Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Neuropharmacology ; 197: 108754, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34389398

ABSTRACT

Alzheimer's disease (AD) is a profoundly debilitating neurodegenerative disorder characterized most notably by progressive cognitive decline, but also agitation and behavioral disturbances that are extremely disruptive to patient and caregiver. Current pharmacological treatments for these symptoms have limited efficacy and significant side effects. We have recently reported the discovery of Compound 24, an M4 positive allosteric modulator (PAM) that is potent, highly selective, and devoid of cholinergic-like side effects in rats. In order to further evaluate the translatability of the effects of compound 24 in primates, here we describe the effect of Compound 24 on three behavioral and cognition assays in rhesus monkeys, the stimulant induced motor activity (SIMA) assay, the object retrieval detour task (ORD), and the visuo-spatial paired-associates learning (vsPAL) task. As far as we know, this is the first such characterization of an M4 PAM in non-human primate. Compound 24 and the clinical standard olanzapine attenuated amphetamine induced hyperactivity to a similar degree. In addition, Compound 24 demonstrated procognitive effects in scopolamine-impaired ORD and vsPAL, and these effects were of similar magnitude to donepezil. These findings suggest that M4 PAMs may be beneficial to diseases such as Alzheimer's disease and schizophrenia, which are marked by behavioral disturbances as well as deficits in cognitive function.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Behavior, Animal/drug effects , Cholinergic Agents/pharmacology , Cognition Disorders/drug therapy , Receptor, Muscarinic M4/drug effects , Schizophrenia/drug therapy , Schizophrenic Psychology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Association Learning/drug effects , Central Nervous System Stimulants , Cholinergic Agents/pharmacokinetics , Cognition Disorders/psychology , Hyperkinesis/chemically induced , Hyperkinesis/prevention & control , Macaca mulatta , Male , Motor Activity/drug effects , Olanzapine/pharmacology , Orientation/drug effects
2.
J Pharmacol Exp Ther ; 355(3): 442-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26446308

ABSTRACT

Improved treatment of Alzheimer disease (AD) is a significant unmet medical need that is becoming even more critical given the rise in the number of patients and the substantial economic burden. The current standards of care, acetylcholinesterase inhibitors (AChEIs), are hindered by gastrointestinal side effects owing to their nonselective activation of muscarinic and nicotinic receptors. Recently, the highly selective M1 positive allosteric modulator PQCA (1-((4-cyano-4-(pyridine-2-yl)piperidin-1-yl)methyl-4-oxo-4 H-quinolizine-3-carboxylic acid) has been demonstrated to improve cognition in a variety of rodent and nonhuman primate cognition models without producing significant gastrointestinal side effects. Here we describe the effect of PQCA and the AChEI donepezil on two clinically relevant and highly translatable touchscreen cognition tasks in nonhuman primates: paired-associates learning (PAL) and the continuous-performance task (CPT). Blockade of muscarinic signaling by scopolamine produced significant impairments in both PAL and CPT. PQCA and donepezil attenuated the scopolamine deficits in both tasks, and the action of these two compounds was similar in magnitude. In addition, the combination of subeffective doses of PQCA and donepezil enhanced PAL performance. These results further suggest that M1-positive allosteric modulators, either as monotherapy or as an add-on to current standards of care, have potential to reduce the cognitive deficits associated with AD.


Subject(s)
Attention/drug effects , Memory/drug effects , Muscarinic Agonists/pharmacology , Piperidines/pharmacology , Psychomotor Performance/drug effects , Quinolizines/pharmacology , Receptor, Muscarinic M1/drug effects , Animals , Association Learning/drug effects , Cholinesterase Inhibitors/pharmacology , Cognition/drug effects , Discrimination, Psychological/drug effects , Donepezil , Dose-Response Relationship, Drug , Indans/pharmacology , Macaca mulatta , Male , Muscarinic Antagonists/pharmacology , Scopolamine/pharmacology
3.
Psychopharmacology (Berl) ; 232(11): 1859-66, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25491927

ABSTRACT

RATIONALE: The standards of care for Alzheimer's disease, acetylcholinesterase inhibitors such as donepezil (Aricept®), are dose-limited due to adverse side-effects. These adverse events lead to significant patient non-compliance, constraining the dose and magnitude of efficacy that can be achieved. Non-selective muscarinic receptor orthosteric agonists such as Xanomeline have been shown to be effective in treating symptoms as well, but were also poorly tolerated. Therefore, there is an unmet medical need for a symptomatic treatment that improves symptoms and is better tolerated. METHODS: We compared donepezil, xanomeline, and the novel selective muscarinic 1 receptor positive allosteric modulator PQCA in combination with donepezil in the object retrieval detour (ORD) cognition test in rhesus macaque. Gastrointestinal (GI) side effects (salivation and feces output) were then assessed with all compounds to determine therapeutic window. RESULTS: All three compounds significantly reduced a scopolamine-induced deficit in ORD. Consistent with what is observed clinically in patients, both donepezil and xanomeline produced significant GI effects in rhesus at doses equal to or less than a fivefold margin from the minimum effective dose that improves cognition. In stark contrast, PQCA produced no GI side effects when tested at the same dose range. CONCLUSIONS: These data suggest M1 positive allosteric modulators have the potential to improve cognition in Alzheimer's disease with a greater therapeutic margin than the current standard of care, addressing an important unmet medical need.


Subject(s)
Cognition/drug effects , Indans/pharmacology , Muscarinic Agonists/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Quinolizines/pharmacology , Receptor, Muscarinic M1/drug effects , Thiadiazoles/pharmacology , Aged , Alzheimer Disease/drug therapy , Animals , Appetitive Behavior/drug effects , Attention/drug effects , Defecation/drug effects , Donepezil , Female , Humans , Indans/toxicity , Macaca mulatta , Male , Neuropsychological Tests , Orientation/drug effects , Piperidines/toxicity , Problem Solving/drug effects , Pyridines/toxicity , Quinolizines/toxicity , Salivation/drug effects , Thiadiazoles/toxicity
4.
Psychopharmacology (Berl) ; 231(3): 511-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24051602

ABSTRACT

Reduced NMDA receptor functioning is hypothesized to underlie the cognitive and negative symptoms associated with schizophrenia. However, because direct activation of the NMDA receptor is accompanied by neurotoxicity, mechanisms that activate the glycine co-agonist site on the NMDA receptor could carry greater therapeutic potential. In the current study, the effects of two glycine transporter 1 (GlyT1) inhibitors, RG1678 and ORG25935, were characterized in the object-retrieval detour (ORD) task in scopolamine-impaired rhesus monkeys and, using positron emission tomography (PET), the GlyT1 occupancy to efficacy relationship of each compound was established. Scopolamine exerted a significant decrease in accuracy in the ORD task. Lower doses of RG1678 (0.3 and 1.0 mg/kg, p.o.) significantly attenuated the impact of scopolamine, whereas the highest dose tested (1.8 mg/kg) did not. The predicted GlyT1 occupancies of RG1678 at the effective doses were ~10 and 30 %. ORG25935 (0.1, 0.3, and 1 mg/kg, p.o.) also significantly attenuated the impact of scopolamine on the ORD task, whereas 3 mg/kg did not. The predicted GlyT1 occupancies of ORG25935 at the effective doses ranged from 16 to 80 %. These data suggest that GlyT1 inhibitors have the potential to improve performance on prefrontal cortex-dependent tests such as the ORD task, but that efficacy is lost when higher occupancies are achieved. Importantly, recent Ph2B data published by Roche suggests that low but not high doses of RG1678 improved negative symptoms in patients with schizophrenia, highlighting the potential translational nature of the current preclinical findings.


Subject(s)
Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Glycine Plasma Membrane Transport Proteins/metabolism , Muscarinic Antagonists/adverse effects , Piperazines/pharmacology , Scopolamine/adverse effects , Sulfones/pharmacology , Tetrahydronaphthalenes/pharmacology , Animals , Central Nervous System Agents/pharmacokinetics , Central Nervous System Agents/pharmacology , Dose-Response Relationship, Drug , Macaca mulatta , Male , Motor Activity/drug effects , Piperazines/pharmacokinetics , Positron-Emission Tomography , Sulfones/pharmacokinetics , Task Performance and Analysis , Tetrahydronaphthalenes/pharmacokinetics
5.
Sci Transl Med ; 5(179): 179ra44, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23552372

ABSTRACT

Current treatments for insomnia, such as zolpidem (Ambien) and eszopiclone (Lunesta), are γ-aminobutyric acid type A (GABAA)-positive allosteric modulators that carry a number of side effects including the potential to disrupt cognition. In an effort to develop better tolerated medicines, we have identified dual orexin 1 and 2 receptor antagonists (DORAs), which promote sleep in preclinical animal models and humans. We compare the effects of orally administered eszopiclone, zolpidem, and diazepam to the dual orexin receptor antagonist DORA-22 on sleep and the novel object recognition test in rat, and on sleep and two cognition tests (delayed match to sample and serial choice reaction time) in the rhesus monkey. Each compound's minimal dose that promoted sleep versus the minimal dose that exerted deficits in these cognitive tests was determined, and a therapeutic margin was established. We found that DORA-22 has a wider therapeutic margin for sleep versus cognitive impairment in rat and rhesus monkey compared to the other compounds tested. These data were further supported with the demonstration of a wider therapeutic margin for DORA-22 compared to the other compounds on sleep versus the expression of hippocampal activity-regulated cytoskeletal-associated protein (Arc), an immediate-early gene product involved in synaptic plasticity. These findings suggest that DORAs might provide an effective treatment for insomnia with a greater therapeutic margin for sleep versus cognitive disturbances compared to the GABAA-positive allosteric modulators currently in use.


Subject(s)
Cognition/drug effects , Hypnotics and Sedatives/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , Sleep/drug effects , Administration, Oral , Animals , Attention/drug effects , Azabicyclo Compounds/administration & dosage , Azabicyclo Compounds/pharmacology , Choice Behavior/drug effects , Cytoskeletal Proteins/metabolism , Diazepam/administration & dosage , Diazepam/pharmacology , Eszopiclone , Hippocampus/drug effects , Hippocampus/metabolism , Macaca mulatta , Male , Memory, Short-Term/drug effects , Nerve Tissue Proteins/metabolism , Orexin Receptors , Piperazines/administration & dosage , Piperazines/pharmacology , Piperidines/administration & dosage , Piperidines/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Recognition, Psychology , Task Performance and Analysis , Time Factors , Triazoles/administration & dosage , Triazoles/pharmacology , Zolpidem , gamma-Aminobutyric Acid/metabolism
6.
Neuropharmacology ; 64: 191-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22659472

ABSTRACT

The cognitive deficits associated with schizophrenia are recognized as a core component of the disorder, yet there remain no available therapeutics to treat these symptoms of the disease. As a result, there is a need for establishing predictive preclinical models to identify the therapeutic potential of novel compounds. In the present study, rhesus monkeys were trained in the object retrieval-detour task, which is dependent on the prefrontal cortex, a brain region implicated in the cognitive deficits associated with schizophrenia. The NMDA receptor antagonist ketamine significantly impaired performance without affecting measures of motor or visuospatial abilities. Pre-treatment with the nicotinic α7 agonist GTS-21 (0.03 mg/kg) significantly attenuated the ketamine-induced impairment, consistent with reports from clinical trials suggesting that nicotinic α7 receptor agonism has pro-cognitive potential in clinical populations. In contrast, pretreatment with the acetylcholinesterase inhibitor donepezil failed to reverse the ketamine-induced impairment, consistent with studies showing a lack of pro-cognitive effects in patients with schizophrenia. These data suggest that the ketamine-impaired object retrieval-detour task could provide a model with improved predictive validity for drug development, and confirm the need for additional efforts in back-translation. This article is part of a Special Issue entitled 'Cognitive Enhancers'.


Subject(s)
Benzylidene Compounds/therapeutic use , Cognition Disorders/prevention & control , Disease Models, Animal , Nicotinic Agonists/therapeutic use , Nootropic Agents/therapeutic use , Pyridines/therapeutic use , Receptors, Nicotinic/metabolism , Schizophrenia/drug therapy , Animals , Behavior, Animal/drug effects , Cholinesterase Inhibitors/adverse effects , Cholinesterase Inhibitors/therapeutic use , Cognition/drug effects , Cognition Disorders/etiology , Donepezil , Drug Evaluation, Preclinical/methods , Excitatory Amino Acid Antagonists , Indans/adverse effects , Indans/therapeutic use , Ketamine , Macaca mulatta , Male , Molecular Targeted Therapy , Nicotinic Agonists/adverse effects , Nootropic Agents/adverse effects , Piperidines/adverse effects , Piperidines/therapeutic use , Psychomotor Performance/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Nicotinic/chemistry , Schizophrenia/physiopathology , alpha7 Nicotinic Acetylcholine Receptor
7.
Psychopharmacology (Berl) ; 225(1): 21-30, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22825578

ABSTRACT

RATIONALE: The current standards of care for Alzheimer's disease, acetylcholinesterase inhibitors, have limited efficacy due to a host of mechanism-related side effects arising from indiscriminate activation of muscarinic and nicotinic receptors. The M1 muscarinic receptor is predominantly expressed in the brain in regions involved in cognition, and therefore selective activation of the M1 receptor would be expected to boost cognitive performance with reduced risk of peripheral side effects. OBJECTIVES: Here we investigated whether the selective M1 muscarinic receptor positive allosteric modulator, PQCA, improves cognitive performance and cerebral blood flow. RESULTS: PQCA attenuated a scopolamine-induced deficit in novel object recognition in rat, self-ordered spatial search in cynomolgus macaque, and the object retrieval detour task in rhesus macaque. Beneficial effects in each of these assays and species were observed at similar plasma drug concentrations. Furthermore, at similar drug concentrations that were effective in the behavioral studies, PQCA increased blood flow in the frontal cortex of mice, providing a translational biomarker that could be used to guide dose selection for clinical studies. CONCLUSIONS: These findings provide a framework for appropriately testing an M1 selective compound in patients with Alzheimer's disease.


Subject(s)
Brain/drug effects , Cognition/drug effects , Piperidines/pharmacology , Quinolizines/pharmacology , Receptor, Muscarinic M1/drug effects , Allosteric Regulation , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Brain/blood supply , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Macaca fascicularis , Macaca mulatta , Male , Mice , Piperidines/administration & dosage , Quinolizines/administration & dosage , Rats , Rats, Wistar , Receptor, Muscarinic M1/metabolism , Regional Blood Flow/drug effects , Scopolamine/toxicity , Species Specificity
8.
Neuropharmacology ; 64: 215-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22750078

ABSTRACT

Phosphodiesterase 10A (PDE10A) is a novel target for the treatment of schizophrenia that may address multiple symptomatic domains associated with this disorder. PDE10A is highly expressed in the brain and functions to metabolically inactivate the important second messengers cAMP and cGMP. Here we describe effects of a potent and orally bioavailable PDE10A inhibitor [2-(6-chloropyridin-3-yl)-4-(2-methoxyethoxy)-7,8-dihydropyrido[4,3-d]pyrimidin-6(5H)-yl](imidazo[1,5-a]pyridin-1-yl)methanone] (THPP-1) on striatal signaling pathways, in behavioral tests that predict antipsychotic potential, and assays that measure episodic-like memory in rat and executive function in rhesus monkey. THPP-1 exhibits nanomolar potency on the PDE10A enzyme, demonstrates excellent pharmacokinetic properties in multiple preclinical animal species, and is selective for PDE10A over other PDE families of enzymes. THPP-1 significantly increased phosphorylation of proteins in the striatum involved in synaptic plasticity, including the a-amino-3-hydroxy-5-methylisoxazole-4-proprionic acid receptor (AMPA) GluR1 subunit, extracellular receptor kinase (ERK), and cAMP-response element binding protein (CREB). THPP-1 produced dose-dependent effects in preclinical assays predictive of antipsychotic activity including attenuation of MK-801-induced psychomotor activation and condition avoidance responding in rats. At similar plasma exposures, THPP-1 significantly increased object recognition memory in rat and attenuated a ketamine-induced deficit in the object retrieval detour task in rhesus monkey. These findings suggest that PDE10A inhibitors have the potential to impact multiple symptomatic domains of schizophrenia including positive symptoms and cognitive impairment. This article is part of a Special Issue entitled 'Cognitive Enhancers'.


Subject(s)
Antipsychotic Agents/therapeutic use , Cognition Disorders/prevention & control , Molecular Targeted Therapy , Nootropic Agents/therapeutic use , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/metabolism , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/blood , Antipsychotic Agents/pharmacokinetics , Behavior, Animal/drug effects , Cognition Disorders/etiology , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Executive Function/drug effects , Macaca mulatta , Male , Memory, Episodic , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Nootropic Agents/administration & dosage , Nootropic Agents/blood , Nootropic Agents/pharmacokinetics , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/blood , Phosphodiesterase Inhibitors/pharmacokinetics , Phosphoric Diester Hydrolases/chemistry , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Pyridines/administration & dosage , Pyridines/blood , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Random Allocation , Rats , Rats, Wistar , Schizophrenia/blood , Schizophrenia/metabolism , Schizophrenia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...