Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cancer Res ; 82(14): 2552-2564, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35584009

ABSTRACT

The therapeutic benefit of approved BRAF and MEK inhibitors (BRAFi/MEKi) in patients with brain metastatic BRAF V600E/K-mutated melanoma is limited and transient. Resistance largely occurs through the restoration of MAPK signaling via paradoxical BRAF activation, highlighting the need for more effective therapeutic options. Aiming to address this clinical challenge, we characterized the activity of a potent, brain-penetrant paradox breaker BRAFi (compound 1a, C1a) as first-line therapy and following progression upon treatment with approved BRAFi and BRAFi/MEKi therapies. C1a activity was evaluated in vitro and in vivo in melanoma cell lines and patient-derived models of BRAF V600E-mutant melanoma brain metastases following relapse after treatment with BRAFi/MEKi. C1a showed superior efficacy compared with approved BRAFi in both subcutaneous and brain metastatic models. Importantly, C1a manifested potent and prolonged antitumor activity even in models that progressed on BRAFi/MEKi treatment. Analysis of mechanisms of resistance to C1a revealed MAPK reactivation under drug treatment as the predominant resistance-driving event in both subcutaneous and intracranial tumors. Specifically, BRAF kinase domain duplication was identified as a frequently occurring driver of resistance to C1a. Combination therapies of C1a and anti-PD-1 antibody proved to significantly reduce disease recurrence. Collectively, these preclinical studies validate the outstanding antitumor activity of C1a in brain metastasis, support clinical investigation of this agent in patients pretreated with BRAFi/MEKi, unveil genetic drivers of tumor escape from C1a, and identify a combinatorial treatment that achieves long-lasting responses. SIGNIFICANCE: A brain-penetrant BRAF inhibitor demonstrates potent activity in brain metastatic melanoma, even upon relapse following standard BRAF inhibitor therapy, supporting further investigation into its clinical utility.


Subject(s)
Brain Neoplasms , Melanoma , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases , Mutation , Neoplasm Recurrence, Local/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf
2.
Clin Cancer Res ; 28(4): 770-780, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34782366

ABSTRACT

PURPOSE: Disease progression in BRAF V600E/K positive melanomas to approved BRAF/MEK inhibitor therapies is associated with the development of resistance mediated by RAF dimer inducing mechanisms. Moreover, progressing disease after BRAFi/MEKi frequently involves brain metastasis. Here we present the development of a novel BRAF inhibitor (Compound Ia) designed to address the limitations of available BRAFi/MEKi. EXPERIMENTAL DESIGN: The novel, brain penetrant, paradox breaker BRAFi is comprehensively characterized in vitro, ex vivo, and in several preclinical in vivo models of melanoma mimicking peripheral disease, brain metastatic disease, and acquired resistance to first-generation BRAFi. RESULTS: Compound Ia manifested elevated potency and selectivity, which triggered cytotoxic activity restricted to BRAF-mutated models and did not induce RAF paradoxical activation. In comparison to approved BRAFi at clinical relevant doses, this novel agent showed a substantially improved activity in a number of diverse BRAF V600E models. In addition, as a single agent, it outperformed a currently approved BRAFi/MEKi combination in a model of acquired resistance to clinically available BRAFi. Compound Ia presents high central nervous system (CNS) penetration and triggered evident superiority over approved BRAFi in a macro-metastatic and in a disseminated micro-metastatic brain model. Potent inhibition of MAPK by Compound Ia was also demonstrated in patient-derived tumor samples. CONCLUSIONS: The novel BRAFi demonstrates preclinically the potential to outperform available targeted therapies for the treatment of BRAF-mutant tumors, thus supporting its clinical investigation.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Brain/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
3.
Mol Ther Oncolytics ; 20: 166-174, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33575479

ABSTRACT

Glioblastoma is an invariably deadly disease. A subpopulation of glioma stem-like cells (GSCs) drives tumor progression and treatment resistance. Two recent studies demonstrated that neurons form oncogenic glutamatergic electrochemical synapses with post-synaptic GSCs. This led us to explore whether glutamate signaling through G protein-coupled metabotropic receptors would also contribute to the malignancy of glioblastoma. We found that glutamate metabotropic receptor (Grm)3 is the predominantly expressed Grm in glioblastoma. Associations of GRM3 gene expression levels with survival are confined to the proneural gene expression subtype, which is associated with enrichment of GSCs. Using multiplexed single-cell qRT-PCR, GSC marker-based cell sorting, database interrogations, and functional assays in GSCs derived from patients' tumors, we establish Grm3 as a novel marker and potential therapeutic target in GSCs. We confirm that Grm3 inhibits adenylyl cyclase and regulates extracellular signal-regulated kinase. Targeting Grm3 disrupts self-renewal and promotes differentiation of GSCs. Thus, we hypothesize that Grm3 signaling may complement oncogenic functions of glutamatergic ionotropic receptor activity in neuroglial synapses, supporting a link between neuronal activity and the GSC phenotype. The novel class of highly specific Grm3 inhibitors that we characterize herein have been clinically tested as cognitive enhancers in humans with a favorable safety profile.

4.
J Pharmacol Exp Ther ; 357(1): 17-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26801398

ABSTRACT

Nociceptin/orphanin FQ receptor (NOP) agonists have been reported to produce antinociceptive effects in rhesus monkeys with comparable efficacy to µ-opioid receptor (MOP) agonists, but without their limiting side effects. There are also known to be species differences between rodents and nonhuman primates (NHPs) in the behavioral effects of NOP agonists. The aims of this study were the following: 1) to determine if the NOP agonist Ro 64-6198 could be trained as a discriminative stimulus; 2) to evaluate its pharmacological selectivity as a discriminative stimulus; and 3) to establish the order of potency with which Ro 64-6198 produces discriminative stimulus effects compared with analgesic effects in NHPs. Two groups of rhesus monkeys were trained to discriminate either fentanyl or Ro 64-6198 from vehicle. Four monkeys were trained in the warm-water tail-withdrawal procedure to measure antinociception. Ro 64-6198 produced discriminative stimulus effects that were blocked by the NOP antagonist J-113397 and not by naltrexone. The discriminative stimulus effects of Ro 64-6198 partially generalized to diazepam, but not to fentanyl, SNC 80, ketocyclazocine, buprenorphine, phencyclidine, or chlorpromazine. Fentanyl produced stimulus effects that were blocked by naltrexone and not by J-113397, and Ro 64-6198 did not produce fentanyl-appropriate responding in fentanyl-trained animals. In measures of antinociception, fentanyl, but not Ro 64-6198, produced dose-dependent increases in tail-withdrawal latency. Together, these results demonstrate that Ro 64-6198 produced stimulus effects in monkeys that are distinct from other opioid receptor agonists, but may be somewhat similar to diazepam. In contrast to previous findings, Ro 64-6198 did not produce antinociception in the majority of animals tested even at doses considerably greater than those that produced discriminative stimulus effects.


Subject(s)
Discrimination, Psychological/drug effects , Imidazoles/pharmacology , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Analgesics, Opioid/pharmacology , Animals , Benzimidazoles/pharmacology , Conditioning, Operant/drug effects , Diazepam/pharmacology , Dose-Response Relationship, Drug , Female , Fentanyl/antagonists & inhibitors , Fentanyl/pharmacology , GABA Modulators/pharmacology , Macaca mulatta , Male , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Pain Measurement/drug effects , Piperidines/pharmacology , Nociceptin Receptor
5.
J Med Chem ; 58(3): 1358-71, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25565255

ABSTRACT

Negative allosteric modulators (NAMs) of metabotropic glutamate receptor 5 (mGlu5) have potential for the treatment of psychiatric diseases including depression, fragile X syndrome (FXS), anxiety, obsessive-compulsive disorders, and levodopa induced dyskinesia in Parkinson's disease. Herein we report the optimization of a weakly active screening hit 1 to the potent and selective compounds chloro-4-[1-(4-fluorophenyl)-2,5-dimethyl-1H-imidazol-4-ylethynyl]pyridine (basimglurant, 2) and 2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1H-imidazol-4-yl)ethynyl)pyridine (CTEP, 3). Compound 2 is active in a broad range of anxiety tests reaching the same efficacy but at a 10- to 100-fold lower dose compared to diazepam and is characterized by favorable DMPK properties in rat and monkey as well as an excellent preclinical safety profile and is currently in phase II clinical studies for the treatment of depression and fragile X syndrome. Analogue 3 is the first reported mGlu5 NAM with a long half-life in rodents and is therefore an ideal tool compound for chronic studies in mice and rats.


Subject(s)
Depression/drug therapy , Drug Discovery , Fragile X Syndrome/drug therapy , Imidazoles/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Dose-Response Relationship, Drug , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Macaca mulatta , Male , Mice , Mice, Inbred Strains , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship
6.
Neurobiol Learn Mem ; 98(3): 254-60, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22982481

ABSTRACT

We previously reported that the selective nociceptin orphanin peptide (NOP) receptor agonist, Ro64-6198, impairs mnemonic function through glutamatergic-dependent mechanisms. The aim of the current study was to determine whether the amnesic effects of Ro64-6198 involve a cholinergic component. The effects of systemic administration of Ro64-6198 (0.3 and 1 mg/kg, i.p.), the cholinergic nicotinic receptor antagonist, mecamylamine (0.1 and 1 mg/kg, s.c.), the cholinergic muscarinic receptor antagonist, scopolamine (0.1 and 0.3 mg/kg, s.c.), and the glutamatergic NMDA receptor antagonist, MK-801 (0.03 and 0.1 mg/kg, s.c.), were studied in the mouse object recognition task. All compounds tested were effective in disrupting formation of long-term (24-h delay) recognition memory. Drug interaction studies were then conducted to reveal the existence of functional interactions between NOP receptors and cholinergic and/or NMDA receptors. Co-administration of silent doses of Ro64-6198 (0.3 mg/kg) and MK-801 (0.01 mg/kg) produced clear-cut memory impairment. Similar synergistic effects were observed with the combination of mecamylamine (0.03 mg/kg) and scopolamine (0.1 mg/kg). In contrast, co-administration of Ro64-6198 (0.3 mg/kg) with either mecamylamine (0.03 and 0.1 mg/kg) or scopolamine (0.1 mg/kg) was without any effect on recognition memory. These findings suggest that NOP receptor may modulate memory formation through a functional interaction with glutamatergic but not cholinergic receptors.


Subject(s)
Cholinergic Antagonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Imidazoles/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Opioid/agonists , Recognition, Psychology/drug effects , Spiro Compounds/pharmacology , Animals , Dizocilpine Maleate/pharmacology , Drug Interactions , Male , Mecamylamine/pharmacology , Mice , Scopolamine/pharmacology , Nociceptin Receptor
7.
Psychopharmacology (Berl) ; 222(2): 203-14, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22249359

ABSTRACT

RATIONALE: Ro 64-6198, the prototypical non-peptide nociceptin/orphanin FQ peptide (NOP) receptor agonist, has potent anxiolytic-like effects in several preclinical models and species. However the effects of Ro 64-6198 on distinctive anxiety-provoking conditions related to unconditioned conflict behavior as well as its role in despair-like behavior remain to be addressed. OBJECTIVE: Here we examined the effects of Ro 64-6198 on unconditioned conflict anxiety using stimuli with different salience and on regulation of autonomic reactivity and compared these to the effects of benzodiazepine receptor agonists. We also addressed the potential effects of Ro 64-6198 on despair-like behavior. MATERIALS AND METHODS: Ro 64-6198 (0.1 to 10 mg/kg i.p.) and either diazepam or chlordiazepoxide were tested in the Vogel conflict punished drinking test (VCT) in Sprague Dawley rats, in the social approach-avoidance (SAA) test in Lewis rats, in the novelty-induced hypophagia (NIH) in C57BL/6J mice, and in stress-induced hyperthermia in NMRI mice, as well as in the forced swim test (FST) in Sprague Dawley rats and the tail suspension test (TST) in C57BL/6J mice. RESULTS: Ro 64-6198 (0.3 to 3 mg/kg) dose-dependently produced anxiolytic-like effects in the VCT, SAA, NIH, and SIH, similar to benzodiazepine receptor agonists. Ro 64-6198 did not alter immobility time in the FST and TST. CONCLUSIONS: Ro 64-6198 produced marked anxiolytic-like effects in response to a variety of mild to strong anxiogenic stimuli, whereas it did not facilitate depression-related behaviors. This data extend previous literature suggesting that NOP receptors are a viable target for the treatment of anxiety disorders.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Depression/drug therapy , Imidazoles/pharmacology , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Animals , Behavior, Animal/drug effects , Chlordiazepoxide/pharmacology , Conflict, Psychological , Diazepam/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Imidazoles/administration & dosage , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Spiro Compounds/administration & dosage , Stress, Psychological/drug therapy , Nociceptin Receptor
8.
Neuropsychopharmacology ; 37(2): 378-89, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21881568

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) peptide and its receptor (NOP receptor) have been implicated in a host of brain functions and diseases, but the contribution of this neuropeptide system to behavioral processes of relevance to psychosis has not been investigated. We examined the effect of the NOP receptor antagonists, Compound 24 and J-113397, and the synthetic agonist, Ro64-6198, on time function (2-2000 ms prepulse-pulse intervals) of acoustic (80 dB/10 ms prepulse) and visual (1000 Lux/20 ms prepulse) prepulse inhibition of startle reflex (PPI), a preattentive sensory filtering mechanism that is central to perceptual and mental integration. The effects of the dopamine D1-like receptor agonist, SKF-81297, the D2-like receptor agonist, quinelorane, and the mixed D1/D2 agonist, apomorphine, were studied for comparison. When acoustic stimulus was used as prepulse, BALB/cByJ mice displayed a monotonic time function of PPI, and consistent with previous studies, apomorphine and SKF-81279 induced PPI impairment, whereas quinelorane had no effect. None of the NOP receptor ligands was effective on acoustic PPI. When flash light was used as prepulse, BALB/cByJ mice displayed a bell-shaped time function of PPI and all dopamine agonists were active. Ro64-6198 was also effective in reducing visual PPI. NOP receptor antagonists showed no activity but blocked disruptive effect of Ro64-6198. Finally, coadministration of the typical antipsychotic, haloperidol, attenuated PPI impairment induced by Ro64-6198, revealing involvement of a dopaminergic component. These findings show that pharmacological stimulation of NOP or dopamine D2-like receptors is more potent in disrupting visual than acoustic PPI in mice, whereas D1-like receptor activation disrupts both. They further suggest that dysfunction of N/OFQ transmission may be implicated in the pathogenesis of psychotic manifestations.


Subject(s)
Dopamine Agonists/pharmacology , Receptors, Dopamine/physiology , Receptors, Opioid/physiology , Sensory Gating/physiology , Acoustic Stimulation/methods , Animals , Benzimidazoles/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Haloperidol/pharmacology , Imidazoles/antagonists & inhibitors , Imidazoles/pharmacology , Lysine/analogs & derivatives , Lysine/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Narcotic Antagonists , Photic Stimulation/methods , Piperidines/pharmacology , Receptors, Opioid/agonists , Reflex, Startle/drug effects , Reflex, Startle/physiology , Sensory Gating/drug effects , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacology , Nociceptin Receptor
9.
Psychopharmacology (Berl) ; 213(1): 53-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20852848

ABSTRACT

RATIONALE: The synthetic nonpeptide NOP (nociceptin/orphanin FQ peptide) receptor agonist Ro 64-6198 produces antinociception in rhesus monkeys. In rodents, it has much more variable effects on pain responses, but has response rate-increasing effects on punished operant behavior and decreases drug reward. OBJECTIVES: The aim of this study was to compare Ro 64-6198 with the benzodiazepine diazepam in tests of analgesia, drug self-administration, and response-increasing effects in rhesus monkeys. RESULTS: Ro 64-6198 (0.001-0.01 mg/kg, i.v.) produced antinociception against an acute noxious stimulus (50°C water) in the absence of sedation, whereas diazepam (0.32-3.2 mg/kg, i.v.) did not have analgesic effects without sedation. Diazepam (1.0-5.6 mg/kg, i.v.) and the largest dose of Ro 64-6198 (0.32 mg/kg, i.v.) decreased lever pressing maintained by intravenous self-administration of the mu-opioid agonist, remifentanil, but neither effect could be distinguished from sedative effects. Although neither drug consistently increased responding during nonreinforcement, such effects were observed more frequently following diazepam administration. The effects of Ro 64-6198 on lever pressing were blocked by the NOP-receptor antagonist, J-113397, but not by the benzodiazepine antagonist, flumazenil. CONCLUSIONS: These findings suggest that the effects of Ro 64-6198 on operant lever pressing are mediated by NOP receptors and that larger doses are required to impact operant behavior when compared directly with those that produce antinociception. Therefore, the present findings support previous literature suggesting NOP receptors are a viable target for pain management.


Subject(s)
Conditioning, Operant/drug effects , Diazepam/pharmacology , Imidazoles/pharmacology , Piperidines/pharmacology , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Animals , Benzimidazoles/pharmacology , Diazepam/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions , Female , Imidazoles/administration & dosage , Macaca mulatta , Male , Narcotic Antagonists , Pain Measurement/drug effects , Piperidines/administration & dosage , Remifentanil , Self Administration/methods , Spiro Compounds/administration & dosage , Nociceptin Receptor
10.
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
11.
Brain Res ; 1363: 180-90, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-20875805

ABSTRACT

Altered glutamatergic synaptic transmission is among the key events defining the course of Alzheimer's disease (AD). mGlu2 receptors, a subtype of group II metabotropic glutamate receptors, regulate (as autoreceptors) fast synaptic transmission in the CNS via the controlled release of the excitatory amino acid glutamate. Since their pharmacological manipulation in rodents has been reported to affect cognition, they are potential drug targets for AD therapy. We examined the fate of these receptors in cases of AD as well as in aging PS2APP mice--a proposed model of the disease. In vitro binding of [(3)H]LY354740, a selective group II agonist (with selective affinity for mGlu2 receptors, under the assay conditions used) and quantitative radioautography revealed a partial, but highly significant, loss of receptors in amyloid-affected discrete brain regions of AD cases and PS2APP mice. Among the mouse brain regions affected were, above all, the subiculum but also frontolateral cortex, dentate gyrus, lacunosum moleculare and caudate putamen. In AD, significant receptor losses were registered in entorhinal cortex and lacunosum moleculare (40% and 35%, respectively). These findings have implications for the development of selective ligands for symptomatic therapy in AD and for its diagnosis.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Peptide Fragments/metabolism , Presenilin-2/metabolism , Receptors, Metabotropic Glutamate/metabolism , Aged , Aged, 80 and over , Aging/pathology , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Animals , Bridged Bicyclo Compounds/metabolism , Bridged Bicyclo Compounds/pharmacology , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/pathology , Humans , Iodine Radioisotopes , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Peptide Fragments/pharmacology , Presenilin-2/genetics , Radioligand Assay , Rats , Rats, Inbred F344 , Tritium
12.
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
13.
Neuropsychopharmacology ; 34(9): 2088-96, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19279568

ABSTRACT

Behavioral effects of a nonpeptidic NOP (nociceptin/orphanin FQ Peptide) receptor agonist, Ro 64-6198, have not been studied in primate species. The aim of the study was to verify the receptor mechanism underlying the behavioral effects of Ro 64-6198 and to systematically compare behavioral effects of Ro 64-6198 with those of a mu-opioid receptor agonist, alfentanil, in monkeys. Both Ro 64-6198 (0.001-0.06 mg/kg, s.c.) and alfentanil (0.001-0.06 mg/kg, s.c.) produced antinociception against an acute noxious stimulus (50 degrees C water) and capsaicin-induced allodynia. An NOP receptor antagonist, J-113397 (0.01-0.1 mg/kg, s.c.), dose-dependently produced rightward shifts of the dose-response curve of Ro 64-6198-induced antinociception. The apparent pA(2) value of J-113397 was 8.0. Antagonist studies using J-113397 and naltrexone revealed that Ro 64-6198 produced NOP receptor-mediated antinociception independent of mu-opioid receptors. In addition, alfentanil dose-dependently produced respiratory depression and itch/scratching responses, but antinociceptive doses of Ro 64-6198 did not produce such effects. More important, Ro 64-6198 did not produce reinforcing effects comparable with those of alfentanil, cocaine, or methohexital under self-administration procedures in monkeys. These results provide the first functional evidence that the activation of NOP receptors produces antinociception without reinforcing effects in primates. Non-peptidic NOP receptor agonists may have therapeutic value as novel analgesics without abuse liability in humans.


Subject(s)
Analgesics, Opioid/pharmacology , Behavior, Animal/drug effects , Imidazoles/pharmacology , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Alfentanil/pharmacology , Animals , Benzimidazoles/pharmacology , Capsaicin , Central Nervous System Agents/pharmacology , Dose-Response Relationship, Drug , Female , Hot Temperature , Macaca mulatta , Male , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Pain/chemically induced , Pain/drug therapy , Piperidines/pharmacology , Pruritus/drug therapy , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Reinforcement, Psychology , Respiratory Insufficiency/drug therapy , Nociceptin Receptor
14.
Bioorg Med Chem Lett ; 19(6): 1666-9, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19233648

ABSTRACT

Small molecule mGluR1 enhancers, which are 9H-xanthene-9-carboxylic acid [1,2,4]oxadiazol-3-yl- and (2H-tetrazol-5-yl)-amides, have been previously reported. Fluorinated 9H-xanthene-9-carboxylic acid oxazol-2-yl-amides with improved pharmacokinetic properties have been designed and synthesized as useful pharmacological tools for the study of the physiological roles mediated by mGlu1 receptors. The synthesis and the structure-activity relationship of this class of positive allosteric modulators of mGlu1 receptors will be discussed in detail.


Subject(s)
Amides/chemistry , Chemistry, Pharmaceutical/methods , Receptors, Metabotropic Glutamate/chemistry , Administration, Oral , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Design , Electrophysiology/methods , Humans , Models, Chemical , Molecular Structure , Rats , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship
15.
Neurobiol Learn Mem ; 91(4): 393-401, 2009 May.
Article in English | MEDLINE | ID: mdl-19100850

ABSTRACT

The present study investigated whether the selective nociceptin opioid peptide (NOP) receptor agonist, Ro64-6198, impairs acquisition of fear conditioning through glutamatergic mechanisms. Systemic administration of Ro64-6198 (0.3 and 1mg/kg) or the non-competitive NMDA receptor antagonist, MK-801 (0.03 and 0.1mg/kg) prior to conditioning severely impaired contextual but not cued fear learning in C57BL/6N mice. When administered together at sub-effective doses, Ro64-6198 (0.5mg/kg) and MK-801 (0.05mg/kg), synergistically impaired contextual fear learning, but left cued fear learning intact. We next used the immediate shock deficit paradigm (ISD) to examine the effects of Ro64-6198 and MK-801 on contextual memory formation in the absence of the foot-shock. As expected, naive mice that were shocked briefly after being placed in the training chamber displayed no contextual fear conditioning. This learning deficit was elevated by prior exposure of mice to the training context. Furthermore, administration of Ro64-6198 and MK-801, either separately at amnesic doses (1mg/kg and 0.1mg/kg, respectively) or concomitantly at sub-effective doses (0.5mg/kg and 0.05mg/kg, respectively) significantly reduced the facilitating effects of context preexposure. These findings demonstrate the existence of functional antagonism between NOP and NMDA receptors that predominantly contributes to modulation of conditioned fear learning which involves spatial-processing demands.


Subject(s)
Conditioning, Psychological/physiology , Fear , Glutamic Acid/metabolism , Receptors, Opioid/metabolism , Analgesics, Opioid/pharmacology , Analysis of Variance , Animals , Conditioning, Psychological/drug effects , Cues , Dizocilpine Maleate/pharmacology , Electroshock , Excitatory Amino Acid Antagonists/pharmacology , Imidazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Nociceptin Receptor
16.
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
17.
J Neurosci ; 28(9): 2190-8, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18305252

ABSTRACT

Strong evidence suggests a role for nociceptin/orphanin FQ (N/OFQ) neuropeptide and its receptor (NOP) in cognition. However, the signaling mechanisms underlying N/OFQ modulation of memory are less understood. Here, we show that intracerebroventricular or intrahippocampal infusions of N/OFQ impair long-term memory formation in the mouse object recognition task. The synthetic NOP receptor agonist, (1S,3aS)-8-(2,3,3a,4,5,6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-one (Ro64-6198), administered systemically, also produced amnesic effects that were blocked by coinfusion of the NOP receptor antagonist, [Nphe1,Arg14,Lys15]nociceptin-NH2 (UFP-101), into the dorsal hippocampus. In contrast, Ro64-6198 had no effect on short-term memory or recall performances. Immunoblotting analysis revealed a strong suppressive action of Ro64-6198 on learning-induced upregulation of hippocampal extracellular signal-regulated kinase (ERK) phosphorylation, which is crucial for long-term information storage. Accordingly, pharmacological inhibition of ERK activation after systemic injection of SL327 [alpha-[amino[(4-aminophenyl)thio]methylene]-2-(trifluoromethyl)benzene acetonitrile], a selective inhibitor of the upstream kinase MEK (mitogen-activated protein kinase kinase), abolished long-term recognition memory formation. The noncompetitive NMDA receptor antagonist (+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d]cyclohepten-5,10-imine maleate (MK-801), given systemically, also suppressed ERK activation and disrupted recognition memory. In contrast, no effect of MK-801 was observed on recall, as for Ro64-6198. When administered concurrently at subthreshold doses, Ro64-6198 and MK-801 synergistically suppressed hippocampal ERK activation and impaired long-term memory formation. Under resting conditions, neither Ro64-6198 nor MK-801 affected spontaneous ERK activity in the hippocampus at the amnesic doses whereas at higher doses, only MK-801 had a suppressive effect. We conclude that N/OFQ-NOP receptor system negatively regulates long-term recognition memory formation through hippocampal ERK signaling mechanisms. This modulation may in part take place by inhibiting glutamatergic function at the NMDA receptor.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Hippocampus/drug effects , Memory/drug effects , Opioid Peptides/pharmacology , Pattern Recognition, Visual/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/physiology , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Neuropsychological Tests , Signal Transduction/drug effects , Nociceptin
18.
Bioorg Med Chem Lett ; 18(3): 1091-5, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18096387

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 a cyano group by a five-membered heterocycle produced compounds inhibiting the binding of [(3)H]-LY354740 to rat mGluR2 with low nanomolar affinity and consistent functional effect at both mGluR2 and mGluR3. Further modification to improve the physicochemical properties led eventually to compounds with the ability to reverse LY354740-mediated inhibition of field excitatory postsynaptic potentials in the rat dentate gyrus.


Subject(s)
Azepines/chemical synthesis , Azepines/pharmacology , Bridged Bicyclo Compounds/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Animals , Azepines/chemistry , CHO Cells , Cricetinae , Cricetulus , Molecular Structure , Rats , Structure-Activity Relationship
19.
ChemMedChem ; 3(2): 323-35, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18058780

ABSTRACT

The asymmetric synthesis and receptor pharmacology of (1S,2R,3R,5R,6S)-2-amino-3-Hydroxy-bicyclo[3.1.0]hexane-2,6-dicarboxylic Acid (+)-9 (HYDIA) and a few of its O-alkylated derivatives are described. The key step of the synthesis utilizes Sharpless' asymmetric dihydroxylation (AD-beta) for the kinetic resolution of a bicyclic racemic precursor olefin. In contrast to the bicyclic glutamate analogue LY354740, which is a potent and selective agonist for the group II metabotropic glutamate receptors (mGluRs), these new conformationally restricted and also hydroxylated or alkoxylated glutamate analogues are potent and selective antagonists for the group II mGluRs.


Subject(s)
Bridged Bicyclo Compounds/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Receptors, Metabotropic Glutamate/agonists , Alkylation , Animals , Binding, Competitive , Bridged Bicyclo Compounds/chemical synthesis , Excitatory Amino Acid Agonists/chemical synthesis , Glutamic Acid/chemistry , Glutamic Acid/pharmacology , Hydroxylation , Ligands , Mice , Stereoisomerism , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 17(24): 6811-5, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17964783

ABSTRACT

A series of 1,3-dihydrobenzo[b][1,4]diazepin-2-one derivatives was evaluated as non-competitive mGluR2/3 antagonists. Attachment of an 8-(2-aryl)-ethynyl-moiety produced compounds inhibiting the binding of [(3)H]-LY354740 to rat mGluR2 with low nanomolar affinity and consistent functional effect at both mGluR2 and mGluR3.


Subject(s)
Azepines/chemical synthesis , Azepines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Animals , Azepines/chemistry , CHO Cells , Cricetinae , Cricetulus , Molecular Structure , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...