Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Sci Rep ; 11(1): 7700, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833333

ABSTRACT

GABAA-α5 subunit-containing receptors have been shown to play a key modulatory role in cognition and represent a promising drug target for cognitive dysfunction, as well as other disorders. Here we report on the preclinical and early clinical profile of a novel GABAA-α5 selective negative allosteric modulator (NAM), basmisanil, which progressed into Phase II trials for intellectual disability in Down syndrome and cognitive impairment associated with schizophrenia. Preclinical pharmacology studies showed that basmisanil is the most selective GABAA-α5 receptor NAM described so far. Basmisanil bound to recombinant human GABAA-α5 receptors with 5 nM affinity and more than 90-fold selectivity versus α1, α2, and α3 subunit-containing receptors. Moreover, basmisanil inhibited GABA-induced currents at GABAA-α5 yet had little or no effect at the other receptor subtypes. An in vivo occupancy study in rats showed dose-dependent target engagement and was utilized to establish the plasma exposure to receptor occupancy relationship. At estimated receptor occupancies between 30 and 65% basmisanil attenuated diazepam-induced spatial learning impairment in rats (Morris water maze), improved executive function in non-human primates (object retrieval), without showing anxiogenic or proconvulsant effects in rats. During the Phase I open-label studies, basmisanil showed good safety and tolerability in healthy volunteers at maximum GABAA-α5 receptor occupancy as confirmed by PET analysis with the tracer [11C]-Ro 15-4513. An exploratory EEG study provided evidence for functional activity of basmisanil in human brain. Therefore, these preclinical and early clinical studies show that basmisanil has an ideal profile to investigate potential clinical benefits of GABAA-α5 receptor negative modulation.


Subject(s)
GABA-A Receptor Agonists/pharmacology , Receptors, GABA-A/drug effects , Allosteric Regulation , Animals , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Female , HEK293 Cells , Healthy Volunteers , Humans , Learning/drug effects , Macaca fascicularis , Positron-Emission Tomography , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Xenopus laevis
2.
Neuropsychopharmacology ; 44(2): 295-305, 2019 01.
Article in English | MEDLINE | ID: mdl-30120410

ABSTRACT

The pituitary neuropeptide oxytocin promotes social behavior, and is a potential adjunct therapy for social deficits in schizophrenia and autism. Oxytocin may mediate pro-social effects by modulating monoamine release in limbic and cortical areas, which was investigated herein using in vivo microdialysis, after establishing a dose that did not produce accompanying sedative or thermoregulatory effects that could concomitantly influence behavior. The effects of oxytocin (0.03-0.3 mg/kg subcutaneous) on locomotor activity, core body temperature, and social behavior (social interaction and ultrasonic vocalizations) were examined in adult male Lister-hooded rats, using selective antagonists to determine the role of oxytocin and vasopressin V1a receptors. Dopamine and serotonin efflux in the prefrontal cortex and nucleus accumbens of conscious rats were assessed using microdialysis. 0.3 mg/kg oxytocin modestly reduced activity and caused hypothermia but only the latter was attenuated by the V1a receptor antagonist, SR49059 (1 mg/kg intraperitoneal). Oxytocin at 0.1 mg/kg, which did not alter activity and had little effect on temperature, significantly attenuated phencyclidine-induced hyperactivity and increased social interaction between unfamiliar rats without altering the number or pattern of ultrasonic vocalizations. In the same rats, oxytocin (0.1 mg/kg) selectively elevated dopamine overflow in the nucleus accumbens, but not prefrontal cortex, without influencing serotonin efflux. Systemic oxytocin administration attenuated phencyclidine-induced hyperactivity and increased pro-social behavior without decreasing core body temperature and selectively enhanced nucleus accumbens dopamine release, consistent with activation of mesocorticolimbic circuits regulating associative/reward behavior being involved. This highlights the therapeutic potential of oxytocin to treat social behavioral deficits seen in psychiatric disorders such as schizophrenia.


Subject(s)
Behavior, Animal/drug effects , Dopamine/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Nucleus Accumbens/drug effects , Oxytocin/pharmacology , Phencyclidine/pharmacology , Social Behavior , Animals , Body Temperature/drug effects , Male , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Serotonin/metabolism
3.
Psychopharmacology (Berl) ; 232(21-22): 4059-83, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26070547

ABSTRACT

RATIONALE: Current treatments for schizophrenia have modest, if any, efficacy on cognitive dysfunction, creating a need for novel therapies. Their development requires predictive animal models. The N-methyl-D-aspartate (NMDA) hypothesis of schizophrenia indicates the use of NMDA antagonists, like subchronic phencyclidine (scPCP) to model cognitive dysfunction in adult animals. OBJECTIVES: The objective of this study was to assess the scPCP model by (1) reviewing published findings of scPCP-induced neurochemical changes and effects on cognitive tasks in adult rats and (2) comparing findings from a multi-site study to determine scPCP effects on standard and touchscreen cognitive tasks. METHODS: Across four research sites, the effects of scPCP (typically 5 mg/kg twice daily for 7 days, followed by at least 7-day washout) in adult male Lister Hooded rats were studied on novel object recognition (NOR) with 1-h delay, acquisition and reversal learning in Morris water maze and touchscreen-based visual discrimination. RESULTS: Literature findings showed that scPCP impaired attentional set-shifting (ASST) and NOR in several labs and induced a variety of neurochemical changes across different labs. In the multi-site study, scPCP impaired NOR, but not acquisition or reversal learning in touchscreen or water maze. Yet, this treatment regimen induced locomotor hypersensitivity to acute PCP until 13-week post-cessation. CONCLUSIONS: The multi-site study confirmed that scPCP impaired NOR and ASST only and demonstrated the reproducibility and usefulness of the touchscreen approach. Our recommendation, prior to testing novel therapeutics in the scPCP model, is to be aware that further work is required to understand the neurochemical changes and specificity of the cognitive deficits.


Subject(s)
Attention/drug effects , Cognition Disorders/chemically induced , Disease Models, Animal , Phencyclidine , Reversal Learning/drug effects , Schizophrenia/chemically induced , Animals , Cognition Disorders/complications , Cognition Disorders/drug therapy , Male , Rats , Schizophrenia/complications , Schizophrenia/drug therapy
4.
Handb Exp Pharmacol ; 228: 27-57, 2015.
Article in English | MEDLINE | ID: mdl-25977079

ABSTRACT

Cognition is a complex brain function that represents processes such as learning and memory, attention, working memory, and executive functions amongst others. Impairments in cognition are prevalent in many neuropsychiatric and neurological disorders with few viable treatment options. The development of new therapies is challenging, and poor efficacy in clinical development continues to be one of the most consistent reasons compounds fail to advance, suggesting that traditional animal models are not predictive of human conditions and behavior. An effort to improve the construct validity of neuropsychological testing across species with the intent of facilitating therapeutic development has been strengthening over recent years. With an emphasis on understanding the underlying biology, optimizing the use of appropriate systems (e.g., transgenic animals) to model targeted disease states, and incorporating non-rodent species (e.g., non-human primates) that may enable a closer comparison to humans, an improvement in the translatability of the results will be possible. This chapter focuses on some promising translational cognitive paradigms for use in rodents, non-human primates, and humans.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cognition/drug effects , Mental Disorders/drug therapy , Nootropic Agents/therapeutic use , Translational Research, Biomedical/methods , Animals , Attention/drug effects , Brain/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Cognitive Behavioral Therapy/methods , Disease Models, Animal , Electrocardiography , Executive Function/drug effects , Eye Movements , Humans , Magnetic Resonance Imaging , Memory/drug effects , Mental Disorders/diagnosis , Mental Disorders/physiopathology , Mental Disorders/psychology , Neuropsychological Tests , Predictive Value of Tests
5.
Neuropharmacology ; 86: 259-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25107588

ABSTRACT

The NK3 receptor is a GPCR that is prominently expressed in limbic areas of the brain, many of which have been implicated in schizophrenia. Phase II clinical trials in schizophrenia with two selective NK3 antagonists (osanetant and talnetant) have demonstrated significant improvement in positive symptoms. The objective of this study was to characterize the properties of a novel dual NK2/NK3 antagonist, RO5328673. [(3)H]RO5328673 bound to a single saturable site on hNK2, hNK3 and gpNK3 with high-affinity. RO5328673 acted as an insurmountable antagonist at both human and guinea-pig NK3 receptors in the [(3)H]IP accumulation assay. In binding kinetic analyses, [(3)H]RO5328673 had fast association and dissociation rates at hNK2 while it had a fast association rate and a remarkably slow dissociation rate at gp and hNK3. In electrophysiological recordings of gp SNpc, RO5328673 inhibited the senktide-induced potentiation of spontaneous activity of dopaminergic neurons with an insurmountable mechanism of action. RO5328673 exhibited in-vivo activity in gerbils, robustly reversing the senktide-induced locomotor activity. The TM2 residue gpNK3-A114(2.58) (threonine in all other species) was identified as the critical residue for the RO5328673's slower dissociation kinetics and stronger insurmountable mode of antagonism in the guinea-pig as compared to hNK3-T139(2.58). Using site-directed mutagenesis, [(3)H]RO5328673 binding and rhodopsin-based modeling, the important molecular determinants of the RO5328673-binding pocket of hNK3 were determined. A comparison of the RO5328673-binding pocket with that of osanetant showed that two antagonists have similar contact sides on hNK3 binding crevice except for three mutations V95L(1.42), Y247W(5.38), V255I(5.46), which behaved differently between interacting modes of two antagonists in hNK3.


Subject(s)
Carbamates/pharmacology , Neurotransmitter Agents/pharmacology , Piperidines/pharmacology , Receptors, Neurokinin-3/antagonists & inhibitors , Amino Acid Sequence , Animals , Antipsychotic Agents/pharmacology , Binding Sites , Carbamates/pharmacokinetics , Central Nervous System Agents/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Female , Gerbillinae , Guinea Pigs , HEK293 Cells , Humans , Locomotion/drug effects , Locomotion/physiology , Male , Models, Molecular , Molecular Sequence Data , Mutation , Neurotransmitter Agents/pharmacokinetics , Pars Compacta/drug effects , Pars Compacta/physiology , Peptide Fragments/pharmacology , Piperidines/pharmacokinetics , Receptors, Neurokinin-2/antagonists & inhibitors , Receptors, Neurokinin-2/metabolism , Receptors, Neurokinin-3/agonists , Receptors, Neurokinin-3/genetics , Receptors, Neurokinin-3/metabolism , Substance P/analogs & derivatives , Substance P/pharmacology , Tissue Culture Techniques
6.
Biol Psychiatry ; 75(3): 189-97, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23910948

ABSTRACT

BACKGROUND: Fragile X syndrome (FXS) is the most common genetic cause for intellectual disability. Fmr1 knockout (KO) mice are an established model of FXS. Chronic pharmacological inhibition of metabotropic glutamate receptor 5 (mGlu5) in these mice corrects multiple molecular, physiological, and behavioral phenotypes related to patients' symptoms. To better understand the pathophysiology of FXS and the effect of treatment, brain activity was analyzed using functional magnetic resonance imaging in relation to learning and memory performance. METHODS: Wild-type (WT) and Fmr1 KO animals receiving chronic treatment with the mGlu5 inhibitor CTEP or vehicle were evaluated consecutively for 1) learning and memory performance in the inhibitory avoidance and extinction test, and 2) for the levels of brain activity using continuous arterial spin labeling based functional magnetic resonance imaging. Neural activity patterns were correlated with cognitive performance using a multivariate regression analysis. Furthermore, mGlu5 receptor expression in brains of untreated mice was analyzed by autoradiography and saturation analysis using [(3)H]-ABP688. RESULTS: Chronic CTEP treatment corrected the learning deficit observed in Fmr1 KO mice in the inhibitory avoidance and extinction test and prevented memory extinction in WT and Fmr1 KO animals. Chronic CTEP treatment normalized perfusion in the amygdala and the lateral hypothalamus in Fmr1 KO mice and furthermore decreased perfusion in the hippocampus and increased perfusion in primary sensorimotor cortical areas. No significant differences in mGlu5 receptor expression levels between Fmr1 WT and KO mice were detected. CONCLUSIONS: Chronic mGlu5 inhibition corrected the learning deficits and partially normalized the altered brain activity pattern in Fmr1 KO mice.


Subject(s)
Brain/drug effects , Cognition/drug effects , Excitatory Amino Acid Antagonists/therapeutic use , Fragile X Syndrome/drug therapy , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Animals , Avoidance Learning/drug effects , Brain/blood supply , Disease Models, Animal , Electroshock/adverse effects , Excitatory Amino Acid Antagonists/pharmacokinetics , Extinction, Psychological/drug effects , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Imidazoles/therapeutic use , Mice , Mice, Knockout , Oximes/pharmacokinetics , Oxygen/blood , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Receptor, Metabotropic Glutamate 5/metabolism , Tritium/pharmacokinetics
7.
Psychopharmacology (Berl) ; 228(1): 31-42, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23397053

ABSTRACT

RATIONALE: Rearing rats in isolation from weaning is an established preclinical neurodevelopmental model which induces behavioural deficits with apparent translational relevance to some core symptoms of schizophrenia. OBJECTIVE: This study evaluated the ability of the atypical antipsychotic risperidone to reverse behavioural deficits induced by post-weaning social isolation of rat pups and to further characterise the predictive validity of this model. METHOD: Forty-five male Lister hooded rats were housed in groups of 3-4 (n = 16) or singly (n = 29) for 4 weeks immediately after weaning on postnatal day (PND) 22-24. On PND 51, novel cage-induced locomotor activity (LMA) was assessed to subdivide rats into groups balanced for behavioural response. On PNDs 58, 59, 65 and 72, rats received either vehicle (1 ml/kg; i.p.) or risperidone (0.2 or 0.5 mg/kg; i.p.) 30 min prior to testing in LMA, novel object discrimination (NOD), prepulse inhibition (PPI) of acoustic startle and conditioned emotional response (CER) learning paradigms, respectively. RESULTS: Isolation rearing had no effect on PPI, but produced LMA hyperactivity and impaired NOD and CER compared to group-housed controls. Risperidone caused a dose-dependent reduction in LMA, irrespective of rearing condition, but selectively reversed the NOD deficit in isolation-reared rats. Risperidone did not reverse the isolation rearing-induced CER deficit. CONCLUSIONS: Similar to its clinical profile, risperidone only partially reverses the schizophrenic symptomology; since it reversed some, but not all, of the learning and memory deficits induced by post-weaning isolation, the isolation rearing model may be useful to predict antipsychotic activity of novel therapeutic agents.


Subject(s)
Antipsychotic Agents/pharmacology , Memory Disorders/drug therapy , Risperidone/pharmacology , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/administration & dosage , Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Discrimination Learning/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Memory Disorders/etiology , Motor Activity/drug effects , Rats , Reflex, Startle/drug effects , Risperidone/administration & dosage , Schizophrenia/physiopathology , Social Isolation/psychology , Time Factors
8.
Neuropharmacology ; 64: 337-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22992331

ABSTRACT

Cognitive impairment, in particular of attention and memory, is often reported by patients suffering from major depressive disorder (MDD) and deficits in attention are part of the current diagnostic criteria of MDD. Objectively measured cognitive deficits associated with MDD have been described in many studies. They have been conceptualized as an integral facet and epiphenomenon of MDD. However, evidence accumulated in recent years has challenged this notion and demonstrated that in a subset of patients the degree of cognitive deficits cannot be accounted for by the severity of depression. In addition, in some patients cognitive deficits persist despite resolution of depressive symptomatology. It is plausible to assume that cognitive deficits contribute to functional impairment even though supportive data for such a relationship are lacking. However, the exact association between cognitive deficits and major depression and the clinical and neurobiological characteristics of patients with MDD in whom cognitive deficits seem partially or fully independent of the clinical manifestation of depressive symptoms remain poorly understood. This review focuses on objective measures of non-emotional cognitive deficits in MDD and discusses the presence of a subgroup of patients in whom these symptoms can be defined independently and in dissociation from the rest of the depressive symptomatology. The current understanding of brain circuits and molecular events implicated in cognitive impairment in MDD are discussed with an emphasis on the missing elements that could further define the specificity of cognitive impairment in MDD and lead to new therapeutics. Furthermore, this article presents in detail observations made in behavioral studies in rodents with potential novel therapeutic agents, such as negative allosteric modulators at the metabotropic glutamate receptor type 2/3 (mGlu2/3 NAM) which exhibit both cognitive enhancing and antidepressant properties. Such a compound, RO4432717, was tested in tests of short term memory (delayed match to position), cognitive flexibility (Morris water maze, reversal protocol), impulsivity and compulsivity (5-choice serial reaction time) and spontaneous object recognition in rodents, providing first evidence of a profile potentially relevant to address cognitive impairment in MDD. This article is part of a Special Issue entitled 'Cognitive Enhancers'.


Subject(s)
Antidepressive Agents/therapeutic use , Cognition Disorders/prevention & control , Depressive Disorder, Major/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , Molecular Targeted Therapy , Nootropic Agents/therapeutic use , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Antidepressive Agents/adverse effects , Antidepressive Agents/pharmacology , Cognition Disorders/chemically induced , Cognition Disorders/etiology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Agonists/therapeutic use , Excitatory Amino Acid Antagonists/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Interneurons/drug effects , Interneurons/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Nootropic Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism
9.
Neuron ; 74(1): 49-56, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22500629

ABSTRACT

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability. Previous studies have implicated mGlu5 in the pathogenesis of the disease, but a crucial unanswered question is whether pharmacological mGlu5 inhibition is able to reverse an already established FXS phenotype in mammals. Here we have used the novel, potent, and selective mGlu5 inhibitor CTEP to address this issue in the Fmr1 knockout mouse. Acute CTEP treatment corrects elevated hippocampal long-term depression, protein synthesis, and audiogenic seizures. Chronic treatment that inhibits mGlu5 within a receptor occupancy range of 81% ± 4% rescues cognitive deficits, auditory hypersensitivity, aberrant dendritic spine density, overactive ERK and mTOR signaling, and partially corrects macroorchidism. This study shows that a comprehensive phenotype correction in FXS is possible with pharmacological intervention starting in young adulthood, after development of the phenotype. It is of great interest how these findings may translate into ongoing clinical research testing mGlu5 inhibitors in FXS patients.


Subject(s)
Excitatory Amino Acid Antagonists/therapeutic use , Fragile X Syndrome/drug therapy , Imidazoles/therapeutic use , Pyridines/therapeutic use , Receptors, Metabotropic Glutamate/drug effects , Age Factors , Animals , Disease Models, Animal , Drug Administration Schedule , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Phenotype , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/metabolism
10.
Pharmacol Biochem Behav ; 99(2): 130-45, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21463652

ABSTRACT

The investigation of novel drug targets for treating cognitive impairments associated with neurological and psychiatric disorders remains a primary focus of study in central nervous system (CNS) research. Many promising new therapies are progressing through preclinical and clinical development, and offer the potential of improved treatment options for neurodegenerative diseases such as Alzheimer's disease (AD) as well as other disorders that have not been particularly well treated to date like the cognitive impairments associated with schizophrenia (CIAS). Among targets under investigation, cholinergic receptors have received much attention with several nicotinic agonists (α7 and α4ß2) actively in clinical trials for the treatment of AD, CIAS and attention deficit hyperactivity disorder (ADHD). Both glutamatergic and serotonergic (5-HT) agonists and antagonists have profound effects on neurotransmission and improve cognitive function in preclinical experiments with animals; some of these compounds are now in proof-of-concept studies in humans. Several histamine H3 receptor antagonists are in clinical development not only for cognitive enhancement, but also for the treatment of narcolepsy and cognitive deficits due to sleep deprivation because of their expression in brain sleep centers. Compounds that dampen inhibitory tone (e.g., GABA(A) α5 inverse agonists) or elevate excitatory tone (e.g., glycine transporter inhibitors) offer novel approaches for treating diseases such as schizophrenia, AD and Down syndrome. In addition to cell surface receptors, intracellular drug targets such as the phosphodiesterases (PDEs) are known to impact signaling pathways that affect long-term memory formation and working memory. Overall, there is a genuine need to treat cognitive deficits associated with many neuropsychiatric conditions as well as an increasingly aging population.


Subject(s)
Cognition Disorders/drug therapy , Nootropic Agents/pharmacology , Animals , Cognition Disorders/physiopathology , Glycine Plasma Membrane Transport Proteins/drug effects , Glycine Plasma Membrane Transport Proteins/physiology , Humans , Learning/drug effects , Learning/physiology , Memory/drug effects , Memory/physiology , Phosphodiesterase Inhibitors/pharmacology , Receptors, Cholinergic/drug effects , Receptors, Cholinergic/physiology , Receptors, Dopamine/drug effects , Receptors, Dopamine/physiology , Receptors, GABA/drug effects , Receptors, GABA/physiology , Receptors, Glutamate/drug effects , Receptors, Glutamate/physiology , Receptors, Histamine/drug effects , Receptors, Histamine/physiology , Receptors, Serotonin/drug effects , Receptors, Serotonin/physiology
11.
Expert Opin Ther Pat ; 21(5): 637-55, 2011 May.
Article in English | MEDLINE | ID: mdl-21417773

ABSTRACT

INTRODUCTION: The neurokinin 3 (NK(3)) receptor is a GPCR that has been shown to modulate monoaminergic systems within regions of the brain implicated in schizophrenia. Preclinical and Phase II clinical results of osanetant and talnetant in schizophrenic patients have indicated that NK(3) antagonists may provide significant improvement of the positive symptoms and cognitive impairment associated with this disorder. Recent findings have also indicated that neurokinin B (NKB)-NK(3) signaling plays a key role in the hypothalamic regulation of reproduction in humans. AREAS COVERED: This review article discusses the latest medicinal chemistry strategies used to derive novel NK(3) receptor antagonists which have been patented during the period 2005 - 2010. EXPERT OPINION: Since the report of a beneficial effect of osanetant in schizophrenic patients, major pharmaceutical companies have been involved in this field, leading to a very large number of patent applications disclosed. Nevertheless, only three NK(3) selective antagonists entered into Phase II, but were then terminated for various reasons. Currently, the main challenge to move forward a selective NK(3) antagonist into the clinic would be to define a safety margin between the desired therapeutic effect and the effect on testosterone levels. The involvement of NKB-NK(3) signaling in reproduction in humans may also lead to new exciting indications, such as treatment for sex steroid-sensitive cancers of breast and prostate.


Subject(s)
Patents as Topic , Receptors, Neurokinin-3/antagonists & inhibitors , Amino Acid Sequence , Animals , Clinical Trials as Topic , Humans , Luteinizing Hormone/blood , Molecular Sequence Data , Neoplasms, Hormone-Dependent/drug therapy , Receptors, Neurokinin-3/chemistry , Receptors, Neurokinin-3/physiology , Reproduction , Schizophrenia/drug therapy , Testosterone/blood
12.
Bioorg Med Chem Lett ; 20(23): 6969-74, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20971004

ABSTRACT

This study completes a series of papers devoted to the characterization of the non-competitive mGluR2/3 antagonist properties of 1,3-dihydro-benzo[b][1,4]diazepin-2-one derivatives with particular emphasis on derivatizations compatible with brain penetration and in vivo activity. Especially the compounds bearing a para-pyridine consistently showed in vivo activity in rat behavioral models after oral administration, for example, blockade of the mGluR2/3 agonist LY354740-induced hypoactivity and improvement of a working memory deficit induced either by LY354740 or scopolamine in the delayed match to position task (DMTP). Moreover, combination studies with a cholinesterase inhibitor show apparent synergistic effects on working memory impairment induced by scopolamine.


Subject(s)
Azepines/chemical synthesis , Azepines/pharmacology , Benzodiazepinones/chemical synthesis , Benzodiazepinones/pharmacology , Excitatory Amino Acid Antagonists/chemical synthesis , Memory Disorders/drug therapy , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Administration, Oral , Animals , Azepines/chemistry , Behavior, Animal , Benzodiazepinones/chemistry , Brain/metabolism , Brain/physiopathology , Bridged Bicyclo Compounds/pharmacology , Cholinesterase Inhibitors/pharmacology , Drug Synergism , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Memory Disorders/chemically induced , Rats , Scopolamine/pharmacology
13.
Bioorg Med Chem Lett ; 20(22): 6735-8, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20850972

ABSTRACT

The rational design of a novel series of pyrrolidine derivatives as neurokinin-3 receptor antagonists is reported starting from a selective neurokinin-1 receptor antagonist. Typical representatives in this series showed in vivo efficacy after oral administration in a NK3 mediated functional assay. This series of NK3 antagonists shows promise to deliver a novel antipsychotic.


Subject(s)
Pyrrolidines/pharmacology , Receptors, Neurokinin-3/antagonists & inhibitors , Administration, Oral , Drug Design , Models, Molecular , Pyrrolidines/administration & dosage , Pyrrolidines/chemistry
14.
Bioorg Med Chem Lett ; 20(11): 3405-8, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20430616

ABSTRACT

During a program directed at selective NK(1) receptor antagonists, we serendipitously discovered an NK(1) receptor ligand with additional affinity for the NK(3) receptor. Recognising an opportunity for a drug discovery program aiming for dual NK(1)/NK(3) receptor antagonists, we prepared a series of analogues from a novel, versatile building block. From this series emerged compounds with high and balanced affinities for the NK(1) and the NK(3) receptors. Typical representatives of this series were active in the gerbil foot tapping assay after oral administration.


Subject(s)
Neurokinin-1 Receptor Antagonists , Receptors, Neurokinin-3/antagonists & inhibitors , Administration, Oral , Animals , Drug Discovery , Ligands , Models, Molecular , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-3/metabolism
15.
Neuropharmacology ; 58(1): 259-67, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19540857

ABSTRACT

The tachykinin NK(3) receptor shows promise as a novel target for antipsychotics, but knowledge of downstream activity following tachykinin NK(3) receptor activation is lacking. To determine the practical utility of senktide-induced tail whips in mice as a tool for determining and characterizing downstream activity following tachykinin NK(3) receptor activation, mice were injected with 0.05 nmol of senktide i.c.v. and the number of tail whip bouts was counted for 20 min. Strain differences were observed, with NMRI mice showing a stronger tail whip response than C57Bl/6J mice. Tachykinin NK(3) receptor specificity was confirmed by the absence of the senktide-induced tail whip response in tachykinin NK(3) receptor knockout mice. Effects of tachykinin receptor pharmacological agents were tested by pretreatment with tachykinin NK(3) receptor antagonists (SB222200, talnetant and osanetant), which attenuated senktide-induced tail whips, and the tachykinin NK(1) receptor antagonist MK869, which had no effect on senktide-induced tail whips. Pharmacological interactions with other neurotransmitter systems were determined by pretreatment with dopamine D(1), D(2), and D(3) receptor antagonists, atypical antipsychotics, serotonin 5HT(1a) receptor antagonists, serotonin 5HT(2a/c) receptor antagonists, benzodiazepine and putative anxiolytics, antidepressants, and an anticholinergic. Senktide-induced tail whips were attenuated by dopamine D(2) receptor antagonists, atypical antipsychotics, serotonin 5HT(2a/c) antagonists, and benzodiazepine anxiolytics, but unaffected by drugs from other classes. Thus, the senktide-induced tail whip response is easily quantifiable, specific to the tachykinin NK(3) receptor, and provides valuable information on the downstream pharmacology of tachykinin NK(3) receptor activation.


Subject(s)
Behavioral Symptoms/chemically induced , Movement/drug effects , Neurotransmitter Agents/pharmacology , Peptide Fragments/pharmacology , Receptors, Neurokinin-3/agonists , Substance P/analogs & derivatives , Tail/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Behavior, Animal/physiology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Movement/physiology , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/deficiency , Serotonin Antagonists/pharmacology , Species Specificity , Statistics, Nonparametric , Substance P/pharmacology , Tail/physiology
16.
Bioorg Med Chem Lett ; 19(20): 5958-61, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19740657

ABSTRACT

In a search for GABAA alpha5 ligands that combine high subtype binding selectivity with a marked inverse agonism imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepines were identified as a promising class. A short tandem reaction allowed rapid access to this chemical series, thereby facilitating rapid SAR generation which guided the optimization process. Two compounds (10e and 11f) were found to be active in an in vivo paradigm for cognitive improvement.


Subject(s)
Anticonvulsants/chemistry , Benzodiazepines/chemistry , Cognition Disorders/drug therapy , Receptors, GABA-A/metabolism , Triazoles/chemistry , Adjuvants, Anesthesia/pharmacology , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/pharmacokinetics , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacokinetics , Benzodiazepines/pharmacology , Cell Line , Drug Inverse Agonism , GABA-A Receptor Agonists , Humans , Memory, Short-Term/drug effects , Microsomes, Liver/metabolism , Rats , Scopolamine/pharmacology , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/pharmacology
17.
Bioorg Med Chem Lett ; 19(19): 5746-52, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19726184

ABSTRACT

Through iterative design cycles we have discovered a number of novel new classes where the imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepine was deemed the most promising GABA(A) alpha5 inverse agonist class with potential for cognitive enhancement. This class combines a modest subtype binding selectivity with inverse agonism and has the most favourable molecular properties for further lead optimisation towards a central nervous system (CNS) acting medicine.


Subject(s)
Benzodiazepines/chemistry , Nootropic Agents/chemistry , Receptors, GABA-A/metabolism , Triazoles/chemistry , Animals , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacology , Drug Discovery , Drug Inverse Agonism , GABA-A Receptor Agonists , Humans , Nootropic Agents/chemical synthesis , Nootropic Agents/pharmacology , Oocytes/drug effects , Triazoles/chemical synthesis , Triazoles/pharmacology , Xenopus laevis
18.
Psychopharmacology (Berl) ; 202(1-3): 207-23, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18936916

ABSTRACT

RATIONALE: GABAA alpha5 subunit-containing receptors are primarily expressed in the hippocampus and their role in learning and memory has been demonstrated recently by both genetic and pharmacological approaches. OBJECTIVES: The objective of the study is to evaluate the cognitive effects of a novel GABAA alpha5 receptor inverse agonist, RO4938581 in rats and monkeys. MATERIALS AND METHODS: The in vitro profile was determined using radioligand binding and electrophysiological assays for the GABAA alpha1, alpha2, alpha3, and alpha5 receptors. Long-term potentiation (LTP) was performed in mouse hippocampal slices. Cognitive effects were assessed in rats in the delayed match to position (DMTP) task and the Morris water maze. In monkeys, the object retrieval task was used. Pro-convulsant and anxiogenic potentials were evaluated in mice and rats. In vivo receptor occupancy was determined using [3H]-RO0154513. RESULTS: RO4938581 is a potent inverse agonist at the GABAA alpha5 receptor, with both binding and functional selectivity, enhancing hippocampal LTP. RO4938581 reversed scopolamine-induced working memory impairment in the DMTP task (0.3-1 mg/kg p.o.) and diazepam-induced spatial learning impairment (1-10 mg/kg p.o.). RO4938581 improved executive function in monkeys (3-10 mg/kg p.o.). Importantly, RO4938581 showed no anxiogenic and pro-convulsive potential. RO4938581 dose-dependently bound to GABAA alpha5 receptors and approximately 30% receptor occupancy was sufficient to produce enhanced cognition in the rat. CONCLUSIONS: The data further support the potential of GABAA alpha5 receptors as a target for cognition-enhancing drugs. The dual binding and functional selectivity offers an ideal profile for cognition-enhancing effects without the unwanted side effects associated with activity at other GABAA receptor subtypes.


Subject(s)
Benzodiazepines/pharmacology , Cognition/drug effects , Imidazoles/pharmacology , Nootropic Agents/pharmacology , Receptors, GABA-A/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Female , Haplorhini , Hippocampus/drug effects , In Vitro Techniques , Learning/drug effects , Long-Term Potentiation/drug effects , Male , Membranes/drug effects , Memory/drug effects , Mice , Mice, Inbred DBA , Motor Activity/drug effects , Patch-Clamp Techniques , Plasmids , Rats , Rats, Wistar , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Recombinant Proteins , Seizures/chemically induced
19.
Bioorg Med Chem Lett ; 18(8): 2725-9, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18374569

ABSTRACT

A series of 1,3-dihydro-benzo[b][1,4]diazepin-2-one derivatives was evaluated as non-competitive mGluR2/3 antagonists. Replacement of the (2-aryl)-ethynyl-moiety in 8-position with smaller less lipophilic substituents produced compounds inhibiting the binding of [3H]-LY354740 to rat mGluR2 with low nanomolar affinity and consistent functional effect at both mGluR2 and mGluR3. These compounds were able to reverse LY354740-mediated inhibition of field excitatory postsynaptic potentials in the rat dentate gyrus and in vivo activity could be demonstrated by reversal of the LY354740-induced hypoactivity in mice after oral administration.


Subject(s)
Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Animals , Benzodiazepines/chemistry , CHO Cells , Cricetinae , Cricetulus , Mice , Mice, Inbred C57BL , Molecular Structure , Rats , Receptors, Metabotropic Glutamate/genetics , Structure-Activity Relationship
20.
Pharmacol Biochem Behav ; 85(1): 44-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16920181

ABSTRACT

In the current study we examined the effects of serotonin reuptake inhibitors on the locomotor activity of gerbils, and undertook experiments to understand the mechanisms involved in their effects. The selective serotonin reuptake inhibitors (SSRIs) fluoxetine (1-30 mg/kg, i.p.) and escitalopram (0.03-10 mg/kg, i.p.) dose-dependently increased locomotor activity, whereas the serotonin and noradrenaline reuptake inhibitor duloxetine (0.3-30 mg/kg, i.p.) did not. The noradrenaline reuptake inhibitor (NRI) reboxetine, which alone did not significantly affect locomotion (1-30 mg/kg, i.p.), markedly reduced the effects of escitalopram. The locomotor effects of fluoxetine and escitalopram were dependent on novelty since both compounds showed rapid habituation in novel cages and were inactive when tested in home cages. Both diazepam (0.3-10 mg/kg, i.p.) and d-amphetamine (0.3-10 mg/kg, s.c.) increased locomotor activity but only the effects of diazepam were novelty-dependent. The finding that SSRIs increased locomotion, with a negative modulatory role for NRI, in a novelty-dependent manner, similar to diazepam, suggests that anxiety plays an important role in the present paradigm. The increase in locomotion as observed in our test conditions can be readily used as a selective and sensitive in vivo assay for serotonin transport inhibition in gerbils.


Subject(s)
Locomotion/drug effects , Motor Activity/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Gerbillinae , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...