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










Publication year range
2.
Neuropsychopharmacology ; 41(11): 2647-57, 2016 10.
Article in English | MEDLINE | ID: mdl-27109624

ABSTRACT

The A118G single-nucleotide polymorphism (SNP rs1799971) in the µ-opioid receptor gene, OPRM1, has been much studied in relation to alcohol use disorders. The reported effects of allelic variation at this SNP on alcohol-related behaviors, and on opioid receptor antagonist treatments, have been inconsistent. We investigated the pharmacogenetic interaction between A118G variation and the effects of two µ-opioid receptor antagonists in a clinical lab setting. Fifty-six overweight and moderate-heavy drinkers were prospectively stratified by genotype (29 AA homozygotes, 27 carriers of at least 1 G allele) in a double-blind placebo-controlled, three-period crossover design with naltrexone (NTX; 25 mg OD for 2 days, then 50 mg OD for 3 days) and GSK1521498 (10 mg OD for 5 days). The primary end point was regional brain activation by the contrast between alcohol and neutral tastes measured using functional magnetic resonance imaging (fMRI). Secondary end points included other fMRI contrasts, subjective responses to intravenous alcohol challenge, and food intake. GSK1521498 (but not NTX) significantly attenuated fMRI activation by appetitive tastes in the midbrain and amygdala. GSK1521498 (and NTX to a lesser extent) significantly affected self-reported responses to alcohol infusion. Both drugs reduced food intake. Across all end points, there was less robust evidence for significant effects of OPRM1 allelic variation, or for pharmacogenetic interactions between genotype and drug treatment. These results do not support strong modulatory effects of OPRM1 genetic variation on opioid receptor antagonist attenuation of alcohol- and food-related behaviors. However, they do support further investigation of GSK1521498 as a potential therapeutic for alcohol use and eating disorders.


Subject(s)
Drinking/drug effects , Drinking/genetics , Indans/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Polymorphism, Single Nucleotide/genetics , Receptors, Opioid, mu/genetics , Triazoles/pharmacology , Adolescent , Adult , Aged , Alanine/genetics , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Eating/drug effects , Eating/genetics , Female , Glycine/genetics , Humans , Male , Middle Aged , Pharmacogenomic Testing , Receptors, Opioid, mu/agonists , Young Adult
3.
Psychopharmacology (Berl) ; 232(18): 3431-41, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26141191

ABSTRACT

RATIONALE: Using the drinking-in-the-dark (DID) model, we compared the effects of a novel mu-opioid receptor antagonist, GSK1521498, with naltrexone, a licensed treatment of alcohol dependence, on ethanol consumption in mice. OBJECTIVE: We test the ability of GSK1521498 to reduce alcohol consumption and compare its intrinsic efficacy to that of naltrexone by comparing the two drugs at doses matched for equivalent receptor occupancy. METHODS: Thirty-six C57BL/6J mice were tested in a DID procedure. In 2-day cycles, animals experienced one baseline, injection-free session, and one test session when they received two injections, one of test drug and one placebo. All animals received GSK1521498 (0, 0.1, 1 and 3 mg/kg, i.p., 30 min pre-treatment) and naltrexone (0, 0.1, 1 and 3 mg/kg, s.c. 10 min pre-treatment) in a cross-over design. Receptor occupancies following the same doses were determined ex vivo in separate groups by autoradiography, using [3H]DAMGO. Binding in the region of interest was measured integrally by computer-assisted microdensitometry and corrected for non-specific binding. RESULTS: Both GSK1521498 and naltrexone dose-dependently decreased ethanol consumption. When drug doses were matched for 70-75% receptor occupancy, GSK1521498 3 mg/kg, i.p., caused a 2.5-fold greater reduction in alcohol consumption than naltrexone 0.1 mg/kg, s.c. Both GSK1521498 and naltrexone significantly reduced sucrose consumption at a dose of 1 mg/kg but not 0.1 mg/kg. In a test of conditioned taste aversion, GSK1521498 (3 mg/kg) reduced sucrose consumption 24 h following exposure to a conditioning injection. CONCLUSIONS: Both opioid receptor antagonists reduced alcohol consumption but GK1521498 has higher intrinsic efficacy than naltrexone.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Depressants/administration & dosage , Ethanol/administration & dosage , Indans/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Triazoles/pharmacology , Alcohol Drinking , Alcoholism/drug therapy , Animals , Autoradiography , Cross-Over Studies , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Male , Mice , Mice, Inbred C57BL , Radiopharmaceuticals , Self Administration , Tritium
4.
Psychopharmacology (Berl) ; 232(1): 305-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24973897

ABSTRACT

RATIONALE: The novel opioid receptor antagonist, GSK1421498, has been shown to attenuate reward-driven compulsive behaviours, such as stimulant drug seeking or binge eating, in animals and humans. Here, we report new data on the receptor pharmacology of GSK121498, in comparison to naltrexone, naloxone, 6-ß-naltrexol and nalmefene. OBJECTIVES: To determine whether the novel opioid antagonist, GSK1521498, is an orthosteric or allosteric antagonist at the µ opioid receptor (MOPr) and whether it has neutral antagonist or inverse agonist properties. METHODS: A combination of radioligand binding assays and [(35)S]GTPγS binding assays was employed. RESULTS: GSK1521498 completely displaced [(3)H]naloxone binding to MOPr and did not alter the rate of [(3)H]naloxone dissociation from MOPr observations compatible with it binding to the orthosteric site on MOPr. GSK1521498 exhibited inverse agonism when MOPr was overexpressed but not when the level of MOPr expression was low. In parallel studies under conditions of high receptor expression density, naloxone, naltrexone, 6-ß-naltrexol and nalmefene exhibited partial agonism, not inverse agonism as has been reported previously for naloxone and naltrexone. In brain tissue from mice receiving a prolonged morphine pre-treatment, GSK1521498 exhibited slight inverse agonism. CONCLUSIONS: Differences between GSK1521498 and naltrexone in their effects on compulsive reward seeking are arguably linked to the more selective and complete MOPr antagonism of GSK1521498 versus the partial MOPr agonism of naltrexone. GSK1521498 is also pharmacologically differentiated by its inverse agonist efficacy at high levels of MOPr expression, but this may be less likely to contribute to behavioural differentiation at patho-physiological levels of expression.


Subject(s)
Compulsive Behavior/metabolism , Indans/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Reward , Triazoles/pharmacology , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , CHO Cells , Compulsive Behavior/drug therapy , Cricetinae , Cricetulus , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Humans , Indans/metabolism , Indans/therapeutic use , Ligands , Male , Mice , Morphine/metabolism , Morphine/pharmacology , Morphine/therapeutic use , Naloxone/metabolism , Naloxone/pharmacology , Naloxone/therapeutic use , Naltrexone/analogs & derivatives , Naltrexone/metabolism , Naltrexone/pharmacology , Naltrexone/therapeutic use , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Protein Binding/physiology , Receptors, Opioid, mu/metabolism , Treatment Outcome , Triazoles/metabolism , Triazoles/therapeutic use
5.
PLoS One ; 8(11): e74133, 2013.
Article in English | MEDLINE | ID: mdl-24244264

ABSTRACT

It has been suggested that the BDNF Val66Met polymorphism modulates episodic memory performance via effects on hippocampal neural circuitry. However, fMRI studies have yielded inconsistent results in this respect. Moreover, very few studies have examined the effect of met allele load on activation of memory circuitry. In the present study, we carried out a comprehensive analysis of the effects of the BDNF polymorphism on brain responses during episodic memory encoding and retrieval, including an investigation of the effect of met allele load on memory related activation in the medial temporal lobe. In contrast to previous studies, we found no evidence for an effect of BDNF genotype or met load during episodic memory encoding. Met allele carriers showed increased activation during successful retrieval in right hippocampus but this was contrast-specific and unaffected by met allele load. These results suggest that the BDNF Val66Met polymorphism does not, as previously claimed, exert an observable effect on neural systems underlying encoding of new information into episodic memory but may exert a subtle effect on the efficiency with which such information can be retrieved.


Subject(s)
Alleles , Brain-Derived Neurotrophic Factor/genetics , Memory, Episodic , Nerve Net/physiology , Polymorphism, Genetic , Adult , Brain-Derived Neurotrophic Factor/metabolism , Double-Blind Method , Female , Humans , Middle Aged
6.
J Clin Pharmacol ; 53(10): 1078-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23934621

ABSTRACT

The mu-opioid system has a key role in hedonic and motivational processes critical to substance addiction. However, existing mu-opioid antagonists have had limited success as anti-addiction treatments. GSK1521498 is a selective and potent mu-opioid antagonist being developed for the treatment of overeating and substance addictions. In this study, 28 healthy participants were administered single doses of GSK1521498 20 mg, ethanol 0.5 g/kg body weight, or both in combination, in a double blind placebo controlled four-way crossover design. The primary objective was to determine the risk of significant adverse pharmacodynamic and pharmacokinetic (PK) interactions. The effects of GSK1521498 on hedonic and consummatory responses to alcohol and the attentional processing of alcohol-related stimuli, and their modulation by the OPRM1 A118G polymorphism were also explored. GSK1521498 20 mg was well tolerated alone and in combination with ethanol. There were mild transient effects of GSK1521498 on alertness and mood that were greater when it was combined with ethanol. These effects were not of clinical significance. There were no effects of GSK1521498 on reaction time, hedonic or consummatory responses. These findings provide encouraging safety and PK data to support continued development of GSK1521498 for the treatment of alcohol addiction.


Subject(s)
Ethanol/administration & dosage , Indans/administration & dosage , Triazoles/administration & dosage , Adult , Affect/drug effects , Cross-Over Studies , Double-Blind Method , Drug Interactions , Ethanol/adverse effects , Ethanol/pharmacokinetics , Humans , Indans/adverse effects , Indans/pharmacokinetics , Middle Aged , Polymorphism, Genetic , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Triazoles/adverse effects , Triazoles/pharmacokinetics , Young Adult
7.
Biol Psychiatry ; 73(9): 887-94, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23245760

ABSTRACT

BACKGROUND: Binge eating is associated with obesity and has been conceptualized as "food addiction." However, this view has received only inconsistent support in humans, and limited evidence relates key neurocircuitry to the disorder. Moreover, relatively few studies have used pharmacologic functional magnetic resonance imaging to probe the underlying basis of altered eating behaviors. METHODS: In a double-blind, placebo-controlled, parallel group study, we explored the effects of a potent mu-opioid receptor antagonist, GSK1521498, in obese individuals with moderate binge eating. Subjects were tested during a baseline placebo run-in period and retested after 28-days of drug (n = 21) or placebo (n = 21) treatment. Using functional magnetic resonance imaging and behavioral measures, we determined the drug's effects on brain responses to food images and, separately, on motivation to expend energy to view comparable images. RESULTS: Compared with placebo, GSK1521498 was associated with a significant reduction in pallidum/putamen responses to pictures of high-calorie food and a reduction in motivation to view images of high-calorie food. Intriguingly, although motivational responding was reduced, subjective liking for the same images actually increased following drug treatment. CONCLUSIONS: Stimulus-specific putamen/pallidal responses in obese people with binge eating are sensitive to altered mu-opioid function. This neuromodulation was accompanied by reductions in motivational responding, as measured by grip force, although subjective liking responses to the same stimuli actually increased. As well as providing evidence for a link between the opioid system and food-related behavior in binge-eating obese individuals, these results support a dissociation across measures of motivation and liking associated with food-related stimuli in these individuals.


Subject(s)
Binge-Eating Disorder/physiopathology , Brain/drug effects , Indans/pharmacology , Narcotic Antagonists/pharmacology , Obesity/physiopathology , Receptors, Opioid, mu/antagonists & inhibitors , Triazoles/pharmacology , Adolescent , Adult , Binge-Eating Disorder/complications , Brain/physiopathology , Brain Mapping , Double-Blind Method , Feeding Behavior/drug effects , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Motivation/drug effects , Motivation/physiology , Obesity/complications
8.
Psychopharmacology (Berl) ; 224(4): 501-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22752384

ABSTRACT

RATIONALE: Translational research implicates the mu opioid neurochemical system in hedonic processing, but its role in dissociable high-level cognitive functions is not well understood. Binge-eating represents a useful model of 'behavioural addiction' for exploring this issue. OBJECTIVE: The aim of this study was to objectively assess the cognitive effects of a mu opioid receptor antagonist in obese individuals with binge-eating symptoms. METHODS: Adults with moderate to severe binge-eating and body mass index ≥30 kg/m² received 4 weeks of treatment with a mu opioid receptor antagonist (GSK1521498) 2 or 5 mg per day, or placebo, in a double-blind randomised parallel design. Neuropsychological assessment was undertaken at baseline and endpoint to quantify processing bias for food stimuli (visual dot probe with 500- and 2,000-ms stimulus presentations and food Stroop tasks) and other distinct cognitive functions (N-back working memory, sustained attention, and power of attention tasks). RESULTS: GSK1521498 5 mg/day significantly reduced attentional bias for food cues on the visual dot probe task versus placebo (p = 0.042), with no effects detected on other cognitive tasks (all p > 0.10). The effect on attentional bias was limited to the longer stimulus duration condition in the higher dose cohort alone. CONCLUSIONS: These findings support a central role for mu opioid receptors in aspects of attentional processing of food cues but militate against the notion of major modulatory influences of mu opioid receptors in working memory and sustained attention. The findings have implications for novel therapeutic directions and suggest that the role of different opioid receptors in cognition merits further research.


Subject(s)
Binge-Eating Disorder/drug therapy , Indans/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Triazoles/pharmacology , Adolescent , Adult , Attention , Binge-Eating Disorder/psychology , Cognition/drug effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Indans/administration & dosage , Male , Middle Aged , Narcotic Antagonists/administration & dosage , Neuropsychological Tests , Severity of Illness Index , Time Factors , Triazoles/administration & dosage , Young Adult
9.
J Clin Pharmacol ; 52(4): 464-74, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21610207

ABSTRACT

Endogenous opioids and µ-opioid receptors have been linked to hedonic and rewarding aspects of palatable food intake. The authors examined the safety, pharmacokinetic, and pharmacodynamic profile of GSK1521498, a µ-opioid receptor inverse agonist that is being investigated primarily for the treatment of overeating behavior in obesity. In healthy participants, GSK1521498 oral solution and capsule formulations were well tolerated up to a dose of 100 mg. After single doses (10-150 mg), the maximum concentration (C(max)) and area under the curve (AUC) in plasma increased in a dose-proportional manner. GSK1521498 selectively reduced sensory hedonic ratings of high-sugar and high-fat dairy products and caloric intake of high-fat/high-sucrose snack foods. These findings provide encouraging data in support of the development of GSK1521498 for the treatment of disorders of maladaptive ingestive behavior or compulsive consumption.


Subject(s)
Eating/drug effects , Food Preferences/drug effects , Indans/pharmacology , Receptors, Opioid, mu/agonists , Triazoles/pharmacology , Administration, Oral , Adult , Area Under Curve , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Inverse Agonism , Humans , Indans/adverse effects , Indans/pharmacokinetics , Male , Single-Blind Method , Triazoles/adverse effects , Triazoles/pharmacokinetics
10.
J Clin Pharmacol ; 52(10): 1456-67, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22162534

ABSTRACT

The endogenous opioid system and µ-opioid receptors in particular have been demonstrated to play a fundamental role in hedonic and motivational behaviors reinforced by rewards. In healthy participants, the authors examined the multiple-dose safety, pharmacokinetic, and secondary pharmacodynamic profile of GSK1521498, a µ-opioid receptor inverse agonist that is being developed for treatment of disorders of compulsive consumption. Clinically relevant doses of GSK1521498 (2, 5, and 10 mg) following once-daily administration for 10 days, were well tolerated with no clinically relevant changes in vital signs, chemistry, or hematologic parameters and with a favorable neuropsychiatric profile. Following oral administration, median first time to reach maximum observed plasma concentration for GSK1521498 occurred 2 to 5 hours after dosing, with individual values ranging from 1 to 8 hours. Systemic exposure to GSK1521498 (area under the curve [0-∞] and maximum observed plasma concentration) increased in a slightly greater-than-dose-proportional manner, and steady-state plasma levels were reached within approximately 7 days. The secondary pharmacodynamic effects of GSK1521498 on cognition and pain threshold and tolerance were dose related, with mild to moderate impairments in measures of attention and reductions of pressure pain threshold and tolerance at the highest dose. These findings provide encouraging safety, tolerability, and pharmacokinetic data in support of the continued clinical development of GSK1521498.


Subject(s)
Indans/administration & dosage , Receptors, Opioid, mu/agonists , Triazoles/administration & dosage , Adult , Affect/drug effects , Cognition/drug effects , Double-Blind Method , Drug Administration Schedule , Female , Hot Temperature , Humans , Indans/adverse effects , Indans/pharmacokinetics , Male , Middle Aged , Pain Threshold/drug effects , Pressure , Triazoles/adverse effects , Triazoles/pharmacokinetics , Young Adult
11.
Br J Clin Pharmacol ; 69(3): 252-61, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20233196

ABSTRACT

AIMS: To estimate the pharmacologically active dose range of a new investigational compound S-0139, a selective endothelin A (ET(A)) receptor antagonist, in man, and to examine the duration of its pharmacodynamic effect. METHODS: Venous occlusion plethysmography was performed to assess changes in forearm blood flow following intra-brachial administration of endothelin-1 (ET-1). ET(A) antagonists have been shown to block ET-1-induced vasoconstriction in this model. The study was conducted in three parts: (1) a pilot study to explore dose-response (dose range 0.08-13.33 microg kg(-1) min(-1)), (2) a randomized study to confirm dose-response (placebo, 2.5, 6.67 and 15 microg kg(-1) min(-1)), and (3) a delayed administration study (15.7 microg kg(-1) min(-1)) to explore the duration of the pharmacodynamic effect. In all studies a 3-h infusion of S-0139 was given and during the last 90 min of the infusion, ET-1 was infused concurrently for 90 min. In study (3) a second ET-1 infusion was given starting 3 h after completion of the first. RESULTS: Intravenously administered S-0139 resulted in significant inhibition of ET-1-induced vasoconstriction in the forearm (plasma concentration 800-2000 ng ml(-1)). In the delayed administration study, the same extent of inhibition was still present when ET-1 was administered 3 h after the end of infusion of S-0139, even though the S-0139 plasma concentrations (mean 17 ng ml(-1)) were well below pharmacologically active concentrations as determined in studies 1 and 2. CONCLUSIONS: S-0139 dose-dependently blocks ET-1-mediated vasoconstriction in the forearm and has a prolonged duration of effect beyond that expected from its pharmacokinetic profile.


Subject(s)
Caffeic Acids , Endothelin A Receptor Antagonists , Forearm/blood supply , Oleanolic Acid/analogs & derivatives , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Adolescent , Adult , Blood Pressure/drug effects , Caffeic Acids/pharmacology , Double-Blind Method , Endothelin-1 , Humans , Infusions, Intravenous , Male , Middle Aged , Oleanolic Acid/pharmacology , Plethysmography , Single-Blind Method , Young Adult
12.
Br J Clin Pharmacol ; 66(3): 396-404, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18662287

ABSTRACT

AIM: To characterize the effects of lamotrigine on QT interval in healthy subjects. METHODS: Healthy subjects received a single oral dose of moxifloxacin (400 mg) or placebo in crossover design, followed by a dose-escalating regimen of lamotrigine (n = 76) over a 77-day period, or matched placebo (n = 76). Blood samples were taken for determination of moxifloxacin and lamotrigine concentrations and digital 12-lead ECGs were recorded. The relationships between individual QT values and respective individual moxifloxacin or lamotrigine concentrations were explored using population pharmacokinetic-pharmacodynamic (PK-PD) modelling. RESULTS: Moxifloxacin was associated with a maximum mean increase from baseline in QTcF of 14.81 ms [90% confidence interval (CI) 13.50, 16.11] 2.5 h after dosing. Steady-state exposure to lamotrigine (50, 150 or 200 mg b.d.) was not associated with an increase in QTc interval. Small reductions in QTcF (maximum mean difference from placebo -7.48 ms, 90% CI -10.49, -4.46) and small increases in heart rate (maximum mean difference from placebo 5.94 bpm, 90% CI 3.81, 8.06) were observed with lamotrigine 200 mg b.d. vs. placebo. No effect of lamotrigine on QRS duration or blood pressure was observed. No outliers with QTcF > 450 ms, or with an increase from baseline of >60 ms were observed in the lamotrigine group. PK-PD modelling indicated statistically significant decreases in individually corrected QT intervals for lamotrigine and statistically significant increases in individually corrected QT intervals for moxifloxacin over the concentration ranges studied. CONCLUSIONS: Therapeutic doses of lamotrigine (50-200 mg b.d.) were not associated with QT prolongation in healthy subjects.


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Aza Compounds/administration & dosage , Electrocardiography/methods , Long QT Syndrome/chemically induced , Quinolines/administration & dosage , Triazines/administration & dosage , Administration, Oral , Adolescent , Adult , Anti-Arrhythmia Agents/pharmacokinetics , Aza Compounds/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fluoroquinolones , Heart Rate/drug effects , Humans , Lamotrigine , Male , Middle Aged , Moxifloxacin , Quinolines/pharmacokinetics , Treatment Outcome , Triazines/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL