with atomoxetine, it is important that the clinician sets appropriate expectations for the patient and their family with regard to the likelihood of a gradual response, which often builds over time.


Subject(s)
Adrenergic Uptake Inhibitors/administration & dosage , Attention Deficit Disorder with Hyperactivity/drug therapy , Propylamines/administration & dosage , Adolescent , Adrenergic Uptake Inhibitors/pharmacokinetics , Atomoxetine Hydrochloride , Attention Deficit Disorder with Hyperactivity/complications , Child , Comorbidity , Humans , Propylamines/pharmacokinetics , Randomized Controlled Trials as Topic
11.
Anal Bioanal Chem ; 407(12): 3457-70, 2015 May.
Article in English | MEDLINE | ID: mdl-25711990

ABSTRACT

The number of so-called new psychoactive substances (NPS) is still increasing by modification of the chemical structure of known (scheduled) drugs. As analogues of amphetamines, 2-aminopropyl-benzofurans were sold. They were consumed because of their euphoric and empathogenic effects. After the 5-(2-aminopropyl)benzofurans, the 6-(2-aminopropyl)benzofuran isomers appeared. Thus, the question arose whether the metabolic fate, the mass spectral fragmentation, and the detectability in urine are comparable or different and how an intake can be differentiated. In the present study, 6-(2-aminopropyl)benzofuran (6-APB) and its N-methyl derivative 6-MAPB (N-methyl-6-(2-aminopropyl)benzofuran) were investigated to answer these questions. The metabolites of both drugs were identified in rat urine and human liver preparations using GC-MS and/or liquid chromatography-high resolution-mass spectrometry (LC-HR-MS(n)). Besides the parent drug, the main metabolite of 6-APB was 4-carboxymethyl-3-hydroxy amphetamine and the main metabolites of 6-MAPB were 6-APB (N-demethyl metabolite) and 4-carboxymethyl-3-hydroxy methamphetamine. The cytochrome P450 (CYP) isoenzymes involved in the 6-MAPB N-demethylation were CYP1A2, CYP2D6, and CYP3A4. An intake of a common users' dose of 6-APB or 6-MAPB could be confirmed in rat urine using the authors' GC-MS and the LC-MS(n) standard urine screening approaches with the corresponding parent drugs as major target allowing their differentiation. Furthermore, a differentiation of 6-APB and 6-MAPB in urine from their positional isomers 5-APB and 5-MAPB was successfully performed after solid phase extraction and heptafluorobutyrylation by GC-MS via their retention times.


Subject(s)
Benzofurans/metabolism , Benzofurans/urine , Designer Drugs/pharmacokinetics , Mass Spectrometry/methods , Propylamines/metabolism , Propylamines/urine , Animals , Benzofurans/pharmacokinetics , Chromatography, Liquid/methods , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/metabolism , Designer Drugs/metabolism , Gas Chromatography-Mass Spectrometry/methods , Humans , Inactivation, Metabolic , Isomerism , Male , Microsomes, Liver/drug effects , Propylamines/pharmacokinetics , Rats, Wistar , Solid Phase Extraction
12.
BMC Gastroenterol ; 14: 188, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25407279

ABSTRACT

BACKGROUND: The γ-aminobutyric acid type B-receptor agonist lesogaberan (AZD3355) has been developed for use in patients with gastroesophageal reflux disease (GERD) symptoms despite proton pump inhibitor (PPI) therapy (partial responders). This study aimed to explore the dose-response effect of lesogaberan on reflux episodes in partial responders. METHODS: In this randomized, single-centre, double-blind, crossover, placebo-controlled study, partial responders taking optimised PPI therapy were given 30, 90, 120 and 240 mg doses of lesogaberan. Each dose was given twice (12 h apart) during a 24-h period, during which impedance-pH measurements were taken. RESULTS: Twenty-five patients were included in the efficacy analysis and 27 in the safety analysis. The effect of lesogaberan on the mean number of reflux episodes was dose-dependent, and all doses significantly reduced the mean number of reflux episodes relative to placebo. Lesogaberan also dose-dependently reduced the mean number of acid reflux episodes (except the 30 mg dose) and weakly acid reflux episodes (all doses) significantly, relative to placebo. Regardless of dose, lesogaberan had a similar effect on the percentage of time with esophageal pH < 4 [mean reduction: 68.5% (30 mg), 54.2% (90 mg), 65.9% (120 mg), 72.1% (240 mg); p < 0.05 except 90 mg dose]. No adverse events led to discontinuation and no serious adverse events occurred during active treatment. CONCLUSIONS: Lesogaberan inhibited reflux in a dose-dependent manner in partial responders taking optimised PPI therapy, and these effects were significant versus placebo. All lesogaberan doses were well tolerated and were not associated with clinically relevant adverse events. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT01043185.


Subject(s)
GABA-A Receptor Agonists/administration & dosage , Gastroesophageal Reflux/drug therapy , Gastrointestinal Agents/administration & dosage , Phosphinic Acids/administration & dosage , Propylamines/administration & dosage , Adult , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Esophagus/physiopathology , Female , GABA-A Receptor Agonists/adverse effects , GABA-A Receptor Agonists/pharmacokinetics , Gastroesophageal Reflux/physiopathology , Headache/chemically induced , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Nausea/chemically induced , Phosphinic Acids/adverse effects , Phosphinic Acids/pharmacokinetics , Propylamines/adverse effects , Propylamines/pharmacokinetics , Young Adult
13.
J Membr Biol ; 247(9-10): 1043-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24898094

ABSTRACT

Amphipols (APols) are polymeric surfactants that keep membrane proteins (MPs) water-soluble in the absence of detergent, while stabilizing them. They can be used to deliver MPs and other hydrophobic molecules in vivo for therapeutic purposes, e.g., vaccination or targeted delivery of drugs. The biodistribution and elimination of the best characterized APol, a polyacrylate derivative called A8-35, have been examined in mice, using two fluorescent APols, grafted with either Alexa Fluor 647 or rhodamine. Three of the most common injection routes have been used, intravenous (IV), intraperitoneal (IP), and subcutaneous (SC). The biodistribution has been studied by in vivo fluorescence imaging and by determining the concentration of fluorophore in the main organs. Free rhodamine was used as a control. Upon IV injection, A8-35 distributes rapidly throughout the organism and is found in most organs but the brain and spleen, before being slowly eliminated (10-20 days). A similar pattern is observed after IP injection, following a brief latency period during which the polymer remains confined to the peritoneal cavity. Upon SC injection, A8-35 remains essentially confined to the point of injection, from which it is only slowly released. An interesting observation is that A8-35 tends to accumulate in fat pads, suggesting that it could be used to deliver anti-obesity drugs.


Subject(s)
Drug Delivery Systems , Organ Specificity/physiology , Polymers/administration & dosage , Polymers/pharmacokinetics , Propylamines/administration & dosage , Propylamines/pharmacokinetics , Adipose Tissue/metabolism , Animals , Female , Injections, Intraperitoneal , Injections, Intravenous , Injections, Subcutaneous , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Mice, Nude , Tissue Distribution
14.
Neuroimage ; 86: 164-71, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23933039

ABSTRACT

BACKGROUND: Atomoxetine (ATX), a drug for treatment of depression and ADHD, has a high affinity for the norepinephrine transporter (NET); however, our previous study showed it had a blocking effect similar to fluoxetine on binding of [(11)C]DASB, a selective serotonin transporter (SERT) ligand. Whether the therapeutic effects of ATX are due to inhibition of either or both transporters is not known. Here we report our comparative PET imaging studies with [(11)C]MRB (a NET ligand) and [(11)C]AFM (a SERT ligand) to evaluate in vivo IC50 values of ATX in monkeys. METHODS: Rhesus monkeys were scanned up to four times with each tracer with up to four doses of ATX. ATX or saline (placebo) infusion began 2h before each PET scan, lasting until the end of the 2-h scan. The final infusion rates were 0.01-0.12mg/kg/h and 0.045-1.054mg/kg/h for the NET and SERT studies, respectively. ATX plasma levels and metabolite-corrected arterial input functions were measured. Distribution volumes (VT) and IC50 values were estimated. RESULTS: ATX displayed dose-dependent occupancy on both NET and SERT, with a higher occupancy on NET: IC50 of 31±10 and 99±21ng/mL plasma for NET and SERT, respectively. At a clinically relevant dose (1.0-1.8mg/kg, approx. 300-600ng/mL plasma), ATX would occupy >90% of NET and >85% of SERT. This extrapolation assumes comparable free fraction of ATX in humans and non-human primates. CONCLUSION: Our data suggests that ATX at clinically relevant doses greatly occupies both NET and SERT. Thus, therapeutic modes of ATX action for treatment of depression and ADHD may be more complex than selective blockade of NET.


Subject(s)
Brain/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Propylamines/administration & dosage , Propylamines/pharmacokinetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Adrenergic Uptake Inhibitors/administration & dosage , Adrenergic Uptake Inhibitors/pharmacokinetics , Animals , Atomoxetine Hydrochloride , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/metabolism , Brain/drug effects , Depression/drug therapy , Depression/metabolism , Dose-Response Relationship, Drug , Macaca mulatta , Positron-Emission Tomography/methods , Tissue Distribution
15.
J Clin Psychopharmacol ; 34(1): 139-42, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24346747

ABSTRACT

Atomoxetine is a selective norepinephrine reuptake inhibitor indicated for the treatment of attention-deficit/hyperactivity disorder. Atomoxetine metabolism is mediated by CYP2D6 and CYP2C19. This study aimed to investigate the effect of the CYP2C19 genetic polymorphism on the pharmacokinetics of atomoxetine and its metabolites, 4-hydroxyatomoxetine and N-desmethylatomoxetine. A single 40-mg oral dose of atomoxetine was administered to 40 subjects with different CYP2C19 genotypes (all participants carried the CYP2D6*1/*10 genotype). Concentrations of atomoxetine and its metabolites were analyzed using high-performance liquid chromatography with tandem mass spectrometry in plasma samples that were collected up to 24 hours after drug intake. For atomoxetine, the CYP2C19 poor metabolizer (PM) group showed significantly increased maximum plasma concentration and AUC0-∞ (area under the plasma concentration-time curve from 0 to infinity) and decreased apparent oral clearance compared with samples of the CYP2C19 extensive metabolizer (EM) and intermediate metabolizer (IM) groups (P < 0.001 for all). The half-life of atomoxetine in the CYP2C19PM group was also significantly longer than in the other genotype groups (P < 0.01 for CYP2C19EM and P < 0.05 for CYP2C19IM groups). The maximum plasma concentration and AUC 0-∞ of 4-hydroxyatomoxetine were significantly higher in the CYP2C19PM group compared with those in the CYP2C19EM and IM groups (P < 0.001 for CYP2C19EM and P < 0.05 for CYP2C19IM, respectively), whereas the corresponding values for N-desmethylatomoxetine in the CYP2C19PM group were significantly lower than those in the 2 genotype groups (P < 0.001 for both genotype groups). These results suggest that the genetic polymorphisms of CYP2C19 significantly affect the pharmacokinetics of atomoxetine.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Polymorphism, Genetic , Propylamines/pharmacokinetics , Administration, Oral , Adrenergic Uptake Inhibitors/administration & dosage , Adrenergic Uptake Inhibitors/blood , Area Under Curve , Atomoxetine Hydrochloride , Biotransformation , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2C19 , Genotype , Half-Life , Humans , Male , Metabolic Clearance Rate , Pharmacogenetics , Phenols/blood , Phenotype , Phenyl Ethers/blood , Propylamines/administration & dosage , Propylamines/blood , Tandem Mass Spectrometry
16.
Drug Res (Stuttg) ; 63(11): 564-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23812961

ABSTRACT

To evaluate the bioequivalence of a new formulation of atomoxetine hydrochloride (CAS 82248-59-7) capsules (test) and an available branded capsules (reference) after administration of a single 40 mg dose, randomized, open-label, 2-period crossover study was conducted in 22 healthy male Chinese subjects with a 1-week wash-out period. This study was designed for/the Honglin Pharmaceutical Co. Ltd and contracted to be done by the Beijing Anding Hospital in order to satisfy Chinese regulatory requirements to allow marketing of this generic product and performed according to the criteria of SFDA. Blood samples were collected before and 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 9, 12, 16 and 24 h after drug administration. Plasma concentrations were determined by high-performance liquid chromatography (HPLC) with UV detection. A non-compartmental method was used to calculate the pharmacokinetic parameters and evaluate bioequivalence of the 2 formulations. The 90% confidence interval (CI) of the ratios (test/reference) of atomoxetine for AUC0-24, AUC0-∞ and Cmax were 100.9% (93.6-108.8%), 103.1% (95.1-111.7%) and 105.2% (92.8-119.4%), respectively, which fell within the interval of 80-125% and 75-133%. No clinically significant changes or abnormalities were noted in laboratory data and vital signs. From these results it can be concluded that the test formulation of atomoxetine capsules met the regulatory criterion for bioequivalence to the reference formulation.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacokinetics , Propylamines/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Atomoxetine Hydrochloride , Capsules , Cross-Over Studies , Fasting , Humans , Male , Therapeutic Equivalency , Young Adult
17.
Br J Clin Pharmacol ; 75(2): 538-49, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22803597

ABSTRACT

AIM: The effects of atomoxetine (20 and 60 mg twice daily), 400 mg moxifloxacin and placebo on QT(c) in 131 healthy CYP2D6 poor metabolizer males were compared. METHODS: Atomoxetine doses were selected to result in plasma concentrations that approximated expected plasma concentrations at both the maximum recommended dose and at a supratherapeutic dose in CYP2D6 extensive metabolizers. Ten second electrocardiograms were obtained for time-matched baseline on days -2 and -1, three time points after dosing on day 1 for moxifloxacin and five time points on day 7 for atomoxetine and placebo. Maximum mean placebo-subtracted change from baseline model-corrected QT (QT(c)M) on day 7 was the primary endpoint. RESULTS: QT(c)M differences for atomoxetine 20 and 60 mg twice daily were 0.5 ms (upper bound of the one-sided 95% confidence interval 2.2 ms) and 4.2 ms (upper bound of the one-sided 95% confidence interval 6.0 ms), respectively. As plasma concentration of atomoxetine increased, a statistically significant increase in QT(c) was observed. The moxifloxacin difference from placebo met the a priori definition of non-inferiority. Maximum mean placebo-subtracted change from baseline QT(c)M for moxifloxacin was 4.8 ms and this difference was statistically significant. Moxifloxacin plasma concentrations were below the concentrations expected from the literature. However, the slope of the plasma concentration-QT(c) change observed was consistent with the literature. CONCLUSION: Atomoxetine was not associated with a clinically significant change in QT(c). However, a statistically significant increase in QT(c) was associated with increasing plasma concentrations.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Aza Compounds/pharmacology , Cytochrome P-450 CYP2D6/metabolism , Electrocardiography/drug effects , Heart Rate/drug effects , Propylamines/pharmacology , Quinolines/pharmacology , Topoisomerase II Inhibitors/pharmacology , Adrenergic Uptake Inhibitors/pharmacokinetics , Adult , Atomoxetine Hydrochloride , Aza Compounds/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Fluoroquinolones , Humans , Male , Moxifloxacin , Propylamines/pharmacokinetics , Quinolines/pharmacokinetics , Topoisomerase II Inhibitors/pharmacokinetics , Young Adult
18.
Bioorg Med Chem ; 21(1): 186-95, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23211968

ABSTRACT

The aim of this project was to synthesize and evaluate three novel fluorine-18 labeled derivatives of propargyl amine as potential PET radioligands to visualize monoamine oxidase B (MAO-B) activity. The three fluorinated derivatives of propargyl amine ((S)-1-fluoro-N,4-dimethyl-N-(prop-2-ynyl)-pent-4-en-2-amine (5), (S)-N-(1-fluoro-3-(furan-2-yl)propan-2-yl)-N-methylprop-2-yn-1-amine (10) and (S)-1-fluoro-N,4-dimethyl-N-(prop-2-ynyl)pentan-2-amine (15)) were synthesized in multi-step organic syntheses. IC(50) values for inhibition were determined for compounds 5, 10 and 15 in order to determine their specificity for binding to MAO-B. Compound 5 inhibited MAO-B with an IC(50) of 664 ± 48.08 nM. No further investigation was carried out with this compound. Compound 10 inhibited MAO-B with an IC(50) of 208.5 ± 13.44 nM and compound 15 featured an IC(50) of 131.5 ± 0.71 nM for its MAO-B inhibitory activity. None of the compounds inhibited MAO-A activity (IC(50) > 2 µM). The fluorine-18 labeled analogues of the two higher binding affinity compounds (10 and 15) (S)-N-(1-[(18)F]fluoro-3-(furan-2-yl)propan-2-yl)-N-methylprop-2-yn-1-amine (16) and (S)-1-[(18)F]fluoro-N,4-dimethyl-N-(prop-2-ynyl)pentan-2-amine (18) were both prepared from the corresponding precursors 9A, 9B and 14A, 14B by a one-step fluorine-18 nucleophilic substitution reaction. Autoradiography experiments on human postmortem brain tissue sections were performed with 16 and 18. Only compound 18 demonstrated a high selectivity for MAO-B over MAO-A and was, therefore, chosen for further examination by PET in a cynomolgus monkey. The initial uptake of 18 in the monkey brain was 250% SUV at 4 min post injection. The highest uptake of radioactivity was observed in the striatum and thalamus, regions with high MAO-B activity, whereas lower levels of radioactivity were detected in the cortex and cerebellum. The percentage of unchanged radioligand 18 was 30% in plasma at 90min post injection. In conclusion, compound 18 is a selective inhibitor of MAO-B in vitro and demonstrated a MAO-B specific binding pattern in vivo by PET in monkey. It can, therefore, be considered as a candidate for further investigation in human by PET.


Subject(s)
Brain/diagnostic imaging , Brain/enzymology , Fluorine Radioisotopes/analysis , Monoamine Oxidase/metabolism , Pargyline/analogs & derivatives , Propylamines/analysis , Animals , Autoradiography , Fluorine Radioisotopes/metabolism , Fluorine Radioisotopes/pharmacokinetics , Humans , Macaca fascicularis , Pargyline/analysis , Pargyline/metabolism , Pargyline/pharmacokinetics , Positron-Emission Tomography , Propylamines/metabolism , Propylamines/pharmacokinetics , Radiography
19.
J Addict Med ; 6(4): 265-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22987022

ABSTRACT

OBJECTIVES: Atomoxetine has been considered as an agonist replacement therapy for cocaine. We investigated the safety of the interaction of atomoxetine with cocaine and also whether cognitive function was affected by atomoxetine during short-term administration. METHODS: In a double-blind placebo-controlled inpatient study of 20 cocaine-dependent volunteers, participants received atomoxetine 80 mg daily followed by 100 mg daily for 5 days each. On the fourth and fifth day at each dose, cocaine (20 and 40 mg) was infused intravenously in sequential daily sessions. RESULTS: Preinfusion mean systolic pressures showed a small but statistically significant difference between placebo and both doses of atomoxetine. Preinfusion mean diastolic pressures were significant between placebo and atomoxetine 80 mg only. The diastolic pressure response to 40 mg cocaine was statistically significant only between the 80- and 100-mg atomoxetine doses. All electrocardiogram parameters were unchanged. Visual Analog Scale (VAS) scores for "bad effect" in the atomoxetine group were significantly higher at baseline, then declined, and for "likely to use" declined with atomoxetine treatment. On the Addiction Research Center Inventory, the atomoxetine group scored significantly lower on amphetamine, euphoria, and energy subscales (P < 0.0001). Other VAS descriptors, Brief Substance Craving Scale, Profile of Moods State, and Brief Psychiatric Rating Scale showed no differences. Atomoxetine did not affect cocaine pharmacokinetics. In tests of working memory, sustained attention, cognitive flexibility, and decision-making, atomoxetine improved performance on the visual n-back task. There were no differences in any pharmacokinetic parameters for cocaine with atomoxetine. CONCLUSIONS: Atomoxetine was tolerated safely by all participants. Certain cognitive improvements and a dampening effect on VAS scores after cocaine were observed, but should be weighed against small but significant differences in hemodynamic responses after atomoxetine.


Subject(s)
Adrenergic Uptake Inhibitors/administration & dosage , Adrenergic Uptake Inhibitors/adverse effects , Cocaine-Related Disorders/complications , Cocaine-Related Disorders/rehabilitation , Cocaine/adverse effects , Cognition/drug effects , Opiate Substitution Treatment/adverse effects , Propylamines/administration & dosage , Propylamines/adverse effects , Substance Abuse, Intravenous/rehabilitation , Adrenergic Uptake Inhibitors/pharmacokinetics , Adult , Affect/drug effects , Atomoxetine Hydrochloride , Attention Deficit Disorder with Hyperactivity/blood , Attention Deficit Disorder with Hyperactivity/rehabilitation , Blood Pressure/drug effects , Cocaine/agonists , Cocaine/pharmacokinetics , Cocaine-Related Disorders/blood , Comorbidity , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Drug Interactions , Electrocardiography/drug effects , Heart Rate/drug effects , Humans , Male , Middle Aged , Motivation/drug effects , Neuropsychological Tests , Opiate Substitution Treatment/methods , Propylamines/pharmacokinetics , Substance Abuse, Intravenous/blood , Young Adult
20.
Expert Opin Drug Metab Toxicol ; 8(5): 543-52, 2012 May.
Article in English | MEDLINE | ID: mdl-22413882

ABSTRACT

INTRODUCTION: Pharmacotherapy is frequently used to treat symptoms of attention-deficit/hyperactivity disorder (ADHD), the most common neurobehavioral disorder of childhood. The typically prescribed agents for ADHD have varying durations of effect and degrees of efficacy. The broad range of pharmacological treatments available allows for both single and combination therapies for achieving optimal therapeutic effects. Metabolic, toxicological, and safety information are critical for an informed evaluation of the risk/benefit considerations in prescribing practices. AREAS COVERED: This article focuses on the medications with current FDA approval for use in the treatment of ADHD in pediatric and adult populations. This review covers the stimulants (amphetamine and methylphenidate) and non-stimulants (atomoxetine, clonidine extended release, and guanfacine extended release) used to treat ADHD and presents an overview of their respective metabolic, toxicological, and safety features. A literature search and review of the relevant medications were carried out using the PubMed database up to November 2011. EXPERT OPINION: New trends in study design based on drug profiles include the use of adjuvant therapies and the inclusion of patients with comorbidities. The recent expansion of inclusion/exclusion criteria in pediatric clinical trials of ADHD allows for a more rigorous analysis of associated benefits and risks with the use of adjuvant therapy.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Drug-Related Side Effects and Adverse Reactions , Amphetamines/adverse effects , Amphetamines/pharmacokinetics , Atomoxetine Hydrochloride , Central Nervous System Stimulants/adverse effects , Central Nervous System Stimulants/pharmacokinetics , Clinical Trials as Topic , Clonidine/adverse effects , Clonidine/pharmacokinetics , Comorbidity , Drug Combinations , Guanfacine/adverse effects , Guanfacine/pharmacokinetics , Humans , Methylphenidate/adverse effects , Methylphenidate/pharmacokinetics , Propylamines/adverse effects , Propylamines/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...