Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Psychopharmacol ; 36(5): 614-625, 2022 05.
Article in English | MEDLINE | ID: mdl-35212235

ABSTRACT

BACKGROUND: Intranasal esketamine demonstrates rapid improvement of depressive symptoms. However, transient adverse effects (dissociation, sedation and dizziness) may occur, which could impact driving performance. AIMS: To evaluate the effects of 84 mg intranasal esketamine on driving performance in unipolar major depressive disorder (MDD) or persistent depressive disorder (PDD) patients. METHODS: The study consisted of two parts. Part A was a single-blind, double-dummy, randomized three-period, cross-over study to compare effects of esketamine versus placebo on next morning driving, 18 ± 2 h post-treatment. Alcohol was administered to demonstrate assay sensitivity. In Part B, same-day driving, 6 ± 0.5 hours post-treatment, was assessed during twice weekly esketamine administration for 3 weeks. Twenty-seven patients with mild-to-moderate MDD or PDD without psychotic features completed a 100 km on-the-road driving test on a public highway in normal traffic. The primary outcome was standard deviation of lateral position (SDLP; cm; weaving of car). RESULTS: In Part A, alcohol impaired driving performance compared to placebo: Least-square means (95% CI), p-value for delta SDLP (cm) compared with placebo: (ΔSDLP = + 1.83 (1.03; 2.62), p < 0.001), whereas esketamine did not: (ΔSDLP = -0.23 (-1.04; 0.58), p = 0.572). In Part B, weekly driving tests showed no differences between placebo baseline SDLP and after esketamine administration over 3 weeks: Day 11: (ΔSDLP = -0.96 (-3.72; 1.81), p = 0.493), Day 18: (ΔSDLP = -0.56 (-3.33; 2.20), p = 0.686) and Day 25: (ΔSDLP = -1.05 (-3.82; 1.71), p = 0.451). CONCLUSIONS: In this study, esketamine did not impair on-road driving performance the next morning following a single dose, or on same day after repeated administration.


Subject(s)
Automobile Driving , Depressive Disorder, Major , Antidepressive Agents/adverse effects , Cross-Over Studies , Depressive Disorder, Major/drug therapy , Double-Blind Method , Humans , Ketamine , Psychomotor Performance , Single-Blind Method
2.
J Clin Psychiatry ; 81(3)2020 04 28.
Article in English | MEDLINE | ID: mdl-32316080

ABSTRACT

OBJECTIVE: To evaluate long-term safety and efficacy of esketamine nasal spray plus a new oral antidepressant (OAD) in patients with treatment-resistant depression (TRD). METHODS: This phase 3, open-label, multicenter, long-term (up to 1 year) study was conducted between October 2015 and October 2017. Patients (≥ 18 years) with TRD (DSM-5 diagnosis of major depressive disorder and nonresponse to ≥ 2 OAD treatments) were enrolled directly or transferred from a short-term study (patients aged ≥ 65 years). Esketamine nasal spray (28-mg, 56-mg, or 84-mg) plus new OAD was administered twice a week in a 4-week induction (IND) phase and weekly or every-other-week for patients who were responders and entered a 48-week optimization/maintenance (OP/MAINT) phase. RESULTS: Of 802 enrolled patients, 86.2% were direct-entry and 13.8% were transferred-entry; 580 (74.5%) of 779 patients who entered the IND phase completed the phase, and 150 (24.9%) of 603 who entered the OP/MAINT phase completed the phase. Common treatment-emergent adverse events (TEAEs) were dizziness (32.9%), dissociation (27.6%), nausea (25.1%), and headache (24.9%). Seventy-six patients (9.5%) discontinued esketamine due to TEAEs. Fifty-five patients (6.9%) experienced serious TEAEs. Most TEAEs occurred on dosing days, were mild or moderate in severity, and resolved on the same day. Two deaths were reported; neither was considered related to esketamine. Cognitive performance generally either improved or remained stable postbaseline. There was no case of interstitial cystitis or respiratory depression. Treatment-emergent dissociative symptoms were transient and generally resolved within 1.5 hours postdose. Montgomery-Åsberg Depression Rating Scale total score decreased during the IND phase, and this reduction persisted during the OP/MAINT phase (mean [SD] change from baseline of respective phase to endpoint: IND, -16.4 [8.76]; OP/MAINT, 0.3 [8.12]). CONCLUSIONS: Long-term esketamine nasal spray plus new OAD therapy had a manageable safety profile, and improvements in depression appeared to be sustained in patients with TRD. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02497287.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Treatment-Resistant/drug therapy , Ketamine/therapeutic use , Administration, Intranasal , Administration, Oral , Adolescent , Adult , Aged , Antidepressive Agents/administration & dosage , Cognition/drug effects , Drug Therapy, Combination , Female , Humans , Ketamine/administration & dosage , Ketamine/adverse effects , Male , Middle Aged , Nasal Sprays , Young Adult
3.
Int J Neuropsychopharmacol ; 23(7): 426-433, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32270176

ABSTRACT

BACKGROUND: Esketamine nasal spray was recently approved for treatment-resistant depression. The current analysis evaluated the impact of symptom-based treatment frequency changes during esketamine treatment on clinical outcomes. METHODS: This is a post-hoc analysis of an open-label, long-term (up to 1 year) study of esketamine in patients with treatment-resistant depression (SUSTAIN 2). During a 4-week induction phase, 778 patients self-administered esketamine twice weekly plus a new oral antidepressant daily. In responders (≥50% reduction in Montgomery-Åsberg Depression Rating Scale total score from baseline), esketamine treatment frequency was thereafter decreased during an optimization/maintenance phase to weekly for 4 weeks and then adjusted to the lowest frequency (weekly or every other week) that maintained remission (Montgomery-Åsberg Depression Rating Scale ≤ 12) based on a study-defined algorithm. The relationship between treatment frequency and symptom response, based on clinically meaningful change in Clinical Global Impression-Severity score, was subsequently evaluated 4 weeks after treatment frequency adjustments in the optimization/maintenance phase. RESULTS: Among 580 responders treated with weekly esketamine for the first 4 weeks in the optimization/maintenance phase (per protocol), 26% continued to improve, 50% maintained clinical benefit, and 24% worsened. Thereafter, when treatment frequency could be reduced from weekly to every other week, 19% further improved, 49% maintained benefit, and 32% worsened. For patients no longer in remission after treatment frequency reduction, an increase (every other week to weekly) resulted in 47% improved, 43% remained unchanged, and 10% worsened. CONCLUSIONS: These findings support individualization of esketamine nasal spray treatment frequency to optimize treatment response in real-world clinical practice. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02497287.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Treatment-Resistant/drug therapy , Ketamine/therapeutic use , Administration, Intranasal , Adolescent , Adult , Aged , Algorithms , Antidepressive Agents/administration & dosage , Clinical Trials, Phase III as Topic , Data Interpretation, Statistical , Depressive Disorder, Major/drug therapy , Double-Blind Method , Female , Humans , Ketamine/administration & dosage , Male , Middle Aged , Patient Compliance , Psychiatric Status Rating Scales , Treatment Outcome , Young Adult
4.
Curr Protoc Pharmacol ; 82(1): e42, 2018 09.
Article in English | MEDLINE | ID: mdl-30129249

ABSTRACT

Imaging neuronal activity in awake behaving mice with miniature fluorescence microscopes requires the implementation of a variety of procedures. Surgeries are performed to gain access to the cell population of interest and to implant microscope components. After a recovery period, mice are trained to exhibit a desired behavior. Finally, neuronal activity is imaged and synchronized with that behavior. To take full advantage of the technology, selection of the calcium indicator and experimental design must be carefully considered. In this article, we explain the procedures and considerations that are critical for obtaining high-quality calcium imaging data. As an example, we describe how to utilize miniature fluorescence microscopy to image hippocampal place cell activity during linear track running in Thy1.GCaMP6f transgenic mice. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Place Cells/physiology , Animals , Behavior, Animal , Calcium/physiology , Hippocampus/cytology , Hippocampus/physiology , Mice, Transgenic , Microscopy, Fluorescence , Motor Activity
5.
Neuropsychopharmacology ; 43(13): 2586-2596, 2018 12.
Article in English | MEDLINE | ID: mdl-30026598

ABSTRACT

Emerging data continues to point towards a relationship between neuroinflammation and neuropsychiatric disorders. ATP-induced activation of P2X7 results in IL-1ß release causing neuroinflammation and microglial activation. This study describes the in-vitro and in-vivo neuropharmacology of a novel brain-penetrant P2X7 antagonist, JNJ-55308942, currently in clinical development. JNJ-55308942 is a high-affinity, selective, brain-penetrant (brain/plasma of 1) P2X7 functional antagonist. In human blood and in mouse blood and microglia, JNJ-55308942 attenuated IL-1ß release in a potent and concentration-dependent manner. After oral dosing, the compound exhibited both dose and concentration-dependent occupancy of rat brain P2X7 with an ED50 of 0.07 mg/kg. The P2X7 antagonist (3 mg/kg, oral) blocked Bz-ATP-induced brain IL-1ß release in conscious rats, demonstrating functional effects of target engagement in the brain. JNJ-55308942 (30 mg/kg, oral) attenuated LPS-induced microglial activation in mice, assessed at day 2 after a single systemic LPS injection (0.8 mg/kg, i.p.), suggesting a role for P2X7 in microglial activation. In a model of BCG-induced depression, JNJ-55308942 dosed orally (30 mg/kg), reversed the BCG-induced deficits of sucrose preference and social interaction, indicating for the first time a role of P2X7 in the BCG model of depression, probably due to the neuroinflammatory component induced by BCG inoculation. Finally, in a rat model of chronic stress induced sucrose intake deficit, JNJ-55308942 reversed the deficit with concurrent high P2X7 brain occupancy as measured by autoradiography. This body of data demonstrates that JNJ-55308942 is a potent P2X7 antagonist, engages the target in brain, modulates IL-1ß release and microglial activation leading to efficacy in two models of anhedonia in rodents.


Subject(s)
Anhedonia/drug effects , Disease Models, Animal , Drug Delivery Systems/methods , Inflammation Mediators/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, Purinergic P2X7/physiology , Anhedonia/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Purinergic P2X Receptor Antagonists/chemistry , Purinergic P2X Receptor Antagonists/therapeutic use , Pyridines/chemistry , Pyridines/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Rats, Wistar
6.
J Med Chem ; 61(1): 207-223, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29211470

ABSTRACT

A single pot dipolar cycloaddition reaction/Cope elimination sequence was developed to access novel 1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridine P2X7 antagonists that contain a synthetically challenging chiral center. The structure-activity relationships of the new compounds are described. Two of these compounds, (S)-(2-fluoro-3-(trifluoromethyl)phenyl)(1-(5-fluoropyrimidin-2-yl)-6-methyl-1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridin-5-yl)methanone (compound 29) and (S)-(3-fluoro-2-(trifluoromethyl)pyridin-4-yl)(1-(5-fluoropyrimidin-2-yl)-6-methyl-1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridin-5-yl)methanone (compound 35), were found to have robust P2X7 receptor occupancy at low doses in rat with ED50 values of 0.06 and 0.07 mg/kg, respectively. Compound 35 had notable solubility compared to 29 and showed good tolerability in preclinical species. Compound 35 was chosen as a clinical candidate for advancement into phase I clinical trials to assess safety and tolerability in healthy human subjects prior to the initiation of proof of concept studies for the treatment of mood disorders.


Subject(s)
Drug Design , Purinergic P2X Receptor Antagonists/chemical synthesis , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Receptors, Purinergic P2X7/metabolism , Animals , Cycloaddition Reaction , Dogs , Humans , Male , Mice , Models, Molecular , Molecular Conformation , Purinergic P2X Receptor Antagonists/chemistry , Purinergic P2X Receptor Antagonists/pharmacokinetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Rats , Stereoisomerism , Tissue Distribution
7.
J Med Chem ; 60(11): 4559-4572, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28493698

ABSTRACT

The synthesis and preclinical characterization of novel 4-(R)-methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridines that are potent and selective brain penetrant P2X7 antagonists are described. Optimization efforts based on previously disclosed unsubstituted 6,7-dihydro-4H-triazolo[4,5-c]pyridines, methyl substituted 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyrazines, and several other series lead to the identification of a series of 4-(R)-methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridines that are selective P2X7 antagonists with potency at the rodent and human P2X7 ion channels. These novel P2X7 antagonists have suitable physicochemical properties, and several analogs have an excellent pharmacokinetic profile, good partitioning into the CNS and show robust in vivo target engagement after oral dosing. Improvements in metabolic stability led to the identification of JNJ-54175446 (14) as a candidate for clinical development. The drug discovery efforts and strategies that resulted in the identification of the clinical candidate are described herein.


Subject(s)
Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/pharmacology , Receptors, Purinergic P2X7/drug effects , Animals , Biological Availability , Humans , Purinergic P2X Receptor Antagonists/pharmacokinetics
8.
Front Neurosci ; 10: 53, 2016.
Article in English | MEDLINE | ID: mdl-26973444

ABSTRACT

Prolonged exposure to abnormally high calcium concentrations is thought to be a core mechanism underlying hippocampal damage in epileptic patients; however, no prior study has characterized calcium activity during seizures in the live, intact hippocampus. We have directly investigated this possibility by combining whole-brain electroencephalographic (EEG) measurements with microendoscopic calcium imaging of pyramidal cells in the CA1 hippocampal region of freely behaving mice treated with the pro-convulsant kainic acid (KA). We observed that KA administration led to systematic patterns of epileptiform calcium activity: a series of large-scale, intensifying flashes of increased calcium fluorescence concurrent with a cluster of low-amplitude EEG waveforms. This was accompanied by a steady increase in cellular calcium levels (>5 fold increase relative to the baseline), followed by an intense spreading calcium wave characterized by a 218% increase in global mean intensity of calcium fluorescence (n = 8, range [114-349%], p < 10(-4); t-test). The wave had no consistent EEG phenotype and occurred before the onset of motor convulsions. Similar changes in calcium activity were also observed in animals treated with 2 different proconvulsant agents, N-methyl-D-aspartate (NMDA) and pentylenetetrazol (PTZ), suggesting the measured changes in calcium dynamics are a signature of seizure activity rather than a KA-specific pathology. Additionally, despite reducing the behavioral severity of KA-induced seizures, the anticonvulsant drug valproate (VA, 300 mg/kg) did not modify the observed abnormalities in calcium dynamics. These results confirm the presence of pathological calcium activity preceding convulsive motor seizures and support calcium as a candidate signaling molecule in a pathway connecting seizures to subsequent cellular damage. Integrating in vivo calcium imaging with traditional assessment of seizures could potentially increase translatability of pharmacological intervention, leading to novel drug screening paradigms and therapeutics designed to target and abolish abnormal patterns of both electrical and calcium excitation.

9.
J Pharmacol Exp Ther ; 354(3): 471-82, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26177655

ABSTRACT

Dual orexin receptor antagonists have been shown to promote sleep in various species, including humans. Emerging research indicates that selective orexin-2 receptor (OX2R) antagonists may offer specificity and a more adequate sleep profile by preserving normal sleep architecture. Here, we characterized JNJ-42847922 ([5-(4,6-dimethyl-pyrimidin-2-yl)-hexahydro-pyrrolo[3,4-c]pyrrol-2-yl]-(2-fluoro-6-[1,2,3]triazol-2-yl-phenyl)-methanone), a high-affinity/potent OX2R antagonist. JNJ-42847922 had an approximate 2-log selectivity ratio versus the human orexin-1 receptor. Ex vivo receptor binding studies demonstrated that JNJ-42847922 quickly occupied OX2R binding sites in the rat brain after oral administration and rapidly cleared from the brain. In rats, single oral administration of JNJ-42847922 (3-30 mg/kg) during the light phase dose dependently reduced the latency to non-rapid eye movement (NREM) sleep and prolonged NREM sleep time in the first 2 hours, whereas REM sleep was minimally affected. The reduced sleep onset and increased sleep duration were maintained upon 7-day repeated dosing (30 mg/kg) with JNJ-42847922, then all sleep parameters returned to baseline levels following discontinuation. Although the compound promoted sleep in wild-type mice, it had no effect in OX2R knockout mice, consistent with a specific OX2R-mediated sleep response. JNJ-42847922 did not increase dopamine release in rat nucleus accumbens or produce place preference in mice after subchronic conditioning, indicating that the compound lacks intrinsic motivational properties in contrast to zolpidem. In a single ascending dose study conducted in healthy subjects, JNJ-42847922 increased somnolence and displayed a favorable pharmacokinetic and safety profile for a sedative/hypnotic, thus emerging as a promising candidate for further clinical development for the treatment of insomnia.


Subject(s)
Orexin Receptor Antagonists , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep/drug effects , Animals , Binding Sites/drug effects , Brain/drug effects , Brain/metabolism , CHO Cells , Cell Line , Cricetulus , Dopamine/metabolism , HEK293 Cells , Humans , Hypnotics and Sedatives/pharmacology , Male , Mice , Mice, Knockout , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Sleep Initiation and Maintenance Disorders/metabolism , Sleep Stages/drug effects , Sleep, REM/drug effects , Zolpidem
10.
ACS Med Chem Lett ; 6(4): 450-4, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25893048

ABSTRACT

The preclinical characterization of novel phenyl(piperazin-1-yl)methanones that are histamine H3 receptor antagonists is described. The compounds described are high affinity histamine H3 antagonists. Optimization of the physical properties of these histamine H3 antagonists led to the discovery of several promising lead compounds, and extensive preclinical profiling aided in the identification of compounds with optimal duration of action for wake promoting activity. This led to the discovery of two development candidates for Phase I and Phase II clinical trials.

11.
PLoS One ; 9(11): e112068, 2014.
Article in English | MEDLINE | ID: mdl-25372144

ABSTRACT

Therapeutic drugs for cognitive and psychiatric disorders are often characterized by their molecular mechanism of action. Here we demonstrate a new approach to elucidate drug action on large-scale neuronal activity by tracking somatic calcium dynamics in hundreds of CA1 hippocampal neurons of pharmacologically manipulated behaving mice. We used an adeno-associated viral vector to express the calcium sensor GCaMP3 in CA1 pyramidal cells under control of the CaMKII promoter and a miniaturized microscope to observe cellular dynamics. We visualized these dynamics with and without a systemic administration of Zolpidem, a GABAA agonist that is the most commonly prescribed drug for the treatment of insomnia in the United States. Despite growing concerns about the potential adverse effects of Zolpidem on memory and cognition, it remained unclear whether Zolpidem alters neuronal activity in the hippocampus, a brain area critical for cognition and memory. Zolpidem, when delivered at a dose known to induce and prolong sleep, strongly suppressed CA1 calcium signaling. The rate of calcium transients after Zolpidem administration was significantly lower compared to vehicle treatment. To factor out the contribution of changes in locomotor or physiological conditions following Zolpidem treatment, we compared the cellular activity across comparable epochs matched by locomotor and physiological assessments. This analysis revealed significantly depressive effects of Zolpidem regardless of the animal's state. Individual hippocampal CA1 pyramidal cells differed in their responses to Zolpidem with the majority (∼ 65%) significantly decreasing the rate of calcium transients, and a small subset (3%) showing an unexpected and significant increase. By linking molecular mechanisms with the dynamics of neural circuitry and behavioral states, this approach has the potential to contribute substantially to the development of new therapeutics for the treatment of CNS disorders.


Subject(s)
CA1 Region, Hippocampal , Calcium/metabolism , GABA-A Receptor Agonists/pharmacology , Molecular Imaging/methods , Optical Imaging/methods , Pyramidal Cells , Pyridines/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Locomotion/drug effects , Locomotion/physiology , Mice , Microscopy, Fluorescence/methods , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Zolpidem
12.
J Pharmacol Exp Ther ; 351(3): 628-41, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25271258

ABSTRACT

In the central nervous system, the ATP-gated Purinergic receptor P2X ligand-gated ion channel 7 (P2X7) is expressed in glial cells and modulates neurophysiology via release of gliotransmitters, including the proinflammatory cytokine interleukin (IL)-1ß. In this study, we characterized JNJ-42253432 [2-methyl-N-([1-(4-phenylpiperazin-1-yl)cyclohexyl]methyl)-1,2,3,4-tetrahydroisoquinoline-5-carboxamide] as a centrally permeable (brain-to-plasma ratio of 1), high-affinity P2X7 antagonist with desirable pharmacokinetic and pharmacodynamic properties for in vivo testing in rodents. JNJ-42253432 is a high-affinity antagonist for the rat (pKi 9.1 ± 0.07) and human (pKi 7.9 ± 0.08) P2X7 channel. The compound blocked the ATP-induced current and Bz-ATP [2'(3')-O-(4-benzoylbenzoyl)adenosine-5'-triphosphate tri(triethylammonium)]-induced release of IL-1ß in a concentration-dependent manner. When dosed in rats, JNJ-42253432 occupied the brain P2X7 channel with an ED50 of 0.3 mg/kg, corresponding to a mean plasma concentration of 42 ng/ml. The compound blocked the release of IL-1ß induced by Bz-ATP in freely moving rat brain. At higher doses/exposure, JNJ-42253432 also increased serotonin levels in the rat brain, which is due to antagonism of the serotonin transporter (SERT) resulting in an ED50 of 10 mg/kg for SERT occupancy. JNJ-42253432 reduced electroencephalography spectral power in the α-1 band in a dose-dependent manner; the compound also attenuated amphetamine-induced hyperactivity. JNJ-42253432 significantly increased both overall social interaction and social preference, an effect that was independent of stress induced by foot-shock. Surprisingly, there was no effect of the compound on either neuropathic pain or inflammatory pain behaviors. In summary, in this study, we characterize JNJ-42253432 as a novel brain-penetrant P2X7 antagonist with high affinity and selectivity for the P2X7 channel.


Subject(s)
Central Nervous System Agents/metabolism , Central Nervous System Agents/pharmacology , Isoquinolines/metabolism , Isoquinolines/pharmacology , Piperazines/metabolism , Piperazines/pharmacology , Purinergic P2X Receptor Antagonists/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/metabolism , Animals , Animals, Newborn , Central Nervous System Agents/therapeutic use , Dose-Response Relationship, Drug , Humans , Isoquinolines/therapeutic use , Male , Mice , Neuralgia/drug therapy , Neuralgia/metabolism , Piperazines/therapeutic use , Purinergic P2X Receptor Antagonists/therapeutic use , Rats , Rats, Sprague-Dawley
13.
Front Neurosci ; 8: 107, 2014.
Article in English | MEDLINE | ID: mdl-24904253

ABSTRACT

The orexin/hypocretin neuropeptides are produced by a cluster of neurons within the lateral posterior hypothalamus and participate in neuronal regulation by activating their receptors (OX1 and OX2 receptors). The orexin system projects widely through the brain and functions as an interface between multiple regulatory systems including wakefulness, energy balance, stress, reward, and emotion. Recent studies have demonstrated that orexins and glutamate interact at the synaptic level and that orexins facilitate glutamate actions. We tested the hypothesis that orexins modulate glutamate signaling via OX1 receptors by monitoring levels of glutamate in frontal cortex of freely moving mice using enzyme coated biosensors under inhibited OX1 receptor conditions. MK-801, an NMDA receptor antagonist, was administered subcutaneously (0.178 mg/kg) to indirectly disinhibit pyramidal neurons and therefore increase cortical glutamate release. In wild-type mice, pretreatment with the OX1 receptor antagonist GSK-1059865 (10 mg/kg S.C.) which had no effect by itself, significantly attenuated the cortical glutamate release elicited by MK-801. OX1 receptor knockout mice had a blunted glutamate release response to MK-801 and exhibited about half of the glutamate release observed in wild-type mice in agreement with the data obtained with transient blockade of OX1 receptors. These results indicate that pharmacological (transient) or genetic (permanent) inhibition of the OX1 receptor similarly interfere with glutamatergic function in the cortex. Selectively targeting the OX1 receptor with an antagonist may normalize hyperglutamatergic states and thus may represent a novel therapeutic strategy for the treatment of various psychiatric disorders associated with hyperactive states.

14.
Br J Pharmacol ; 170(3): 624-40, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23889535

ABSTRACT

BACKGROUND AND PURPOSE: An increasing body of evidence suggests that the purinergic receptor P2X, ligand-gated ion channel, 7 (P2X7) in the CNS may play a key role in neuropsychiatry, neurodegeneration and chronic pain. In this study, we characterized JNJ-47965567, a centrally permeable, high-affinity, selective P2X7 antagonist. EXPERIMENTAL APPROACH: We have used a combination of in vitro assays (calcium flux, radioligand binding, electrophysiology, IL-1ß release) in both recombinant and native systems. Target engagement of JNJ-47965567 was demonstrated by ex vivo receptor binding autoradiography and in vivo blockade of Bz-ATP induced IL-1ß release in the rat brain. Finally, the efficacy of JNJ-47965567 was tested in standard models of depression, mania and neuropathic pain. KEY RESULTS: JNJ-47965567 is potent high affinity (pKi 7.9 ± 0.07), selective human P2X7 antagonist, with no significant observed speciation. In native systems, the potency of the compound to attenuate IL-1ß release was 6.7 ± 0.07 (human blood), 7.5 ± 0.07 (human monocytes) and 7.1 ± 0.1 (rat microglia). JNJ-47965567 exhibited target engagement in rat brain, with a brain EC50 of 78 ± 19 ng·mL(-1) (P2X7 receptor autoradiography) and functional block of Bz-ATP induced IL-1ß release. JNJ-47965567 (30 mg·kg(-1) ) attenuated amphetamine-induced hyperactivity and exhibited modest, yet significant efficacy in the rat model of neuropathic pain. No efficacy was observed in forced swim test. CONCLUSION AND IMPLICATIONS: JNJ-47965567 is centrally permeable, high affinity P2X7 antagonist that can be used to probe the role of central P2X7 in rodent models of CNS pathophysiology.


Subject(s)
Brain/drug effects , Niacinamide/analogs & derivatives , Piperazines/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Analgesics/metabolism , Analgesics/pharmacology , Animals , Antidepressive Agents/pharmacology , Antimanic Agents/pharmacology , Behavior, Animal/drug effects , Binding, Competitive , Bipolar Disorder/metabolism , Bipolar Disorder/prevention & control , Bipolar Disorder/psychology , Blood-Brain Barrier/metabolism , Brain/metabolism , Calcium Signaling/drug effects , Capillary Permeability , Cell Line , Depression/metabolism , Depression/prevention & control , Depression/psychology , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Humans , Interleukin-1beta/metabolism , Macaca , Male , Mice , Mice, Inbred C57BL , Neuralgia/metabolism , Neuralgia/prevention & control , Neuralgia/psychology , Niacinamide/metabolism , Niacinamide/pharmacology , Piperazines/metabolism , Protein Binding , Purinergic P2X Receptor Antagonists/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X7/metabolism , Time Factors , Transfection
15.
J Pharmacol Exp Ther ; 342(2): 429-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22570363

ABSTRACT

In rodents 5-hydroxytryptamine type 7 (5-HT(7)) receptor blockade has been shown to be effective in models of depression and to increase the latency to rapid eye movement (REM) sleep and decrease REM duration. In the clinic, the REM sleep reduction observed with many antidepressants may serve as a biomarker. We report here the preclinical and clinical evaluation of a 5-HT(7) receptor antagonist, (3-(4-chlorophenyl)-1,4,5,6,7,8-hexahydro-1-(phenylmethyl)pyrazolo[3,4-d]azepine 2-hydroxy-1,2,3-propanetricarboxylate) (JNJ-18038683). In rodents, JNJ-18038683 increased the latency to REM sleep and decreased REM duration, and this effect was maintained after repeated administration for 7 days. The compound was effective in the mouse tail suspension test. JNJ-18038683 enhanced serotonin transmission, antidepressant-like behavior, and REM sleep suppression induced by citalopram in rodents. In healthy human volunteers JNJ-18038683 prolonged REM latency and reduced REM sleep duration, demonstrating that the effect of 5-HT(7) blockade on REM sleep translated from rodents to humans. Like in rats, JNJ-18038683 enhanced REM sleep suppression induced by citalopram in humans, although a drug-drug interaction could not be ruled out. In a double-blind, active, and placebo-controlled clinical trial in 225 patients suffering from major depressive disorder, neither treatment with pharmacologically active doses of JNJ-18038683 or escitalopram separated from placebo, indicating a failed study lacking assay sensitivity. Post hoc analyses using an enrichment window strategy, where all the efficacy data from sites with an implausible high placebo response [placebo group Montgomery-Åsberg Depression Rating Scale (MADRS) < = 12] and from sites with no placebo response (MADRS > = 28) are removed, there was a clinically meaningful difference between JNJ-18038683 and placebo. Further clinical studies are required to characterize the potential antidepressant efficacy of JNJ-18038683.


Subject(s)
Antidepressive Agents/pharmacology , Azepines/pharmacology , Depressive Disorder, Major/drug therapy , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacology , Sleep, REM/drug effects , Tricarboxylic Acids/pharmacology , Adolescent , Adult , Animals , Antidepressive Agents/therapeutic use , Azepines/therapeutic use , Cell Line, Transformed , Citalopram/pharmacology , Cohort Studies , Cross-Over Studies , Depressive Disorder, Major/metabolism , Double-Blind Method , Female , HEK293 Cells , Hindlimb Suspension/methods , Humans , Hypothermia/drug therapy , Hypothermia/metabolism , Hypothermia/physiopathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Serotonin Antagonists/therapeutic use , Tricarboxylic Acids/therapeutic use , Young Adult
16.
PLoS One ; 6(6): e20210, 2011.
Article in English | MEDLINE | ID: mdl-21701689

ABSTRACT

The role of 5-HT7 receptor has been demonstrated in various animal models of mood disorders; however its function in cognition remains largely speculative. This study evaluates the effects of SB-269970, a selective 5-HT7 antagonist, in a translational model of working memory deficit and investigates whether it modulates cortical glutamate and/or dopamine neurotransmission in rats. The effect of SB-269970 was evaluated in the delayed non-matching to position task alone or in combination with MK-801, a non-competitive NMDA receptor antagonist, and, in separate experiments, with scopolamine, a non-selective muscarinic antagonist. SB-269970 (10 mg/kg) significantly reversed the deficits induced by MK-801 (0.1 mg/kg) but augmented the deficit induced by scopolamine (0.06 mg/kg). The ability of SB-269970 to modulate MK-801-induced glutamate and dopamine extracellular levels was separately evaluated using biosensor technology and microdialysis in the prefrontal cortex of freely moving rats. SB-269970 normalized MK-801 -induced glutamate but not dopamine extracellular levels in the prefrontal cortex. Rat plasma and brain concentrations of MK-801 were not affected by co-administration of SB-269970, arguing for a pharmacodynamic rather than a pharmacokinetic mechanism. These results indicate that 5-HT7 receptor antagonists might reverse cognitive deficits associated with NMDA receptor hypofunction by selectively normalizing glutamatergic neurotransmission.


Subject(s)
Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Memory Disorders/drug therapy , Memory, Short-Term/drug effects , Receptors, Serotonin/metabolism , Synaptic Transmission/drug effects , Animals , Cerebral Cortex/drug effects , Dizocilpine Maleate/pharmacokinetics , Dizocilpine Maleate/therapeutic use , Male , Memory Disorders/metabolism , Phenols/pharmacokinetics , Phenols/therapeutic use , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacokinetics , Serotonin Antagonists/therapeutic use , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
17.
Psychopharmacology (Berl) ; 214(4): 829-41, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21086115

ABSTRACT

RATIONALE: A few recent studies suggest that brain histamine levels and signaling via H(3) receptors play an important role in modulation of alcohol stimulation and reward in rodents. OBJECTIVE: The present study characterized the effects of a novel, selective, and brain penetrant H(3) receptor antagonist (JNJ-39220675) on the reinforcing effects of alcohol in rats. METHODS: The effect of JNJ-39220675 on alcohol intake and alcohol relapse-like behavior was evaluated in selectively bred alcohol-preferring (P) rats using the standard two-bottle choice method. The compound was also tested on operant alcohol self administration in non-dependent rats and on alcohol-induced ataxia using the rotarod apparatus. In addition, alcohol-induced dopamine release in the nucleus accumbens was tested in freely moving rats. RESULTS: Subcutaneous administration of the selective H(3) receptor antagonist dose-dependently reduced both alcohol intake and preference in alcohol-preferring rats. JNJ-39220675 also reduced alcohol preference in the same strain of rats following a 3-day alcohol deprivation. The compound significantly and dose-dependently reduced alcohol self-administration without changing saccharin self-administration in alcohol non-dependent rats. Furthermore, the compound did not change the ataxic effects of alcohol, alcohol elimination rate, nor alcohol-induced dopamine release in nucleus accumbens. CONCLUSIONS: These results indicate that blockade of H(3) receptor should be considered as a new attractive mechanism for the treatment of alcoholism.


Subject(s)
Alcoholism/drug therapy , Azepines/therapeutic use , Histamine H3 Antagonists/therapeutic use , Pyridines/therapeutic use , Alcoholism/metabolism , Alcoholism/psychology , Animals , Autoradiography , Azepines/pharmacology , Behavior, Animal/drug effects , Brain/metabolism , Histamine H3 Antagonists/pharmacokinetics , Histamine H3 Antagonists/pharmacology , Injections, Subcutaneous , Male , Microdialysis , Molecular Structure , Motor Activity/drug effects , Protein Binding , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Histamine H3/metabolism , Reinforcement, Psychology , Self Administration
18.
Psychopharmacology (Berl) ; 215(1): 191-203, 2011 May.
Article in English | MEDLINE | ID: mdl-21181123

ABSTRACT

RATIONALE: Orexin-1 receptor antagonists have been shown to block the reinforcing effects of drugs of abuse and food. However, whether blockade of orexin-2 receptor has similar effects has not been determined. We have recently described the in vitro and in vivo effects of JNJ-10397049, a selective and brain penetrant orexin-2 receptor antagonist. OBJECTIVE: The goal of these studies was to evaluate whether systemic administration of JNJ-10397049 blocks the rewarding effects of ethanol and reverses ethanol withdrawal in rodents. As a comparison, SB-408124, a selective orexin-1 receptor antagonist, was also evaluated. METHODS: Rats were trained to orally self-administer ethanol (8% v/v) or saccharin (0.1% v/v) under a fixed-ratio 3 schedule of reinforcement. A separate group of rats received a liquid diet of ethanol (8% v/v) and withdrawal signs were evaluated 4 h after ethanol discontinuation. In addition, ethanol-induced increases in extracellular dopamine levels in the nucleus accumbens were tested. In separate experiments, the acquisition, expression, and reinstatement of conditioned place preference (CPP) were evaluated in mice. RESULTS: Our results indicate that JNJ-10397049 (1, 3, and 10 mg/kg, sc) dose-dependently reduced ethanol self-administration without changing saccharin self-administration, dopamine levels, or withdrawal signs in rats. Treatment with JNJ-10397049 (10 mg/kg, sc) attenuated the acquisition, expression, and reinstatement of ethanol CPP and ethanol-induced hyperactivity in mice. Surprisingly, SB-408124 (3, 10 and 30 mg/kg, sc) did not have any effect in these procedures. CONCLUSIONS: Collectively, these results indicate, for the first time, that blockade of orexin-2 receptors is effective in reducing the reinforcing effects of ethanol.


Subject(s)
Behavior, Addictive/prevention & control , Behavior, Animal/drug effects , Conditioning, Operant/drug effects , Dioxanes/therapeutic use , Ethanol/administration & dosage , Phenylurea Compounds/therapeutic use , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , Alcoholism/metabolism , Alcoholism/prevention & control , Alcoholism/psychology , Animals , Behavior, Addictive/psychology , Dioxanes/administration & dosage , Dioxanes/pharmacology , Disease Models, Animal , Male , Mice , Mice, Inbred DBA , Motor Activity/drug effects , Orexin Receptors , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacology , Rats , Rats, Wistar , Reinforcement Schedule , Reinforcement, Psychology , Self Administration
19.
J Neurochem ; 115(2): 475-82, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20722970

ABSTRACT

Neuropeptide S (NPS) is known to produce anxiolytic-like effects and facilitate extinction of conditioned fear. Catecholaminergic neurotransmission in the medial prefrontal cortex (mPFC) has been suggested to be crucially involved in these brain functions. In the current study, we investigated the effect of NPS on the release of dopamine and serotonin in the mPFC by in vivo microdialysis in rats. Central administration of NPS dose-dependently enhanced extracellular levels of dopamine and its major metabolite 3,4-dihydroxyphenylacetic acid, with maximal effects lasting up to 120 min. In contrast, no effect on serotonergic neurotransmission was detected. Dopamine release in the mPFC has been previously linked to modulation of anxiety states and fear extinction. The present results may thus provide a physiological and anatomical basis for the reported effects of NPS on these behaviors.


Subject(s)
Dopamine/metabolism , Neuropeptides/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Synaptic Transmission/drug effects , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Area Under Curve , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Hydroxyindoleacetic Acid/metabolism , Male , Microdialysis/methods , Rats , Rats, Sprague-Dawley , Serotonin/metabolism
20.
Bioorg Med Chem Lett ; 20(14): 4210-4, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20561786

ABSTRACT

The pre-clinical characterization of novel aryloxypyridine amides that are histamine H(3) receptor antagonists is described. These compounds are high affinity histamine H(3) ligands that penetrate the CNS and occupy the histamine H(3) receptor in rat brain. Several compounds were extensively profiled pre-clinically leading to the identification of two compounds suitable for nomination as development candidates.


Subject(s)
Azepines/pharmacology , Histamine H3 Antagonists/pharmacology , Pyridines/pharmacology , Amides/chemistry , Animals , Azepines/chemistry , Drug Evaluation, Preclinical , Pyridines/chemistry , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...