Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Cell ; 161(6): 1252-65, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046436

ABSTRACT

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Subject(s)
Drug Discovery , Small Molecule Libraries , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , National Institutes of Health (U.S.) , United States
2.
Molecules ; 29(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38893312

ABSTRACT

Gain-of-function mutations in the KCNT1 gene, which encodes the sodium-activated potassium channel known as SLACK, are associated with the rare but devastating developmental and epileptic encephalopathy known as epilepsy of infancy with migrating focal seizures (EIMFS). The design of small molecule inhibitors of SLACK channels represents a potential therapeutic approach to the treatment of EIMFS, other childhood epilepsies, and developmental disorders. Herein, we describe a hit optimization effort centered on a xanthine SLACK inhibitor (8) discovered via a high-throughput screen. Across three distinct regions of the chemotype, we synthesized 58 new analogs and tested each one in a whole-cell automated patch-clamp assay to develop structure-activity relationships for inhibition of SLACK channels. We further evaluated selected analogs for their selectivity versus a variety of other ion channels and for their activity versus clinically relevant SLACK mutants. Selectivity within the series was quite good, including versus hERG. Analog 80 (VU0948578) was a potent inhibitor of WT, A934T, and G288S SLACK, with IC50 values between 0.59 and 0.71 µM across these variants. VU0948578 represents a useful in vitro tool compound from a chemotype that is distinct from previously reported small molecule inhibitors of SLACK channels.


Subject(s)
Potassium Channel Blockers , Structure-Activity Relationship , Humans , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels, Sodium-Activated , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Xanthine/chemistry , Xanthine/pharmacology , Patch-Clamp Techniques , HEK293 Cells , Molecular Structure , Xanthines/chemistry , Xanthines/pharmacology
3.
Bioorg Med Chem ; 95: 117487, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37812884

ABSTRACT

Malignant migrating partial seizure of infancy (MMPSI) is a devastating and pharmacoresistant form of infantile epilepsy. MMPSI has been linked to multiple gain-of-function (GOF) mutations in the KCNT1 gene, which encodes for a potassium channel often referred to as SLACK. SLACK channels are sodium-activated potassium channels distributed throughout the central nervous system (CNS) and the periphery. The investigation described here aims to discover SLACK channel inhibitor tool compounds and profile their pharmacokinetic and pharmacodynamic properties. A SLACK channel inhibitor VU0531245 (VU245) was identified via a high-throughput screen (HTS) campaign. Structure-activity relationship (SAR) studies were conducted in five distinct regions of the hit VU245. VU245 analogs were evaluated for their ability to affect SLACK channel activity using a thallium flux assay in HEK-293 cells stably expressing wild-type (WT) human SLACK. Selected analogs were tested for metabolic stability in mouse liver microsomes and plasma-protein binding in mouse plasma. The same set of analogs was tested via thallium flux for activity versus human A934T SLACK and other structurally related potassium channels, including SLICK and Maxi-K. In addition, potencies for selected VU245 analogs were obtained using whole-cell electrophysiology (EP) assays in CHO cells stably expressing WT human SLACK through an automated patch clamp system. Results revealed that this scaffold tolerates structural changes in some regions, with some analogs demonstrating improved SLACK inhibitory activity, good selectivity against the other channels tested, and modest improvements in metabolic clearance. Analog VU0935685 represents a new, structurally distinct small-molecule inhibitor of SLACK channels that can serve as an in vitro tool for studying this target.


Subject(s)
Potassium Channels , Thallium , Animals , Cricetinae , Humans , Mice , Cricetulus , HEK293 Cells , Nerve Tissue Proteins/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels, Sodium-Activated/genetics , Potassium Channels, Sodium-Activated/metabolism , Seizures , Thallium/metabolism , Oxadiazoles/chemistry , Oxadiazoles/metabolism
4.
Bioorg Med Chem Lett ; 76: 129013, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36184030

ABSTRACT

In this Letter we describe structure-activity relationship (SAR) studies conducted in five distinct regions of a new 2-amino-N-phenylacetamides series of Slack potassium channel inhibitors exemplified by recently disclosed high-throughput screening (HTS) hit VU0606170 (4). New analogs were screened in a thallium (Tl+) flux assay in HEK-293 cells stably expressing wild-type human (WT) Slack. Selected analogs were screened in Tl+ flux versus A934T Slack and other Slo family members Slick and Maxi-K and evaluated in whole-cell electrophysiology (EP) assays using an automated patch clamp system. Results revealed the series to have flat SAR with significant structural modifications resulting in a loss of Slack activity. More minor changes led to compounds with Slack activity and Slo family selectivity similar to the HTS hit.


Subject(s)
Potassium Channels , Thallium , Humans , HEK293 Cells , Nerve Tissue Proteins/metabolism , Potassium Channels, Sodium-Activated , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 29(1): 47-50, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30446311

ABSTRACT

This letter describes the further chemical optimization of VU0424238 (auglurant), an mGlu5 NAM clinical candidate that failed in non-human primate (NHP) 28 day toxicology due to accumulation of a species-specific aldehyde oxidase (AO) metabolite of the pyrimidine head group. Here, we excised the pyrimidine moiety, identified the minimum pharmacophore, and then developed a new series of saturated ether head groups that ablated any AO contribution to metabolism. Putative back-up compounds in this novel series provided increased sp3 character, uniform CYP450-mediated metabolism across species, good functional potency and high CNS penetration. Key to the optimization was a combination of matrix and iterative libraries that allowed rapid surveillance of multiple domains of the allosteric ligand.


Subject(s)
Drug Discovery , Picolinic Acids/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Structure , Picolinic Acids/chemical synthesis , Picolinic Acids/chemistry , Rats , Receptor, Metabotropic Glutamate 5/metabolism , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 29(2): 342-346, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30503632

ABSTRACT

This letter describes the first account of the chemical optimization (SAR and DMPK profiling) of a new series of mGlu4 positive allosteric modulators (PAMs), leading to the identification of VU0652957 (VU2957, Valiglurax), a compound profiled as a preclinical development candidate. Here, we detail the challenges faced in allosteric modulator programs (e.g., steep SAR, as well as subtle structural changes affecting overall physiochemical/DMPK properties and CNS penetration).


Subject(s)
Drug Discovery , Heterocyclic Compounds, 2-Ring/pharmacology , Isoquinolines/pharmacology , Myotonin-Protein Kinase/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation/drug effects , Dose-Response Relationship, Drug , Heterocyclic Compounds, 2-Ring/chemistry , Humans , Isoquinolines/chemistry , Molecular Structure , Myotonin-Protein Kinase/metabolism , Structure-Activity Relationship
7.
Br J Cancer ; 119(12): 1538-1551, 2018 12.
Article in English | MEDLINE | ID: mdl-30385822

ABSTRACT

BACKGROUND: Although NGLY1 is known as a pivotal enzyme that catalyses the deglycosylation of denatured glycoproteins, information regarding the responses of human cancer and normal cells to NGLY1 suppression is limited. METHODS: We examined how NGLY1 expression affects viability, tumour growth, and responses to therapeutic agents in melanoma cells and an animal model. Molecular mechanisms contributing to NGLY1 suppression-induced anticancer responses were revealed by systems biology and chemical biology studies. Using computational and medicinal chemistry-assisted approaches, we established novel NGLY1-inhibitory small molecules. RESULTS: Compared with normal cells, NGLY1 was upregulated in melanoma cell lines and patient tumours. NGLY1 knockdown caused melanoma cell death and tumour growth retardation. Targeting NGLY1 induced pleiotropic responses, predominantly stress signalling-associated apoptosis and cytokine surges, which synergise with the anti-melanoma activity of chemotherapy and targeted therapy agents. Pharmacological and molecular biology tools that inactivate NGLY1 elicited highly similar responses in melanoma cells. Unlike normal cells, melanoma cells presented distinct responses and high vulnerability to NGLY1 suppression. CONCLUSION: Our work demonstrated the significance of NGLY1 in melanoma cells, provided mechanistic insights into how NGLY1 inactivation leads to eradication of melanoma with limited impact on normal cells, and suggested that targeting NGLY1 represents a novel anti-melanoma strategy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Interferon-gamma/physiology , Melanoma/drug therapy , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/antagonists & inhibitors , Activating Transcription Factor 4/physiology , Animals , Cells, Cultured , Cytokines/analysis , Gene Expression Profiling , Humans , Interferon-gamma/genetics , Melanoma/pathology , Mice , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/physiology , Pluripotent Stem Cells/physiology , Proteasome Endopeptidase Complex/physiology , Signal Transduction/physiology , Transcription Factor CHOP/physiology
8.
Bioorg Med Chem Lett ; 28(10): 1679-1685, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29705142

ABSTRACT

Based on previous work that established fused heterocycles as viable alternatives for the picolinamide core of our lead series of mGlu5 negative allosteric modulators (NAMs), we designed a novel series of 6-(pyrimidin-5-ylmethyl)quinoline-8-carboxamide mGlu5 NAMs. These new quinoline derivatives also contained carbon linkers as replacements for the diaryl ether oxygen atom common to our previously published chemotypes. Compounds were evaluated in a cell-based functional mGlu5 assay, and an exemplar analog 27 was >60-fold selective versus the other seven mGlu receptors. Selected compounds were also studied in metabolic stability assays in rat and human S9 hepatic fractions and exhibited a mixture of P450- and non-P450-mediated metabolism.


Subject(s)
Drug Discovery , Pyrimidines/pharmacology , Quinolines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Dose-Response Relationship, Drug , Humans , Male , Molecular Structure , Pyrimidines/chemistry , Quinolines/chemistry , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/metabolism , Structure-Activity Relationship
9.
Chem Rev ; 116(11): 6707-41, 2016 06 08.
Article in English | MEDLINE | ID: mdl-26882314

ABSTRACT

Allosteric modulation of GPCRs has initiated a new era of basic and translational discovery, filled with therapeutic promise yet fraught with caveats. Allosteric ligands stabilize unique conformations of the GPCR that afford fundamentally new receptors, capable of novel pharmacology, unprecedented subtype selectivity, and unique signal bias. This review provides a comprehensive overview of the basics of GPCR allosteric pharmacology, medicinal chemistry, drug metabolism, and validated approaches to address each of the major challenges and caveats. Then, the review narrows focus to highlight recent advances in the discovery of allosteric ligands for metabotropic glutamate receptor subtypes 1-5 and 7 (mGlu1-5,7) highlighting key concepts ("molecular switches", signal bias, heterodimers) and practical solutions to enable the development of tool compounds and clinical candidates. The review closes with a section on late-breaking new advances with allosteric ligands for other GPCRs and emerging data for endogenous allosteric modulators.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation , Drug Design , Humans , Kinetics , Ligands , Receptors, G-Protein-Coupled/chemistry , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/genetics , Structure-Activity Relationship
10.
Nucleic Acids Res ; 44(9): 4189-99, 2016 05 19.
Article in English | MEDLINE | ID: mdl-26873923

ABSTRACT

RAD52 is a member of the homologous recombination (HR) pathway that is important for maintenance of genome integrity. While single RAD52 mutations show no significant phenotype in mammals, their combination with mutations in genes that cause hereditary breast cancer and ovarian cancer like BRCA1, BRCA2, PALB2 and RAD51C are lethal. Consequently, RAD52 may represent an important target for cancer therapy. In vitro, RAD52 has ssDNA annealing and DNA strand exchange activities. Here, to identify small molecule inhibitors of RAD52 we screened a 372,903-compound library using a fluorescence-quenching assay for ssDNA annealing activity of RAD52. The obtained 70 putative inhibitors were further characterized using biochemical and cell-based assays. As a result, we identified compounds that specifically inhibit the biochemical activities of RAD52, suppress growth of BRCA1- and BRCA2-deficient cells and inhibit RAD52-dependent single-strand annealing (SSA) in human cells. We will use these compounds for development of novel cancer therapy and as a probe to study mechanisms of DNA repair.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Rad52 DNA Repair and Recombination Protein/antagonists & inhibitors , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , DNA Damage , Drug Screening Assays, Antitumor , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Protein Binding , Rad52 DNA Repair and Recombination Protein/chemistry
11.
Proc Natl Acad Sci U S A ; 112(4): 1196-201, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25583490

ABSTRACT

Clinical studies have revealed that genetic variations in metabotropic glutamate receptor 3 (mGlu3) affect performance on cognitive tasks dependent upon the prefrontal cortex (PFC) and may be linked to psychiatric conditions such as schizophrenia, bipolar disorder, and addiction. We have performed a series of studies aimed at understanding how mGlu3 influences PFC function and cognitive behaviors. In the present study, we found that activation of mGlu3 can induce long-term depression in the mouse medial PFC (mPFC) in vitro. Furthermore, in vivo administration of a selective mGlu3 negative allosteric modulator impaired learning in the mPFC-dependent fear extinction task. The results of these studies implicate mGlu3 as a major regulator of PFC function and cognition. Additionally, potentiators of mGlu3 may be useful in alleviating prefrontal impairments associated with several CNS disorders.


Subject(s)
Behavior, Animal , Cognition , Fear , Long-Term Synaptic Depression , Prefrontal Cortex/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred ICR , Mice, Knockout , Rats , Receptors, Metabotropic Glutamate/genetics
12.
Bioorg Med Chem Lett ; 27(21): 4858-4866, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28958625

ABSTRACT

Based on a hypothesis that an intramolecular hydrogen bond was present in our lead series of picolinamide mGlu5 NAMs, we reasoned that an inactive nicotinamide series could be modified through introduction of a fused heterocyclic core to generate potent mGlu5 NAMs. In this Letter, we describe the synthesis and evaluation of compounds that demonstrate the viability of that approach. Selected analogs were profiled in a variety of in vitro assays, and two compounds were evaluated in rat pharmacokinetic studies and a mouse model of obsessive-compulsive disorder. Ancillary pharmacology screening revealed that members of this series exhibited moderate inhibition of the dopamine transporter (DAT), and SAR was developed that expanded the selectivity for mGlu5 versus DAT.


Subject(s)
Amides/chemistry , Receptor, Metabotropic Glutamate 5/metabolism , Allosteric Regulation , Amides/pharmacokinetics , Amides/pharmacology , Animals , Cell Membrane Permeability/drug effects , Dogs , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Plasma Membrane Transport Proteins/metabolism , Drug Evaluation, Preclinical , Half-Life , Humans , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Mice , Microsomes, Liver/metabolism , Pyridines/chemistry , Rats , Receptor, Metabotropic Glutamate 5/chemistry , Structure-Activity Relationship , Triazoles/chemistry
13.
J Pharmacol Exp Ther ; 356(1): 123-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26503377

ABSTRACT

Negative allosteric modulators (NAMs) of metabotropic glutamate receptor subtype 5 (mGlu5) have potential applications in the treatment of fragile X syndrome, levodopa-induced dyskinesia in Parkinson disease, Alzheimer disease, addiction, and anxiety; however, clinical and preclinical studies raise concerns that complete blockade of mGlu5 and inverse agonist activity of current mGlu5 NAMs contribute to adverse effects that limit the therapeutic use of these compounds. We report the discovery and characterization of a novel mGlu5 NAM, N,N-diethyl-5-((3-fluorophenyl)ethynyl)picolinamide (VU0477573) that binds to the same allosteric site as the prototypical mGlu5 NAM MPEP but displays weak negative cooperativity. Because of this weak cooperativity, VU0477573 acts as a "partial NAM" so that full occupancy of the MPEP site does not completely inhibit maximal effects of mGlu5 agonists on intracellular calcium mobilization, inositol phosphate (IP) accumulation, or inhibition of synaptic transmission at the hippocampal Schaffer collateral-CA1 synapse. Unlike previous mGlu5 NAMs, VU0477573 displays no inverse agonist activity assessed using measures of effects on basal [(3)H]inositol phosphate (IP) accumulation. VU0477573 acts as a full NAM when measuring effects on mGlu5-mediated extracellular signal-related kinases 1/2 phosphorylation, which may indicate functional bias. VU0477573 exhibits an excellent pharmacokinetic profile and good brain penetration in rodents and provides dose-dependent full mGlu5 occupancy in the central nervous system (CNS) with systemic administration. Interestingly, VU0477573 shows robust efficacy, comparable to the mGlu5 NAM MTEP, in models of anxiolytic activity at doses that provide full CNS occupancy of mGlu5 and demonstrate an excellent CNS occupancy-efficacy relationship. VU0477573 provides an exciting new tool to investigate the efficacy of partial NAMs in animal models.


Subject(s)
GABA Agonists/pharmacology , Picolinic Acids/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Allosteric Regulation/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Behavior, Animal/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Drug Discovery , GABA Agonists/pharmacokinetics , HEK293 Cells , Humans , Inositol Phosphates/metabolism , MAP Kinase Signaling System/drug effects , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Picolinic Acids/pharmacokinetics , Pyridines/metabolism , Radioligand Assay , Rats , Receptor, Metabotropic Glutamate 5/metabolism , Synaptic Transmission/drug effects
14.
Drug Metab Dispos ; 44(8): 1296-303, 2016 08.
Article in English | MEDLINE | ID: mdl-26936972

ABSTRACT

Marketed drugs cleared by aldehyde oxidase (AO) are few, with no known clinically relevant pharmacokinetic drug interactions associated with AO inhibition, whereas cytochrome P450 (P450) inhibition or induction mediates a number of clinical drug interactions. Little attention has been given to the consequences of coadministering a P450 inhibitor with a compound metabolized by both AO and P450. Upon discovering that VU0409106 (1) was metabolized by AO (to M1) and P450 enzymes (to M4-M6), we sought to evaluate the in vivo disposition of 1 and its metabolites in rats with attenuated P450 activity. Male rats were orally pretreated with the pan-P450 inactivator, 1-aminobenzotriazole (ABT), before an i.p. dose of 1. Interestingly, the plasma area under the curve (AUC) of M1 was increased 15-fold in ABT-treated rats, indicating a metabolic shunt toward AO resulted from the drug interaction condition. The AUC of 1 also increased 7.8-fold. Accordingly, plasma clearance of 1 decreased from 53.5 to 15.3 ml/min per kilogram in ABT-pretreated rats receiving an i.v. dose of 1. Consistent with these data, M1 formation in hepatic S9 increased with NADPH-exclusion to eliminate P450 activity (50% over reactions containing NADPH). These studies reflect possible consequences of a drug interaction between P450 inhibitors and compounds cleared by both AO and P450 enzymes. Notably, increased exposure to an AO metabolite may hold clinical relevance for active metabolites or those mediating toxicity at elevated concentrations. The recent rise in clinical drug candidates metabolized by AO underscores the importance of these findings and the need for clinical studies to fully understand these risks.


Subject(s)
Aldehyde Oxidase/metabolism , Benzamides/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Thiazoles/pharmacokinetics , Triazoles/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Benzamides/administration & dosage , Benzamides/metabolism , Biotransformation , Cytochrome P-450 Enzyme Inhibitors/administration & dosage , Drug Interactions , Humans , Male , Metabolic Clearance Rate , Microsomes, Liver/enzymology , Rats, Sprague-Dawley , Risk Assessment , Substrate Specificity , Thiazoles/administration & dosage , Thiazoles/metabolism , Triazoles/administration & dosage
15.
Bioorg Med Chem Lett ; 26(8): 1894-900, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26988308

ABSTRACT

Selective negative allosteric modulators (NAMs) of each of the group I metabotropic glutamate receptors (mGlu1 and mGlu5) have been well characterized in the literature and offer potential as therapeutics in several disorders of the central nervous system (CNS). Still, compounds that are potent mGlu1/5 NAMs with selectivity versus the other six members of the mGlu family as well as the balance of properties required for use in vivo are lacking. A medicinal chemistry effort centered on the identification of a lead series with the potential of delivering such compounds is described in this Letter. Specifically, a new class of pyrido[1',2':1,5]pyrazolo[4,3-d]pyrimidin-4-amines was designed as a novel isosteric replacement for 4-aminoquinazolines, and compounds from within this chemotype exhibited dual NAM activity at both group I mGlus. One compound, VU0467558 (29), demonstrated near equipotent activity at both receptors, selectivity versus other mGlus, a favorable ancillary pharmacology profile, and CNS exposure in rodents.


Subject(s)
Allosteric Regulation/drug effects , Central Nervous System/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Animals , Central Nervous System/metabolism , Dose-Response Relationship, Drug , Male , Mice , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 26(13): 3029-3033, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27185330

ABSTRACT

This Letter describes the chemical optimization of a novel series of M4 positive allosteric modulators (PAMs) based on a 5,6-dimethyl-4-(piperidin-1-yl)thieno[2,3-d]pyrimidine core, identified from an MLPCN functional high-throughput screen. The HTS hit was potent and selective, but not CNS penetrant. Potency was maintained, while CNS penetration was improved (rat brain:plasma Kp=0.74), within the original core after several rounds of optimization; however, the thieno[2,3-d]pyrimidine core was subject to extensive oxidative metabolism. Ultimately, we identified a 6-fluoroquinazoline core replacement that afforded good M4 PAM potency, muscarinic receptor subtype selectivity and CNS penetration (rat brain:plasma Kp>10). Moreover, this campaign provided fundamentally distinct M4 PAM chemotypes, greatly expanding the available structural diversity for this exciting CNS target.


Subject(s)
Piperidines/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Receptor, Muscarinic M4/metabolism , Thiophenes/pharmacology , Allosteric Regulation , Animals , Brain/drug effects , Brain/metabolism , Humans , Microsomes, Liver/metabolism , Piperidines/chemical synthesis , Piperidines/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Quinazolines/chemical synthesis , Quinazolines/metabolism , Rats , Receptor, Muscarinic M4/agonists , Receptor, Muscarinic M4/antagonists & inhibitors , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/metabolism
17.
Bioorg Med Chem Lett ; 24(15): 3307-14, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24969015

ABSTRACT

Development of SAR in an aryl ether series of mGlu5 NAMs leading to the identification of pyrazine analog VU0431316 is described in this Letter. VU0431316 is a potent and selective non-competitive antagonist of mGlu5 that binds at a known allosteric binding site. VU0431316 demonstrates an attractive DMPK profile, including moderate clearance and good bioavailability in rats. Intraperitoneal (IP) dosing of VU0431316 in a mouse marble burying model of anxiety, an assay known to be sensitive to mGlu5 antagonists and other anxiolytics, produced dose proportional effects.


Subject(s)
Anxiety/drug therapy , Disease Models, Animal , Drug Discovery , Picolinic Acids/pharmacology , Pyrazines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Allosteric Site/drug effects , Animals , Dose-Response Relationship, Drug , Humans , Male , Mice , Molecular Structure , Picolinic Acids/administration & dosage , Picolinic Acids/chemistry , Pyrazines/administration & dosage , Pyrazines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 23(13): 3713-8, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23727046

ABSTRACT

Development of SAR in an N-acyl-N'-arylpiperazine series of negative allosteric modulators of mGlu1 using a functional cell-based assay is described in this Letter. Characterization of selected compounds in protein binding assays was used to aid in selecting VU0469650 for further profiling in ancillary pharmacology assays and pharmacokinetic studies. VU0469650 demonstrated an excellent selectivity profile and good exposure in both plasma and brain samples following intraperitoneal dosing in rats.


Subject(s)
Allosteric Regulation/drug effects , Central Nervous System/drug effects , Piperazines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Central Nervous System/metabolism , Dose-Response Relationship, Drug , Molecular Structure , Piperazines/chemical synthesis , Piperazines/chemistry , Rats , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship
19.
Bioorg Med Chem Lett ; 23(18): 5091-6, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23932792

ABSTRACT

Development of SAR in an octahydropyrrolo[3,4-c]pyrrole series of negative allosteric modulators of mGlu1 using a functional cell-based assay is described in this Letter. The octahydropyrrolo[3,4-c]pyrrole scaffold was chosen as an isosteric replacement for the piperazine ring found in the initial hit compound. Characterization of selected compounds in protein binding assays was used to identify the most promising analogs, which were then profiled in P450 inhibition assays in order to further assess the potential for drug-likeness within this series of compounds.


Subject(s)
Enzyme Inhibitors/pharmacology , Pyrroles/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Pyrroles/chemical synthesis , Pyrroles/chemistry , Rats , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 23(21): 5779-85, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24074843

ABSTRACT

Development of SAR in an aryl ether series of mGlu5 NAMs leading to the identification of tool compound VU0409106 is described in this Letter. VU0409106 is a potent and selective negative allosteric modulator of mGlu5 that binds at the known allosteric binding site and demonstrates good CNS exposure following intraperitoneal dosing in mice. VU0409106 also proved efficacious in a mouse marble burying model of anxiety, an assay known to be sensitive to mGlu5 antagonists as well as clinically efficacious anxiolytics.


Subject(s)
Anxiety/drug therapy , Benzamides/therapeutic use , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Thiazoles/therapeutic use , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Animals , Benzamides/chemistry , Benzamides/pharmacokinetics , Mice , Receptor, Metabotropic Glutamate 5/metabolism , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL