1
|
Krámos B, Hadady Z, Makó A, Szántó G, Felföldi N, Magdó I, Bobok AÁ, Bata I, Román V, Visegrády A, Keserű G, Greiner I, Éles J. Novel-Type GABA B PAMs: Structure-Activity Relationship in Light of the Protein Structure. ACS Med Chem Lett 2024; 15:396-405. [PMID: 38505850 PMCID: PMC10945541 DOI: 10.1021/acsmedchemlett.3c00560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Selecting a known HTS hit with the pyrazolo[1,5-a]pyrimidine core, our project was started from CMPPE, and its optimization was driven by a ligand-based pharmacophore model developed on the basis of published GABAB positive allosteric modulators (PAMs). Our primary goal was to improve the potency by finding new enthalpic interactions. Therefore, we included the lipophilic ligand efficiency (LLE or LipE) as an objective function in the optimization that led to a carboxylic acid derivative (34). This lead candidate offers the possibility to improve potency without drastically inflating the physicochemical properties. Although the discovery of the novel carboxyl feature was surprising, it turned out to be an important element of the GABAB PAM pharmacophore that can be perfectly explained based on the new protein structures. Rationalizing the binding mode of 34, we analyzed the intersubunit PAM binding site of GABAB receptor using the publicly available experimental structures.
Collapse
Affiliation(s)
- Balázs Krámos
- Spectroscopic
Research Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Zsuzsa Hadady
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Attila Makó
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Gábor Szántó
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Nóra Felföldi
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Ildikó Magdó
- Spectroscopic
Research Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Amrita Ágnes Bobok
- Pharmacological
and Drug Safety Research, Gedeon Richter
Plc., Gyömrői
út 19-21, Budapest, 1103 Hungary
| | - Imre Bata
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Viktor Román
- Pharmacological
and Drug Safety Research, Gedeon Richter
Plc., Gyömrői
út 19-21, Budapest, 1103 Hungary
| | - András Visegrády
- Pharmacological
and Drug Safety Research, Gedeon Richter
Plc., Gyömrői
út 19-21, Budapest, 1103 Hungary
| | - György
M. Keserű
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, Budapest, 1117 Hungary
| | - István Greiner
- Research
and Development Director, Gedeon Richter
Plc., Gyömrői
út 19-21, Budapest, 1103 Hungary
| | - János Éles
- Head
of Medicinal Chemistry, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| |
Collapse
|
2
|
The role of thalamic group II mGlu receptors in health and disease. Neuronal Signal 2022; 6:NS20210058. [PMID: 36561092 PMCID: PMC9760452 DOI: 10.1042/ns20210058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 12/25/2022] Open
Abstract
The thalamus plays a pivotal role in the integration and processing of sensory, motor, and cognitive information. It is therefore important to understand how the thalamus operates in states of both health and disease. In the present review, we discuss the function of the Group II metabotropic glutamate (mGlu) receptors within thalamic circuitry, and how they may represent therapeutic targets in treating disease states associated with thalamic dysfunction.
Collapse
|
3
|
Abstract
Gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter in the brain, acts at the ionotropic GABAA and GABAC receptors, and the metabotropic GABAB receptor. This chapter summarizes the studies that have investigated the role of the GABAB receptor in stress-related psychiatric disorders including anxiety and mood disorders. Overall, clinical and preclinical evidences strongly suggest that the GABAB receptor is a therapeutic candidate for depression and anxiety disorders. However, the clinical development of GABAB receptor-based drugs to treat these disorders has been hampered by their potential side-effects, particularly those of agonists. Nevertheless, the discovery of novel GABAB receptor allosteric modulators, and increasing understanding of the influence of specific intracellular GABAB receptor-associated proteins on GABAB receptor activity, may now pave the way towards GABAB receptor therapeutics in the treatment of mood and anxiety disorders.
Collapse
Affiliation(s)
- Daniela Felice
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
4
|
Vlachou S. A Brief History and the Significance of the GABA B Receptor. Curr Top Behav Neurosci 2021; 52:1-17. [PMID: 34595739 DOI: 10.1007/7854_2021_264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the brain. GABA type B (GABAB) receptors (GABABRs) are the only metabotropic G protein-coupled receptors for GABA and can be found distributed not only in the central nervous system, but also in the periphery. This chapter introduces important, fundamental knowledge related to GABABR function and the various potential therapeutic applications of the development of novel GABABR-active compounds, as documented through extensive studies presented in subsequent chapters of this Current Topic in Behavioral Neurosciences volume on the role of the neurobiology of GABABR function. The compounds that have received increased attention in the last few years compared to GABABR agonists and antagonists - the positive allosteric modulators - exhibit better pharmacological profiles and fewer side effects. As we continue to unveil the mystery of GABABRs at the molecular and cellular levels, we further understand the significance of these receptors. Future directions should aim for developing highly selective GABABR compounds for treating neuropsychiatric disorders and their symptomatology.
Collapse
Affiliation(s)
- Styliani Vlachou
- Neuropsychopharmacology Division, Behavioural Neuroscience Laboratory, School of Psychology, Faculty of Science and Health, Dublin City University, Dublin, Ireland.
| |
Collapse
|
5
|
Pérez-Benito L, Llinas del Torrent C, Pardo L, Tresadern G. The computational modeling of allosteric modulation of metabotropic glutamate receptors. FROM STRUCTURE TO CLINICAL DEVELOPMENT: ALLOSTERIC MODULATION OF G PROTEIN-COUPLED RECEPTORS 2020; 88:1-33. [DOI: 10.1016/bs.apha.2020.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
6
|
Felsing DE, Jain MK, Allen JA. Advances in Dopamine D1 Receptor Ligands for Neurotherapeutics. Curr Top Med Chem 2019; 19:1365-1380. [PMID: 31553283 DOI: 10.2174/1568026619666190712210903] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/04/2019] [Accepted: 04/07/2019] [Indexed: 12/15/2022]
Abstract
The dopamine D1 receptor (D1R) is essential for neurotransmission in various brain pathways where it modulates key functions including voluntary movement, memory, attention and reward. Not surprisingly, the D1R has been validated as a promising drug target for over 40 years and selective activation of this receptor may provide novel neurotherapeutics for neurodegenerative and neuropsychiatric disorders. Several pharmacokinetic challenges with previously identified small molecule D1R agonists have been recently overcome with the discovery and advancement of new ligands, including drug-like non-catechol D1R agonists and positive allosteric modulators. From this, several novel molecules and mechanisms have recently entered clinical studies. Here we review the major classes of D1R selective ligands including antagonists, orthosteric agonists, non-catechol biased agonists and positive allosteric modulators, highlighting their structure-activity relationships and medicinal chemistry. Recent chemistry breakthroughs and innovative approaches to selectively target and activate the D1R also hold promise for creating pharmacotherapy for several neurological diseases.
Collapse
Affiliation(s)
- Daniel E Felsing
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555-0615, United States.,Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas, 77555-0615, United States
| | - Manish K Jain
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555-0615, United States.,Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas, 77555-0615, United States
| | - John A Allen
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555-0615, United States.,Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas, 77555-0615, United States
| |
Collapse
|
7
|
Hellyer SD, Albold S, Sengmany K, Singh J, Leach K, Gregory KJ. Metabotropic glutamate receptor 5 (mGlu 5 )-positive allosteric modulators differentially induce or potentiate desensitization of mGlu 5 signaling in recombinant cells and neurons. J Neurochem 2019; 151:301-315. [PMID: 31376155 DOI: 10.1111/jnc.14844] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/17/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022]
Abstract
Allosteric modulators of metabotropic glutamate receptor 5 (mGlu5 ) are a promising therapeutic strategy for a number of neurological disorders. Multiple mGlu5 -positive allosteric modulator (PAM) chemotypes have been discovered that act as either pure PAMs or as PAM-agonists in recombinant and native cells. While these compounds have been tested in paradigms of receptor activation, their effects on receptor regulatory processes are largely unknown. In this study, acute desensitization of mGlu5 mediated intracellular calcium mobilization by structurally diverse mGlu5 orthosteric and allosteric ligands was assessed in human embryonic kidney 293 cells and primary murine neuronal cultures from both striatum and cortex. We aimed to determine the intrinsic efficacy and modulatory capacity of diverse mGlu5 PAMs [(R)-5-((3-fluorophenyl)ethynyl)-N-(3-hydroxy-3-methylbutan-2-yl)picolinamide (VU0424465), N-cyclobutyl-6-((3-fluorophenyl)ethynyl)picolinamide (VU0360172), 1-(4-(2,4-difluorophenyl)piperazin-1-yl)-2-((4-fluorobenzyl)oxy)ethanone (DPFE), ((4-fluorophenyl) (2-(phenoxymethyl)-6,7-dihydrooxazolo[5,4-c]pyridin-5(4H)-yl)methanone) (VU0409551), 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB)] on receptor desensitization and whether cellular context influences receptor regulatory processes. Only VU0424465 and VU0409551 induced desensitization alone in human embryonic kidney 293-mGlu5 cells, while all PAMs enhanced (S)-3,5-dihydroxyphenylglycine (DHPG)-induced desensitization. All mGlu5 PAMs induced receptor desensitization alone and enhanced DHPG-induced desensitization in striatal neurons. VU0424465 and VU0360172 were the only PAMs that induced desensitization alone in cortical neurons. With the exception of (CDPPB), PAMs enhanced DHPG-induced desensitization in cortical neurons. Moreover, differential apparent affinities, efficacies, and cooperativities with DHPG were observed for VU0360172, VU0409551, and VU0424465 when comparing receptor activation and desensitization in a cell type-dependent manner. These data indicate that biased mGlu5 allosteric modulator pharmacology extends to receptor regulatory processes in a tissue dependent manner, adding yet another layer of complexity to rational mGlu5 drug discovery.
Collapse
Affiliation(s)
- Shane D Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Sabine Albold
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Kathy Sengmany
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Junaid Singh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
8
|
Yu Y, Nguyen DT, Jiang J. G protein-coupled receptors in acquired epilepsy: Druggability and translatability. Prog Neurobiol 2019; 183:101682. [PMID: 31454545 DOI: 10.1016/j.pneurobio.2019.101682] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/09/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023]
Abstract
As the largest family of membrane proteins in the human genome, G protein-coupled receptors (GPCRs) constitute the targets of more than one-third of all modern medicinal drugs. In the central nervous system (CNS), widely distributed GPCRs in neuronal and nonneuronal cells mediate numerous essential physiological functions via regulating neurotransmission at the synapses. Whereas their abnormalities in expression and activity are involved in various neuropathological processes. CNS conditions thus remain highly represented among the indications of GPCR-targeted agents. Mounting evidence from a large number of animal studies suggests that GPCRs play important roles in the regulation of neuronal excitability associated with epilepsy, a common CNS disease afflicting approximately 1-2% of the population. Surprisingly, none of the US Food and Drug Administration (FDA)-approved (>30) antiepileptic drugs (AEDs) suppresses seizures through acting on GPCRs. This disparity raises concerns about the translatability of these preclinical findings and the druggability of GPCRs for seizure disorders. The currently available AEDs intervene seizures predominantly through targeting ion channels and have considerable limitations, as they often cause unbearable adverse effects, fail to control seizures in over 30% of patients, and merely provide symptomatic relief. Thus, identifying novel molecular targets for epilepsy is highly desired. Herein, we focus on recent progresses in understanding the comprehensive roles of several GPCR families in seizure generation and development of acquired epilepsy. We also dissect current hurdles hindering translational efforts in developing GPCRs as antiepileptic and/or antiepileptogenic targets and discuss the counteracting strategies that might lead to a potential cure for this debilitating CNS condition.
Collapse
Affiliation(s)
- Ying Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Drug Discovery Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Davis T Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Drug Discovery Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Drug Discovery Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
9
|
Luderman KD, Conroy JL, Free RB, Southall N, Ferrer M, Sanchez-Soto M, Moritz AE, Willette BKA, Fyfe TJ, Jain P, Titus S, Hazelwood LA, Aubé J, Lane JR, Frankowski KJ, Sibley DR. Identification of Positive Allosteric Modulators of the D 1 Dopamine Receptor That Act at Diverse Binding Sites. Mol Pharmacol 2018; 94:1197-1209. [PMID: 30068735 PMCID: PMC6117505 DOI: 10.1124/mol.118.113175] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/27/2018] [Indexed: 01/11/2023] Open
Abstract
The D1 dopamine receptor is linked to a variety of neuropsychiatric disorders and represents an attractive drug target for the enhancement of cognition in schizophrenia, Alzheimer disease, and other disorders. Positive allosteric modulators (PAMs), with their potential for greater selectivity and larger therapeutic windows, may represent a viable drug development strategy, as orthosteric D1 receptor agonists possess known clinical liabilities. We discovered two structurally distinct D1 receptor PAMs, MLS6585 and MLS1082, via a high-throughput screen of the NIH Molecular Libraries program small-molecule library. Both compounds potentiate dopamine-stimulated G protein- and β-arrestin-mediated signaling and increase the affinity of dopamine for the D1 receptor with low micromolar potencies. Neither compound displayed any intrinsic agonist activity. Both compounds were also found to potentiate the efficacy of partial agonists. We tested maximally effective concentrations of each PAM in combination to determine if the compounds might act at separate or similar sites. In combination, MLS1082 + MLS6585 produced an additive potentiation of dopamine potency beyond that caused by either PAM alone for both β-arrestin recruitment and cAMP accumulation, suggesting diverse sites of action. In addition, MLS6585, but not MLS1082, had additive activity with the previously described D1 receptor PAM "Compound B," suggesting that MLS1082 and Compound B may share a common binding site. A point mutation (R130Q) in the D1 receptor was found to abrogate MLS1082 activity without affecting that of MLS6585, suggesting this residue may be involved in the binding/activity of MLS1082 but not that of MLS6585. Together, MLS1082 and MLS6585 may serve as important tool compounds for the characterization of diverse allosteric sites on the D1 receptor as well as the development of optimized lead compounds for therapeutic use.
Collapse
Affiliation(s)
- Kathryn D Luderman
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Jennie L Conroy
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - R Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Noel Southall
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Marc Ferrer
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Marta Sanchez-Soto
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Amy E Moritz
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Blair K A Willette
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Tim J Fyfe
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Prashi Jain
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Steve Titus
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Lisa A Hazelwood
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Jeffrey Aubé
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - J Robert Lane
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - Kevin J Frankowski
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland (K.D.L., J.L.C., R.B.F., M.S.-S., A.E.M., B.K.A.W., L.A.H., D.R.S.); NIH Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.S., M.F., S.T.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (T.J.F., J.R.L.); KU Specialized Chemistry Center, University of Kansas, Lawrence, Kansas (P.J., J.A., K.J.F.); and Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.A., K.J.F.)
| |
Collapse
|
10
|
Efficacy and side effects of baclofen and the novel GABA B receptor positive allosteric modulator CMPPE in animal models for alcohol and cocaine addiction. Psychopharmacology (Berl) 2018; 235:1955-1965. [PMID: 29651507 DOI: 10.1007/s00213-018-4893-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/28/2018] [Indexed: 12/15/2022]
Abstract
RATIONALE Preclinical studies suggest that the GABAB receptor is a potential target for treatment of substance use disorders. However, recent clinical trials report adverse effects in patients treated with the GABAB receptor agonist baclofen and even question efficacy. How can the discrepancy between preclinical and clinical findings be explained? OBJECTIVE To test efficacy and adverse effects of baclofen and the novel GABAB positive allosteric modulator (PAM) CMPPE in rat addiction models, which were developed in accordance with DSM. METHODS We used a well-characterized rat model of long-term alcohol consumption with repeated deprivation phases that result in compulsive alcohol drinking in a relapse situation, and a rat model of long-term intravenous cocaine self-administration resulting in key symptoms of addictive behavior. We tested repeated baclofen (0, 1, and 3 mg/kg; i.p.) and CMPPE doses (0, 10, and 30 mg/kg; i.p.) in relapse-like situations, in either alcohol or cocaine addicted-like rats. RESULTS Baclofen produced a weak anti-relapse effect at the highest dose in alcohol addicted-like rats, and this effect was mainly due to the treatment-induced sedation. CMPPE had a better profile, with a dose-dependent reduction of relapse-like alcohol drinking and without any signs of sedation. The cue-induced cocaine-seeking response was completely abolished by both compounds. CONCLUSION Positive allosteric modulation of the GABAB receptor provides efficacy, and no observable side effects in relapse behavior whereas baclofen may cause, not only sedation, but also considerable impairment of food intake or metabolism. However, targeting GABAB receptors may be effective in reducing certain aspects of addictive-like behavior, such as cue-reactivity.
Collapse
|
11
|
Elucidation of the neural circuits activated by a GABA B receptor positive modulator: Relevance to anxiety. Neuropharmacology 2017; 136:129-145. [PMID: 28734870 DOI: 10.1016/j.neuropharm.2017.07.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 01/09/2023]
Abstract
Although there is much evidence for a role of GABAB receptors in the pathophysiology of anxiety, the underlying neuronal mechanisms are largely unclear. The GABAB receptor allosteric positive modulator, GS39783, exerts anxiolytic effects without interfering with GABAB-mediated modulation of body temperature, cognitive performance and locomotor activity thus offering advantages over GABAB receptor agonists. However, the precise neural circuits underlying the anxiolytic effects of GS39783 are unknown. The aim of the present study was to identify brain structures and associated neuronal circuits that are modulated by GS39783 under either basal or mild stress conditions. To this end, the expression pattern of c-Fos, a marker of neuronal activation, was examined in mice acutely treated with GS39783 under basal conditions or following a mild anxiogenic challenge induced by exposure to the Open Arm (OA) of an Elevated Plus Maze. OA exposure enhanced c-Fos expression in vehicle-treated animals in several brain regions, including the medial prefrontal cortex, lateral septum, amygdala, hippocampus, paraventricular nucleus of the hypothalamus and the periaqueductal gray (PAG). Under basal conditions, GS39783 increased c-Fos in a restricted panel of areas notably amygdala nuclei, cortical areas and PAG subregions, while it inhibited c-Fos expression in the dorsal raphe nucleus (DRN). Under stress conditions, GS39783 reversed OA-induced c-Fos expression in the granular cell layer of the dentate gyrus, no longer increased c-Fos expression in the amygdala nor reduced c-Fos expression in the DRN. These specific patterns of neural activation by GS39783 might explain the neurobiological correlates implicated in GABAB-mediated anti-anxiety effects. This article is part of the "Special Issue Dedicated to Norman G. Bowery".
Collapse
|
12
|
Remesic M, Hruby VJ, Porreca F, Lee YS. Recent Advances in the Realm of Allosteric Modulators for Opioid Receptors for Future Therapeutics. ACS Chem Neurosci 2017; 8:1147-1158. [PMID: 28368571 DOI: 10.1021/acschemneuro.7b00090] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Opioids, and more specifically μ-opioid receptor (MOR) agonists such as morphine, have long been clinically used as therapeutics for severe pain states but often come with serious side effects such as addiction and tolerance. Many studies have focused on bringing about analgesia from the MOR with attenuated side effects, but its underlying mechanism is not fully understood. Recently, focus has been geared toward the design and elucidation of the orthosteric site with ligands of various biological profiles and mixed subtype opioid activities and selectivities, but targeting the allosteric site is an area of increasing interest. It has been shown that allosteric modulators play key roles in influencing receptor function such as its tolerance to a ligand and affect downstream pathways. There has been a high variance of chemical structures that provide allosteric modulation at a given receptor, but recent studies and reviews tend to focus on the altered cellular mechanisms instead of providing a more rigorous description of the allosteric ligand's structure-function relationship. In this review, we aim to explore recent developments in the structural motifs that potentiate orthosteric binding and their influences on cellular pathways in an effort to present novel approaches to opioid therapeutic design.
Collapse
Affiliation(s)
- Michael Remesic
- Department
of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| | - Victor J. Hruby
- Department
of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| | - Frank Porreca
- Department
of Pharmacology, University of Arizona, Tucson, Arizona 85719, United States
| | - Yeon Sun Lee
- Department
of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
13
|
Johnstone S, Albert JS. Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective. Bioorg Med Chem Lett 2017; 27:2239-2258. [PMID: 28408223 DOI: 10.1016/j.bmcl.2017.03.084] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/11/2022]
Abstract
New strategies to potentially improve drug safety and efficacy emerge with allosteric programs. Biased allosteric modulators can be designed with high subtype selectivity and defined receptor signaling endpoints, however, selecting the most meaningful parameters for optimization can be perplexing. Historically, "potency hunting" at the expense of physicochemical and pharmacokinetic optimization has led to numerous tool compounds with excellent pharmacological properties but no path to drug development. Conversely, extensive physicochemical and pharmacokinetic screening with only post hoc bias and allosteric characterization has led to inefficacious compounds or compounds with on-target toxicities. This field is rapidly evolving with new mechanistic understanding, changes in terminology, and novel opportunities. The intent of this digest is to summarize current understanding and debates within the field. We aim to discuss, from a medicinal chemistry perspective, the parameter choices available to drive SAR.
Collapse
Affiliation(s)
- Shawn Johnstone
- Department of Chemistry, IntelliSyn Pharma, 7171 Frederick-Banting, Montreal, Quebec H4S 1Z9, Canada.
| | - Jeffrey S Albert
- Department of Chemistry, IntelliSyn Pharma, 7171 Frederick-Banting, Montreal, Quebec H4S 1Z9, Canada; Department of Chemistry, AviSyn Pharma, 4275 Executive Square, Suite 200, La Jolla, CA 92037, United States.
| |
Collapse
|
14
|
Sturchler E, Li X, de Lourdes Ladino M, Kaczanowska K, Cameron M, Griffin PR, Finn MG, Markou A, McDonald P. GABA B receptor allosteric modulators exhibit pathway-dependent and species-selective activity. Pharmacol Res Perspect 2017; 5:e00288. [PMID: 28357120 PMCID: PMC5368958 DOI: 10.1002/prp2.288] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/02/2016] [Accepted: 11/30/2016] [Indexed: 01/22/2023] Open
Abstract
Positive modulation of the GABAB receptor (GABABR) represents a potentially useful therapeutic approach for the treatment of nicotine addiction. The positive allosteric modulators (PAMs) of GABABR GS39783 and BHF177 enhance GABA‐stimulated [35S]GTPγS‐binding, and have shown efficacy in a rodent nicotine self‐administration procedure reflecting aspects of nicotine dependence. Interestingly, the structural related analog, NVP998, had no effect on nicotine self‐administration in rats despite demonstrating similar pharmacokinetic properties. Extensive in vitro characterization of GS39783, BHF177, and NVP998 activity on GABABR‐regulated signaling events, including modulation of cAMP, intracellular calcium levels, and ERK activation, revealed that these structurally related molecules display distinct pathway‐specific signaling activities that correlate with the dissimilarities observed in rodent models and may be predictive of in vivo efficacy. Furthermore, these GABABR allosteric modulators exhibit species‐dependent activity. Collectively, these data will be useful in guiding the development of GABABR allosteric modulators that display optimal in vivo efficacy in a preclinical model of nicotine dependence, and will identify those that have the potential to lead to novel antismoking therapies.
Collapse
Affiliation(s)
- Emmanuel Sturchler
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - Xia Li
- Department of Psychiatry University of California San Diego 9500 Gilman Drive La Jolla California 92093
| | - Maria de Lourdes Ladino
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458; Present address: School of Medicine Vanderbilt University 2215 Garland Ave Nashville Tennessee 37232
| | - Kasia Kaczanowska
- Department of Chemistry The Scripps Research Institute 10550 North Torrey Pines Road La Jolla California 92037; Present address: Department of Chemistry University of California San Diego, 9500 Gilman Drive La Jolla California 92093
| | - Michael Cameron
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - Patrick R Griffin
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - M G Finn
- Department of Chemistry The Scripps Research Institute 10550 North Torrey Pines Road La Jolla California 92037; Present address: Georgia Institute of Technology School of Chemistry and Biochemistry 901 Atlantic Drive Atlanta Georgia 30332
| | - Athina Markou
- Department of Psychiatry University of California San Diego 9500 Gilman Drive La Jolla California 92093
| | - Patricia McDonald
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| |
Collapse
|
15
|
Kalinichev M, Donovan-Rodriguez T, Girard F, Haddouk H, Royer-Urios I, Schneider M, Bate ST, Marker C, Pomonis JD, Poli S. ADX71943 and ADX71441, novel positive allosteric modulators of the GABA B receptor with distinct central/peripheral profiles, show efficacy in the monosodium iodoacetate model of chronic osteoarthritis pain in the rat. Eur J Pharmacol 2016; 795:43-49. [PMID: 27916555 DOI: 10.1016/j.ejphar.2016.11.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022]
Abstract
We tested novel positive allosteric modulators (PAMs) of the γ-aminobutyric acid receptor B (GABAB), ADX71943 and ADX71441in the monosodium iodoacetate model of chronic osteoarthritis pain in rats with the objective to delineate the role of peripheral versus central GABAB receptor populations in modulation of chronic pain. Anesthetized Sprague-Dawley rats received an injection of monosodium iodoacetate into the knee and were tested for hyperalgesia starting post-MIA day 14. Effects of compounds on ipsilateral joint compression threshold were evaluated on post-MIA day 14 (after acute treatment), as well as after repeated, daily treatment on days 21 and 28 (ADX71943 only) and were compared to those of celecoxib (30mg/kg, p.o.). The PAMs were also tested in the rat rotarod test for potential muscle-relaxant effects. Acutely, ADX71943 (1-30mg/kg, p.o.), the peripherally restricted PAM, resulted in similar increases in pain threshold across the doses on day 14, while showing reduced efficacy on day 21 and no efficacy on day 28. A clear reduction in the efficacy of celecoxib across testing was also noted in this experiment. Acutely ADX71441 (0.3-15mg/kg, p.o.), the central-peripheral PAM, resulted in over 2-fold increases in pain threshold at 15mg/kg (but not at lower doses) on day 14, while causing more modest effects on day 21. Celecoxib increased pain threshold after both acute and daily treatment, showing overall similar efficacy. Thus, early, presumably more inflammatory phase of osteoarthritis pain in more sensitive to GABAB PAMs with peripherally restricted profile, while later, presumably more neuropathic phase is more sensitive to PAMs with central-peripheral profile.
Collapse
Affiliation(s)
- Mikhail Kalinichev
- Addex Therapeutics SA, Chemin des Mines 9, CH-1202, Geneva, Switzerland.
| | | | - Françoise Girard
- Addex Therapeutics SA, Chemin des Mines 9, CH-1202, Geneva, Switzerland
| | - Hasnaá Haddouk
- Addex Therapeutics SA, Chemin des Mines 9, CH-1202, Geneva, Switzerland
| | | | - Manfred Schneider
- Addex Therapeutics SA, Chemin des Mines 9, CH-1202, Geneva, Switzerland
| | - Simon T Bate
- Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Woolley Road Alconbury, Huntingdon PE28 4HS, UK
| | - Cheryl Marker
- Algos Preclinical Serves Inc., 2848 Patton Road, Roseville, MN 55113, USA
| | - James D Pomonis
- Algos Preclinical Serves Inc., 2848 Patton Road, Roseville, MN 55113, USA
| | - Sonia Poli
- Addex Therapeutics SA, Chemin des Mines 9, CH-1202, Geneva, Switzerland
| |
Collapse
|
16
|
Li X, D'Souza MS, Niño AM, Doherty J, Cross A, Markou A. Attenuation of nicotine-taking and nicotine-seeking behavior by the mGlu2 receptor positive allosteric modulators AZD8418 and AZD8529 in rats. Psychopharmacology (Berl) 2016; 233:1801-14. [PMID: 26873083 DOI: 10.1007/s00213-016-4220-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/18/2016] [Indexed: 12/29/2022]
Abstract
RATIONALE Numerous medication development strategies seek to decrease nicotine consumption and prevent relapse to tobacco smoking by blocking glutamate transmission. Decreasing glutamate release by activating presynaptic inhibitory metabotropic glutamate (mGlu)2/3 receptors inhibits the reinforcing effects of nicotine and blocks cue-induced reinstatement of nicotine-seeking behavior in rats. However, the relative contribution of mGlu2 receptors in nicotine dependence is still unknown. OBJECTIVES The present study evaluated the role of mGlu2 receptors in nicotine-taking and nicotine-seeking behavior using the novel, relatively selective mGlu2 positive allosteric modulators (PAMs) AZD8418 and AZD8529. RESULTS Acute treatment with AZD8418 (0.37, 1.12, 3.73, 7.46, and 14.92 mg/kg) and AZD8529 (1.75, 5.83, 17.5, and 58.3 mg/kg) deceased nicotine self-administration and had no effect on food-maintained responding. Chronic treatment with AZD8418 attenuated nicotine self-administration, but tolerance to this effect developed quickly. The inhibition of nicotine self-administration by chronic AZD8529 administration persisted throughout the 14 days of treatment. Chronic treatment with either PAMs inhibited food self-administration. AZD8418 (acute) and AZD8529 (acute and subchronic) blocked cue-induced reinstatement of nicotine- and food-seeking behavior. CONCLUSIONS These findings indicate an important role for mGlu2 receptors in the reinforcing properties of self-administered nicotine and the motivational impact of cues that were previously associated with nicotine administration (i.e., cue-induced reinstatement of nicotine-seeking behavior). Thus, mGlu2 PAMs may be useful medications to assist people to quit tobacco smoking and prevent relapse.
Collapse
Affiliation(s)
- Xia Li
- Department of Psychiatry, M/C 0603, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
| | - Manoranjan S D'Souza
- Department of Psychiatry, M/C 0603, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
| | - Ana M Niño
- Department of Psychiatry, M/C 0603, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
| | - James Doherty
- Present address: Sage Therapeutics, Cambridge, MA, 02142, USA
| | - Alan Cross
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, MA, 02139, USA
| | - Athina Markou
- Department of Psychiatry, M/C 0603, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA.
| |
Collapse
|
17
|
Ito R, Tsujihata Y, Suzuki M, Miyawaki K, Matsuda K, Takeuchi K. Fasiglifam/TAK-875, a Selective GPR40 Agonist, Improves Hyperglycemia in Rats Unresponsive to Sulfonylureas and Acts Additively with Sulfonylureas. J Pharmacol Exp Ther 2016; 357:217-27. [PMID: 26813930 DOI: 10.1124/jpet.115.230730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/21/2016] [Indexed: 03/08/2025] Open
Abstract
Sulfonylureas (SUs) are widely used insulin secretagogues, but they have adverse effects including hypoglycemia and secondary failure. Fasiglifam/TAK-875, a selective GPR40 agonist, enhances glucose-stimulated insulin secretion and improves hyperglycemia. In the present study, we compared the in vivo glucose-lowering effects of fasiglifam with SUs. The risk of secondary failure of fasiglifam and the efficacy in rats desensitized to SUs were also evaluated. Moreover, we assessed whether fasiglifam was effective when combined with SUs. In diabetic neonatally streptozotocin-induced rats 1.5 days after birth (N-STZ-1.5), oral administrations of fasiglifam (3-30 mg/kg) dose dependently improved glucose tolerance; the effect was greater than that of glibenclamide at maximal effective doses (glucose AUC: fasiglifam, -37.6%; glibenclamide, -12.3%). Although the glucose-lowering effects of glibenclamide (10 mg/kg/day) were completely diminished in N-STZ-1.5 rats after 4 weeks of treatment, effects were maintained in rats receiving fasiglifam (10 mg/kg/day), even after 15 weeks. Fasiglifam (3-10 mg/kg) was still effective in two models desensitized to SUs: 15-week glibenclamide-treated N-STZ-1.5 rats and aged Zucker diabetic fatty (ZDF) rats. Acute administration of fasiglifam (3 mg/kg) and glimepiride (10 mg/kg) in combination additively decreased glucose AUC (fasiglifam, -25.3%; glimepiride, -20.0%; combination, -43.1%). Although glimepiride (10 mg/kg) decreased plasma glucose below normal in nonfasted control rats, fasiglifam (3 mg/kg) maintained normoglycemia, and no further exaggeration of hypoglycemia was observed with combination treatment. These results indicate that GPR40 agonists could be more effective and durable than SUs. Our results also provide new insights into GPR40 pharmacology and rationale for the use of GPR40 agonists in diabetic patients with SU failure.
Collapse
Affiliation(s)
- Ryo Ito
- Cardiovascular and Metabolic Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Yoshiyuki Tsujihata
- Cardiovascular and Metabolic Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Masami Suzuki
- Cardiovascular and Metabolic Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Kazumasa Miyawaki
- Cardiovascular and Metabolic Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Kae Matsuda
- Cardiovascular and Metabolic Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Koji Takeuchi
- Cardiovascular and Metabolic Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| |
Collapse
|
18
|
Wang H, Zhang W, Wang X. Elucidation of a CGP7930 in vitro metabolite by liquid chromatography/electrospray ionization quadrupole time-of-flight tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2016; 30:491-496. [PMID: 26777679 DOI: 10.1002/rcm.7465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/15/2015] [Accepted: 11/17/2015] [Indexed: 06/05/2023]
Abstract
RATIONALE γ-Aminobutyric acid-B (GABAB ) receptors are widely expressed in the nervous system and have been implicated as targets for various neurological and psychiatric disorders. CGP7930 is a positive allosteric modulator of GABAB receptors. It has been demonstrated to reduce drug self-administration and has gained increased research as a potential psychotropic treatment. METHODS An in vitro metabolic system with liver microsomes of SD rats has been conducted and evaluated by probe drugs. The predominant in vitro metabolite of CGP7930 was identified and elucidated using liquid chromatography/electrospray ionization quadrupole time-of-flight tandem mass spectrometry (LC/ESI-QTOF-MS/MS). Its structure was determined by comparing the characteristic ions of CGP7930 and those of the metabolite, based on the accurate mass measurement by MS and the fragmentation pattern obtained by MS/MS. RESULTS We found that the main metabolic pathway of CGP7930 was via a monohydroxylation reaction and the hydroxylation site located at the terminal butyl-carbon. The collision-induced dissociation (CID) fragmentation of the hydroxylated metabolite underwent McLafferty rearrangement and α-cleavage. CONCLUSIONS This work provides an understanding of the in vitro metabolism of CGP7930, which is helpful for the further study of the development of potential drug candidates targeting GABAB receptors, for the treatment of depression. The work also demonstrates that the LC/ESI-QTOF-MS/MS method has the advantage of possibly determining the structures of drug metabolites without the use of standards.
Collapse
Affiliation(s)
- Haidong Wang
- Key Laboratory of Catalysis and Materials Science of the State Ethnic Affairs Commission and Ministry of Education, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan, Hubei, 430074, P.R. China
| | - Wenxiang Zhang
- Key Laboratory of Catalysis and Materials Science of the State Ethnic Affairs Commission and Ministry of Education, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan, Hubei, 430074, P.R. China
| | - Xian Wang
- Key Laboratory of Catalysis and Materials Science of the State Ethnic Affairs Commission and Ministry of Education, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan, Hubei, 430074, P.R. China
| |
Collapse
|
19
|
Brown JW, Moeller A, Schmidt M, Turner SC, Nimmrich V, Ma J, Rueter LE, van der Kam E, Zhang M. Anticonvulsant effects of structurally diverse GABA(B) positive allosteric modulators in the DBA/2J audiogenic seizure test: Comparison to baclofen and utility as a pharmacodynamic screening model. Neuropharmacology 2015; 101:358-69. [PMID: 26471422 DOI: 10.1016/j.neuropharm.2015.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 07/31/2015] [Accepted: 10/04/2015] [Indexed: 02/02/2023]
Abstract
The GABA(B) receptor has been indicated as a promising target for multiple CNS-related disorders. Baclofen, a prototypical orthosteric agonist, is used clinically for the treatment of spastic movement disorders, but is associated with unwanted side-effects, such as sedation and motor impairment. Positive allosteric modulators (PAM), which bind to a topographically-distinct site apart from the orthosteric binding pocket, may provide an improved side-effect profile while maintaining baclofen-like efficacy. GABA, the major inhibitory neurotransmitter in the CNS, plays an important role in the etiology and treatment of seizure disorders. Baclofen is known to produce anticonvulsant effects in the DBA/2J mouse audiogenic seizure test (AGS), suggesting it may be a suitable assay for assessing pharmacodynamic effects. Little is known about the effects of GABA(B) PAMs, however. The studies presented here sought to investigate the AGS test as a pharmacodynamic (PD) screening model for GABA(B) PAMs by comparing the profile of structurally diverse PAMs to baclofen. GS39783, rac-BHFF, CMPPE, A-1295120 (N-(3-(4-(4-chloro-3-fluorobenzyl)-6-methoxy-3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)phenyl)acetamide), and A-1474713 (N-(3-(4-(4-chlorobenzyl)-3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)phenyl)acetamide) all produced robust, dose-dependent anticonvulsant effects; a similar profile was observed with baclofen. Pre-treatment with the GABA(B) antagonist SCH50911 completely blocked the anticonvulsant effects of baclofen and CMPPE in the AGS test, indicating such effects are likely mediated by the GABA(B) receptor. In addition to the standard anticonvulsant endpoint of the AGS test, video tracking software was employed to assess potential drug-induced motor side-effects during the acclimation period of the test. This analysis was sensitive to detecting drug-induced changes in total distance traveled, which was used to establish a therapeutic index (TI = hypoactivity/anticonvulsant effects). Calculated TIs for A-1295120, CMPPE, rac-BHFF, GS39783, and A-1474713 were 5.31x, 5.00x, 4.74x, 3.41x, and 1.83x, respectively, whereas baclofen was <1. The results presented here suggest the DBA/2J mouse AGS test is a potentially useful screening model for detecting PD effects of GABA(B) PAMs and can provide an initial read-out on target-related motor side-effects. Furthermore, an improved TI was observed for PAMs compared to baclofen, indicating the PAM approach may be a viable therapeutic alternative to baclofen.
Collapse
Affiliation(s)
- Jordan W Brown
- Neuroscience Discovery, AbbVie, Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States.
| | - Achim Moeller
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, Ludwigshafen 67061, Germany
| | - Martin Schmidt
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, Ludwigshafen 67061, Germany
| | - Sean C Turner
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, Ludwigshafen 67061, Germany
| | - Volker Nimmrich
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, Ludwigshafen 67061, Germany
| | - Junli Ma
- Drug Metabolism and Pharmacokinetics, AbbVie, Inc., North Chicago, IL 60064, United States
| | - Lynne E Rueter
- Neuroscience Discovery, AbbVie, Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States
| | - Elizabeth van der Kam
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, Ludwigshafen 67061, Germany
| | - Min Zhang
- Neuroscience Discovery, AbbVie, Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States
| |
Collapse
|
20
|
Brown KM, Roy KK, Hockerman GH, Doerksen RJ, Colby DA. Activation of the γ-Aminobutyric Acid Type B (GABA(B)) Receptor by Agonists and Positive Allosteric Modulators. J Med Chem 2015; 58:6336-47. [PMID: 25856547 DOI: 10.1021/jm5018913] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Since the discovery of the GABA(B) agonist and muscle relaxant baclofen, there have been substantial advancements in the development of compounds that activate the GABA(B) receptor as agonists or positive allosteric modulators. For the agonists, most of the existing structure-activity data apply to understanding the role of substituents on the backbone of GABA as well as replacing the carboxylic acid and amine groups. In the cases of the positive allosteric modulators, the allosteric binding site(s) and structure-activity relationships are less well-defined; however, multiple classes of molecules have been discovered. The recent report of the X-ray structure of the GABA(B) receptor with bound agonists and antagonists provides new insights for the development of compounds that bind the orthosteric site of this receptor. From a therapeutic perspective, these data have enabled efforts in drug discovery in areas of addiction-related behavior, the treatment of anxiety, and the control of muscle contractility.
Collapse
Affiliation(s)
- Katie M Brown
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University , West Lafayette, Indiana 47907, United States
| | - Kuldeep K Roy
- Department of Biomolecular Sciences, University of Mississippi , University, Mississippi 38677, United States
| | - Gregory H Hockerman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University , West Lafayette, Indiana 47907, United States
| | - Robert J Doerksen
- Department of Biomolecular Sciences, University of Mississippi , University, Mississippi 38677, United States
| | - David A Colby
- Department of Biomolecular Sciences, University of Mississippi , University, Mississippi 38677, United States
| |
Collapse
|
21
|
Bowery NG. Reflections on more than 30 years association with Hanns. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 73:1-11. [PMID: 25637435 DOI: 10.1016/bs.apha.2014.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
I first met Hanns in 1977 and soon learnt of his extraordinary ability as a researcher. He became a friend as well as a mentor providing enthusiasm for my own research. I watched closely over the years how his research uncovered details of the association of the benzodiazepines and GABA and delineated the structural composition of the GABAA receptor associated with the action of individual drugs such as antianxiety and antiepileptic agents. His work produced many important contributions to medicine notable of which was the discovery of the first benzodiazepine antagonists, which are now routinely used in clinical practice. But for me his most important contribution was the discovery of the benzodiazepine receptor. During this time, my group uncovered a novel receptor for GABA and my progress in this work was encouraged and enhanced by discussions with Hanns.
Collapse
Affiliation(s)
- Norman G Bowery
- Department of Pharmacology, University of Birmingham Medical School, Edgbaston, United Kingdom.
| |
Collapse
|
22
|
Abstract
INTRODUCTION There is increasing evidence encouraging the development of drugs that positively modulate the γ-aminobutyric acid type B (GABA(B)) receptor for combating addiction. Compounds that target GABA(B) receptors are unique as anti-abuse therapies because of their impact against multiple addictive drugs. AREAS COVERED The authors present the basic information concerning the drug actions of GABA and GABA(B) receptor orthosteric agonists and positive allosteric modulators (PAM). Furthermore, they discuss several recent excellent reviews and newer results pertaining to GABA(B) receptor drug effects on responses to and self-administration of: alcohol (ethanol), nicotine, cocaine, (meth)amphetamine, and opioids. Preclinical and clinical data are considered. EXPERT OPINION Clinical data exist only for baclofen and mostly for alcohol use disorders. Additional trials are needed, but effects are promising. Whether PAMs, given alone or in combination with a direct GABA(B) receptor agonist, will be clinically effective and have fewer side effects requires investigation. The sedative effects of baclofen, a Food and Drug Administration (FDA)-approved drug, become less severe over time. Based on existing data, baclofen is well-tolerated. However, genetic and physiological differences are likely to contribute to individual responses to different therapeutic agents. The more immediate development of baclofen as a therapeutic for alcohol use disorders may be of significant benefit to some individuals.
Collapse
Affiliation(s)
- Tamara J Phillips
- Oregon Health & Science University, Veterans Affairs Medical Center, Department of Behavioral Neuroscience , 3710 SW US Veterans Hospital Rd, Portland, OR 97239 , USA +1 503 220 8262 Ext. 56674 ; +1 503 721 1029 ;
| | | |
Collapse
|
23
|
Conn PJ, Lindsley CW, Meiler J, Niswender CM. Opportunities and challenges in the discovery of allosteric modulators of GPCRs for treating CNS disorders. Nat Rev Drug Discov 2014; 13:692-708. [PMID: 25176435 PMCID: PMC4208620 DOI: 10.1038/nrd4308] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Novel allosteric modulators of G protein-coupled receptors (GPCRs) are providing fundamental advances in the development of GPCR ligands with high subtype selectivity and novel modes of efficacy that have not been possible with traditional approaches. As new allosteric modulators are advancing as drug candidates, we are developing an increased understanding of the major advantages and broad range of activities that can be achieved with these agents through selective modulation of specific signalling pathways, differential effects on GPCR homodimers versus heterodimers, and other properties. This understanding creates exciting opportunities, as well as unique challenges, in the optimization of novel therapeutic agents for disorders of the central nervous system.
Collapse
Affiliation(s)
- P Jeffrey Conn
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 1215D LH, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 1215D LH, USA
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 1215D LH, USA
| |
Collapse
|
24
|
Cid JM, Tresadern G, Duvey G, Lütjens R, Finn T, Rocher JP, Poli S, Vega JA, de Lucas AI, Matesanz E, Linares ML, Andrés JI, Alcazar J, Alonso JM, Macdonald GJ, Oehlrich D, Lavreysen H, Ahnaou A, Drinkenburg W, Mackie C, Pype S, Gallacher D, Trabanco AA. Discovery of 1-Butyl-3-chloro-4-(4-phenyl-1-piperidinyl)-(1H)-pyridone (JNJ-40411813): A Novel Positive Allosteric Modulator of the Metabotropic Glutamate 2 Receptor. J Med Chem 2014; 57:6495-512. [DOI: 10.1021/jm500496m] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- José María Cid
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | - Gary Tresadern
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | - Guillaume Duvey
- Addex Therapeutics, 12 Chemin
des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Robert Lütjens
- Addex Therapeutics, 12 Chemin
des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
| | | | | | | | - Juan Antonio Vega
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | - Ana Isabel de Lucas
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | - Encarnación Matesanz
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | - María Lourdes Linares
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | - José Ignacio Andrés
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | - Jesús Alcazar
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | - José Manuel Alonso
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| | | | | | | | | | | | | | | | | | - Andrés A. Trabanco
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain
| |
Collapse
|
25
|
QSAR design of triazolopyridine mGlu2 receptor positive allosteric modulators. J Mol Graph Model 2014; 53:82-91. [PMID: 25086773 DOI: 10.1016/j.jmgm.2014.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/08/2014] [Accepted: 07/10/2014] [Indexed: 01/07/2023]
Abstract
Two QSAR approaches were applied to assist the design and to prioritise the synthesis of new active mGlu2 receptor positive allosteric modulators (PAMs). With the aim to explore a particular point of substitution the models successfully prioritised molecules originating from chemistry ideas and a large virtual library. The two methods, 3D topomer CoMFA and support vector machines with 2D ECFP6 fingerprints, delivered good correlation and success in this prospective application. Fourteen molecules with different substituent decoration were identified by the in silico models and synthesised. They were found to be highly active and their mGlu2 receptor PAM activity (pEC50) was predicted within 0.3 and 0.4log units of error with the two methods. The value of the molecules and the models for the future of the project is discussed.
Collapse
|
26
|
Burford NT, Wehrman T, Bassoni D, O'Connell J, Banks M, Zhang L, Alt A. Identification of selective agonists and positive allosteric modulators for µ- and δ-opioid receptors from a single high-throughput screen. ACTA ACUST UNITED AC 2014; 19:1255-65. [PMID: 25047277 DOI: 10.1177/1087057114542975] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hetero-oligomeric complexes of G protein-coupled receptors (GPCRs) may represent novel therapeutic targets exhibiting different pharmacology and tissue- or cell-specific site of action compared with receptor monomers or homo-oligomers. An ideal tool for validating this concept pharmacologically would be a hetero-oligomer selective ligand. We set out to develop and execute a 1536-well high-throughput screen of over 1 million compounds to detect potential hetero-oligomer selective ligands using a β-arrestin recruitment assay in U2OS cells coexpressing recombinant µ- and δ-opioid receptors. Hetero-oligomer selective ligands may bind to orthosteric or allosteric sites, and we might anticipate that the formation of hetero-oligomers may provide novel allosteric binding pockets for ligand binding. Therefore, our goal was to execute the screen in such a way as to identify positive allosteric modulators (PAMs) as well as agonists for µ, δ, and hetero-oligomeric receptors. While no hetero-oligomer selective ligands were identified (based on our selection criteria), this single screen did identify numerous µ- and δ-selective agonists and PAMs as well as nonselective agonists and PAMs. To our knowledge, these are the first µ- and δ-opioid receptor PAMs described in the literature.
Collapse
Affiliation(s)
- Neil T Burford
- Leads Discovery and Optimization, Bristol-Myers Squibb Company, Wallingford, CT, USA
| | | | | | | | - Martyn Banks
- Leads Discovery and Optimization, Bristol-Myers Squibb Company, Wallingford, CT, USA
| | - Litao Zhang
- Leads Discovery and Optimization, Bristol-Myers Squibb Company, Wallingford, CT, USA
| | - Andrew Alt
- Leads Discovery and Optimization, Bristol-Myers Squibb Company, Wallingford, CT, USA
| |
Collapse
|
27
|
Chen LH, Sun B, Zhang Y, Xu TJ, Xia ZX, Liu JF, Nan FJ. Discovery of a Negative Allosteric Modulator of GABAB Receptors. ACS Med Chem Lett 2014; 5:742-7. [PMID: 25050158 PMCID: PMC4094264 DOI: 10.1021/ml500162z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 05/27/2014] [Indexed: 12/11/2022] Open
Abstract
Initialized from the scaffold of CGP7930, an allosteric agonist of GABAB receptors, a series of noncompetitive antagonists were discovered. Among these compounds, compounds 3, 6, and 14 decreased agonist GABA-induced maximal effect of IP3 production in HEK293 cells overexpressing GABAB receptors and Gqi9 proteins without changing the EC50. Compounds 3, 6, and 14 not only inhibited agonist baclofen-induced ERK1/2 phosphorylation but also blocked CGP7930-induced ERK1/2 phosphorylation in HEK293 cells overexpressing GABAB receptors. The results suggested that compounds 3, 6, and 14 are negative allosteric modulators of GABAB receptors. The representative compound 14 decreased GABA-induced IP3 production with IC50 of 37.9 μM and had no effect on other GPCR Class C members such as mGluR1, mGluR2, and mGluR5. Finally, we showed that compound 14 did not bind to the orthosteric binding sites of GABAB receptors, demonstrating that compound 14 negatively modulated GABAB receptors activity as a negative allosteric modulator.
Collapse
Affiliation(s)
- Lin-Hai Chen
- National
Center for Drug Screening, State Key Laboratory of Drug Research,
Shanghai Institute of Materia Medica, Chinese
Academy of Sciences, Shanghai, China
| | - Bing Sun
- Cellular
Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry
of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Zhang
- Cellular
Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry
of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tong-Jie Xu
- Cellular
Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry
of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhi-Xiong Xia
- Cellular
Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry
of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jian-Feng Liu
- Cellular
Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry
of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fa-Jun Nan
- National
Center for Drug Screening, State Key Laboratory of Drug Research,
Shanghai Institute of Materia Medica, Chinese
Academy of Sciences, Shanghai, China
| |
Collapse
|
28
|
Burford NT, Traynor JR, Alt A. Positive allosteric modulators of the μ-opioid receptor: a novel approach for future pain medications. Br J Pharmacol 2014; 172:277-86. [PMID: 24460691 DOI: 10.1111/bph.12599] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/09/2014] [Accepted: 01/19/2014] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED Morphine and other agonists of the μ-opioid receptor are used clinically for acute and chronic pain relief and are considered to be the gold standard for pain medication. However, these opioids also have significant side effects, which are also mediated via activation of the μ-opioid receptor. Since the latter half of the twentieth century, researchers have sought to tease apart the mechanisms underlying analgesia, tolerance and dependence, with the hope of designing drugs with fewer side effects. These efforts have revolved around the design of orthosteric agonists with differing pharmacokinetic properties and/or selectivity profiles for the different opioid receptor types. Recently, μ-opioid receptor-positive allosteric modulators (μ-PAMs) were identified, which bind to a (allosteric) site on the μ-opioid receptor separate from the orthosteric site that binds an endogenous agonist. These allosteric modulators have little or no detectable functional activity when bound to the receptor in the absence of orthosteric agonist, but can potentiate the activity of bound orthosteric agonist, seen as an increase in apparent potency and/or efficacy of the orthosteric agonist. In this review, we describe the potential advantages that a μ-PAM approach might bring to the design of novel therapeutics for pain that may lack the side effects currently associated with opioid therapy. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- N T Burford
- GPCR Lead Discovery & Optimization, Bristol-Myers Squibb Company, Wallingford, CT, USA
| | | | | |
Collapse
|
29
|
Effect of fendiline on the maintenance and expression of methamphetamine-induced conditioned place preference in Sprague-Dawley rats. Psychopharmacology (Berl) 2014; 231:2019-29. [PMID: 24264565 PMCID: PMC3988275 DOI: 10.1007/s00213-013-3347-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 10/25/2013] [Indexed: 01/30/2023]
Abstract
RATIONALE Fendiline is a GABAB receptor-positive allosteric modulator and L-type Ca²⁺ channel blocker that is safe for human use. Based on these pharmacological properties, fendiline may be useful to disrupt associative memories that can drive relapse to drug use in drug-addicted individuals OBJECTIVE The current study evaluated the potential of fendiline to inhibit the maintenance and expression of learned associations between methamphetamine (meth) and an environmental context using conditioned place preference (CPP) in rats, to model for the associative learning that occurs during drug abuse by humans METHODS Following meth conditioning (1 mg/kg), fendiline (5 mg/kg) was administered at various post-conditioning times to ascertain if there was a temporal window during which fendiline would be effective. RESULTS Two once-daily injections of fendiline did not influence the maintenance of CPP regardless of the post-conditioning treatment time while 10 once-daily fendiline treatments inhibited CPP maintenance (p < 0.05). Fendiline administered immediately prior to the CPP test inhibited expression of meth-induced CPP in rats with a fendiline treatment history of 10 once-daily injections (p < 0.05) or those that received two injections that corresponded to the last 2 days of the 10-day treatment (p < 0.05). Fendiline did not produce preference or aversion on its own, nor did it alter motivated motor behavior. CONCLUSION Maintenance and expression of meth CPP is mitigated by repeated fendiline treatments when administered during the days that precede CPP testing. Reduction in the significance of meth-associated cues can reduce relapse; therefore, fendiline may be of value for addiction therapy in abstinent, meth-addicted humans.
Collapse
|
30
|
McCarson KE, Enna SJ. GABA pharmacology: the search for analgesics. Neurochem Res 2014; 39:1948-63. [PMID: 24532294 DOI: 10.1007/s11064-014-1254-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/28/2014] [Accepted: 01/31/2014] [Indexed: 12/28/2022]
Abstract
Decades of research have been devoted to defining the role of GABAergic transmission in nociceptive processing. Much of this work was performed using rigid, orthosteric GABA analogs created by Povl Krogsgaard-Larsen and his associates. A relationship between GABA and pain is suggested by the anatomical distribution of GABA receptors and the ability of some GABA agonists to alter nociceptive responsiveness. Outlined in this report are data supporting this proposition, with particular emphasis on the anatomical localization and function of GABA-containing neurons and the molecular and pharmacological properties of GABAA and GABAB receptor subtypes. Reference is made to changes in overall GABAergic tone, GABA receptor expression and activity as a function of the duration and intensity of a painful stimulus or exposure to GABAergic agents. Evidence is presented that the plasticity of this receptor system may be responsible for the variability in the antinociceptive effectiveness of compounds that influence GABA transmission. These findings demonstrate that at least some types of persistent pain are associated with a regionally selective decline in GABAergic tone, highlighting the need for agents that enhance GABA activity in the affected regions without compromising GABA function over the long-term. As subtype selective positive allosteric modulators may accomplish these goals, such compounds might represent a new class of analgesic drugs.
Collapse
Affiliation(s)
- Kenneth E McCarson
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Boulevard, Mail Stop 1018, Kansas City, KS, 66160, USA
| | | |
Collapse
|
31
|
Discovery of positive allosteric modulators and silent allosteric modulators of the μ-opioid receptor. Proc Natl Acad Sci U S A 2013; 110:10830-5. [PMID: 23754417 DOI: 10.1073/pnas.1300393110] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
μ-Opioid receptors are among the most studied G protein-coupled receptors because of the therapeutic value of agonists, such as morphine, that are used to treat chronic pain. However, these drugs have significant side effects, such as respiratory suppression, constipation, allodynia, tolerance, and dependence, as well as abuse potential. Efforts to fine tune pain control while alleviating the side effects of drugs, both physiological and psychological, have led to the development of a wide variety of structurally diverse agonist ligands for the μ-opioid receptor, as well as compounds that target κ- and δ-opioid receptors. In recent years, the identification of allosteric ligands for some G protein-coupled receptors has provided breakthroughs in obtaining receptor subtype-selectivity that can reduce the overall side effect profiles of a potential drug. However, positive allosteric modulators (PAMs) can also have the specific advantage of only modulating the activity of the receptor when the orthosteric agonist occupies the receptor, thus maintaining spatial and temporal control of receptor signaling in vivo. This second advantage of allosteric modulators may yield breakthroughs in opioid receptor research and could lead to drugs with improved side-effect profiles or fewer tolerance and dependence issues compared with orthosteric opioid receptor agonists. Here, we describe the discovery and characterization of μ-opioid receptor PAMs and silent allosteric modulators, identified from high-throughput screening using a β-arrestin-recruitment assay.
Collapse
|
32
|
Trabanco AA, Cid JM. mGluR2 positive allosteric modulators: a patent review (2009 - present). Expert Opin Ther Pat 2013; 23:629-47. [PMID: 23452205 DOI: 10.1517/13543776.2013.777043] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The mGlu2 receptor, which belongs to the group II subfamily of metabotropic glutamate receptors (mGlu) along with the mGlu3 receptor, has proven to be of particular importance in neuropharmacology. Preferentially expressed on presynaptic nerve terminals, the mGlu2 receptor negatively modulates glutamate and GABA release and is widely distributed in the brain. High levels of mGlu2 receptors are seen in brain areas such as prefrontal cortex, hippocampus and amygdala where glutamate hyperfunction may be implicated in disorders and diseases such as anxiety and schizophrenia. Given the promise offered by mGlu2/3 receptor activation, there is increased interest in identifying small molecules which activate the receptor. A preferred approach is via positive allosteric modulators (PAMs) which bind at an alternative site to agonists. AREAS COVERED This review covers the patent applications which were published between April 2009 and December 2012 on PAMs of the mGlu2, and it is a continuation of an earlier review published in this journal. EXPERT OPINION Advances in medicinal chemistry and pharmacology have set the stage in the field of mGlu2 receptor PAMs. Compounds currently advancing in clinical trials will soon establish the therapeutic potential of this allosteric approach.
Collapse
Affiliation(s)
- Andrés A Trabanco
- Janssen Research and Development, Neuroscience Medicinal Chemistry Department, Toledo, Spain.
| | | |
Collapse
|
33
|
Cid JM, Tresadern G, Vega JA, de Lucas AI, Matesanz E, Iturrino L, Linares ML, Garcia A, Andrés JI, Macdonald GJ, Oehlrich D, Lavreysen H, Megens A, Ahnaou A, Drinkenburg W, Mackie C, Pype S, Gallacher D, Trabanco AA. Discovery of 3-cyclopropylmethyl-7-(4-phenylpiperidin-1-yl)-8-trifluoromethyl[1,2,4]triazolo[4,3-a]pyridine (JNJ-42153605): a positive allosteric modulator of the metabotropic glutamate 2 receptor. J Med Chem 2012; 55:8770-89. [PMID: 23072213 DOI: 10.1021/jm3010724] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Advanced leads from a series of 1,2,4-triazolo[4,3-a]pyridines with mGlu2 receptor PAM activity are reported. By modification of the analogous imidazo[1,2-a]pyridine series, the newly reported leads have improved potency, in vitro ADMET, and hERG as well as good in vivo PK profile. The optimization of the series focused on improving metabolic stability while controlling lipophilicity by introducing small modifications to the scaffold substituents. Analysis of this series combined with our previously reported mGlu2 receptor PAMs showed how lipophilic ligand efficiency was improved during the course of the program. Among the best compounds, example 20 (JNJ-42153605) showed a central in vivo efficacy by inhibition of REM sleep state at a dose of 3 mg/kg po in the rat sleep-wake EEG paradigm, a phenomenon shown earlier to be mGlu2 mediated. In mice, compound 20 reversed PCP-induced hyperlocomotion with an ED₅₀ of 5.4 mg/kg sc, indicative of antipsychotic activity.
Collapse
Affiliation(s)
- Jose María Cid
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007 Toledo, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Andrés JI, Alcázar J, Cid JM, De Angelis M, Iturrino L, Langlois X, Lavreysen H, Trabanco AA, Celen S, Bormans G. Synthesis, evaluation, and radiolabeling of new potent positive allosteric modulators of the metabotropic glutamate receptor 2 as potential tracers for positron emission tomography imaging. J Med Chem 2012; 55:8685-99. [PMID: 22992024 DOI: 10.1021/jm300912k] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The synthesis and in vitro and in vivo evaluation of a new series of 7-(phenylpiperidinyl)-1,2,4-triazolo[4,3-a]pyridines, which were conveniently radiolabeled with carbon-11, as potential positron emission tomography (PET) radiotracers for in vivo imaging of the allosteric binding site of the metabotropic glutamate (mGlu) receptor subtype 2 are described. The synthesized compounds proved to be potent and selective positive allosteric modulators (PAMs) of the mGlu receptor 2 (mGluR2) in a [³⁵S]GTPγS binding assay and were able to displace an mGluR2 PAM radioligand, which we had previously developed, with IC₅₀ values in the low nanomolar range. The most promising candidates were radiolabeled and subjected to biodistribution studies and radiometabolite analysis in rats. Preliminary small-animal PET (μPET) studies in rats indicated that [¹¹C]20f binds specifically and reversibly to an mGluR2 allosteric site, strongly suggesting that it is a promising candidate for PET imaging of mGluR2 in the brain.
Collapse
Affiliation(s)
- José-Ignacio Andrés
- Medicinal Chemistry, Janssen Research & Development , Janssen-Cilag S.A., C/Jarama 75, 45007 Toledo, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kruse LC, Linsenbardt DN, Boehm SL. Positive allosteric modulation of the GABA(B) receptor by GS39783 attenuates the locomotor stimulant actions of ethanol and potentiates the induction of locomotor sensitization. Alcohol 2012; 46:455-62. [PMID: 22560291 DOI: 10.1016/j.alcohol.2012.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 03/12/2012] [Accepted: 03/20/2012] [Indexed: 12/20/2022]
Abstract
Acute ethanol-induced locomotor stimulation and ethanol-induced locomotor sensitization are two behavioral assays thought to model the rewarding effects of ethanol. Recent evidence suggests that GS39783, a GABA(B) positive allosteric modulator, may be effective at reducing both the rewarding and reinforcing effects of several drugs of abuse, including ethanol. The goal of this study was to determine if GS39783 was capable of altering acute ethanol-induced stimulation, and the induction and expression of ethanol-induced locomotor sensitization, without effecting basal locomotion levels. Several doses of GS39783 (ranging from 0 to 100 mg/kg, depending on experiment) were tested on adult male DBA/2J mice in four experiments using 3-day basal locomotion and acute ethanol stimulation paradigms, and 14-day induction and expression of ethanol sensitization paradigms. The results of experiment 1 are in agreement with current literature, suggesting that 30 mg/kg doses of GS39783 and lower do not alter basal locomotor activity. In experiment 2, we found that GS39783 significantly decreased acute ethanol stimulation, but only at the 30 mg/kg dose, supporting our hypothesis and other publications suggesting that GABA(B) receptors modulate acute ethanol stimulation. Contrary to our hypothesis, GS39783 did not alter the expression of locomotor sensitization. Additionally, repeated administration of GS39783 in conjunction with ethanol unexpectedly potentiated ethanol-induced locomotor sensitization. Further study of GS39783 is warranted as it may be a more tolerable treatment for alcoholism than full agonists, due to its behavioral efficacy at doses that lack sedative side effects. Our results add to current literature suggesting that the GABA(B) receptor system is indeed involved in the modulation of ethanol-induced locomotor stimulation and sensitization.
Collapse
Affiliation(s)
- Lauren C Kruse
- Department of Psychology, Indiana Alcohol Research Center, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | | | | |
Collapse
|
36
|
Noblin DJ, Bertekap RL, Burford NT, Hendricson A, Zhang L, Knox R, Banks M, O'Connell J, Alt A. Development of a high-throughput calcium flux assay for identification of all ligand types including positive, negative, and silent allosteric modulators for G protein-coupled receptors. Assay Drug Dev Technol 2012; 10:457-67. [PMID: 22746835 DOI: 10.1089/adt.2011.443] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In recent years, the increased use of cell-based functional assays for G protein-coupled receptors in high-throughput screening has enabled the design of robust assays to identify allosteric modulators (AMs) in addition to the more traditional orthosteric agonists and antagonists. In this article, the authors describe a screening format able to identify all ligand types using a triple-add assay that measures changes in cytosolic calcium concentration with three separate additions and reads in the same assay plate. This triple-add assay captures more small molecule ligand types than previously described assay formats without a significant increase in screening cost. Finally, the customizability of the triple-add assay to suit the needs of various AM screening programs is demonstrated.
Collapse
Affiliation(s)
- Devin J Noblin
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lahti JL, Tang GW, Capriotti E, Liu T, Altman RB. Bioinformatics and variability in drug response: a protein structural perspective. J R Soc Interface 2012; 9:1409-37. [PMID: 22552919 DOI: 10.1098/rsif.2011.0843] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Marketed drugs frequently perform worse in clinical practice than in the clinical trials on which their approval is based. Many therapeutic compounds are ineffective for a large subpopulation of patients to whom they are prescribed; worse, a significant fraction of patients experience adverse effects more severe than anticipated. The unacceptable risk-benefit profile for many drugs mandates a paradigm shift towards personalized medicine. However, prior to adoption of patient-specific approaches, it is useful to understand the molecular details underlying variable drug response among diverse patient populations. Over the past decade, progress in structural genomics led to an explosion of available three-dimensional structures of drug target proteins while efforts in pharmacogenetics offered insights into polymorphisms correlated with differential therapeutic outcomes. Together these advances provide the opportunity to examine how altered protein structures arising from genetic differences affect protein-drug interactions and, ultimately, drug response. In this review, we first summarize structural characteristics of protein targets and common mechanisms of drug interactions. Next, we describe the impact of coding mutations on protein structures and drug response. Finally, we highlight tools for analysing protein structures and protein-drug interactions and discuss their application for understanding altered drug responses associated with protein structural variants.
Collapse
Affiliation(s)
- Jennifer L Lahti
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | | | | | | |
Collapse
|
38
|
Trabanco AA, Tresadern G, Macdonald GJ, Vega JA, de Lucas AI, Matesanz E, García A, Linares ML, Alonso de Diego SA, Alonso JM, Oehlrich D, Ahnaou A, Drinkenburg W, Mackie C, Andrés JI, Lavreysen H, Cid JM. Imidazo[1,2-a]pyridines: Orally Active Positive Allosteric Modulators of the Metabotropic Glutamate 2 Receptor. J Med Chem 2012; 55:2688-701. [PMID: 22352782 DOI: 10.1021/jm201561r] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Andrés A. Trabanco
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Gary Tresadern
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Gregor J. Macdonald
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Juan Antonio Vega
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Ana Isabel de Lucas
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Encarnación Matesanz
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Aránzazu García
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - María Lourdes Linares
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Sergio A. Alonso de Diego
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - José Manuel Alonso
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Daniel Oehlrich
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Abdelah Ahnaou
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Wilhelmus Drinkenburg
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Claire Mackie
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - José Ignacio Andrés
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Hilde Lavreysen
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - José María Cid
- Neuroscience Medicinal Chemistry and ‡Research Informatics and Integrative Genomics, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, and ⊥Discovery ADME/Tox, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| |
Collapse
|
39
|
Castelli MP, Casu A, Casti P, Lobina C, Carai MAM, Colombo G, Solinas M, Giunta D, Mugnaini C, Pasquini S, Tafi A, Brogi S, Gessa GL, Corelli F. Characterization of COR627 and COR628, two novel positive allosteric modulators of the GABA(B) receptor. J Pharmacol Exp Ther 2012; 340:529-38. [PMID: 22129594 DOI: 10.1124/jpet.111.186460] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The potential efficacy of GABA(B) receptor agonists in the treatment of pain, drug addiction, epilepsy, cognitive dysfunctions, and anxiety disorders is supported by extensive preclinical and clinical evidence. However, the numerous side effects produced by the GABA(B) receptor agonist baclofen considerably limit the therapeutic use of this compound. The identification of positive allosteric modulators (PAMs) of the GABA(B) receptor may constitute a novel approach in the pharmacological manipulation of the GABA(B) receptor, leading to fewer side effects. The present study reports the identification of two novel compounds, methyl 2-(1-adamantanecarboxamido)-4-ethyl-5-methylthiophene-3-carboxylate (COR627) and methyl 2-(cyclohexanecarboxamido)-4-ethyl-5-methylthiophene-3-carboxylate (COR628), which act as GABA(B) PAMs in 1) rat cortical membranes and 2) in vivo assay. Both compounds potentiated GABA- and baclofen-stimulated guanosine 5'-O-(3-[(35)S]thio)-triphosphate binding to native GABA(B) receptors, while producing no effect when given alone. GABA concentration-response curves in the presence of fixed concentrations of COR627 and COR628 revealed an increase of potency of GABA rather than its maximal efficacy. In radioligand binding experiments [displacement of the GABA(B) receptor antagonist, 3-N-[1-((S)-3,4dichlorophenyl)-ethylaminol]-2-(S)hydroxypropyl cyclo-hexylmethyl phosphinic acid ([(3)H]CGP54626)], both COR627 and COR628 increased the affinity of high- and low-affinity binding sites for GABA, producing no effect when administered alone up to a concentration of 1 mM. In vivo experiments indicated that pretreatment with per se ineffective doses of COR627 and COR628 potentiated the sedative/hypnotic effect of baclofen. In conclusion, COR627 and COR628 may represent two additional tools for use in investigating the roles and functions of positive allosteric modulatory binding sites of the GABA(B) receptor.
Collapse
Affiliation(s)
- M Paola Castelli
- Department of Neuroscience Bernard B. Brodie, University of Cagliari, Cagliari, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cid JM, Duvey G, Tresadern G, Nhem V, Furnari R, Cluzeau P, Vega JA, de Lucas AI, Matesanz E, Alonso JM, Linares ML, Andrés JI, Poli SM, Lutjens R, Himogai H, Rocher JP, Macdonald GJ, Oehlrich D, Lavreysen H, Ahnaou A, Drinkenburg W, Mackie C, Trabanco AA. Discovery of 1,4-disubstituted 3-cyano-2-pyridones: a new class of positive allosteric modulators of the metabotropic glutamate 2 receptor. J Med Chem 2012; 55:2388-405. [PMID: 22364337 DOI: 10.1021/jm2016864] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The discovery and characterization of compound 48, a selective and in vivo active mGlu2 receptor positive allosteric modulator (PAM), are described. A key to the discovery was the rational exploration of the initial HTS hit 13 guided by an overlay model built with reported mGlu2 receptor PAM chemotypes. The initial weak in vitro activity of the hit 13 was quickly improved, although compounds still had suboptimal druglike properties. Subsequent modulation of the physicochemical properties resulted in compounds having a more balanced profile, combining good potency and in vivo pharmacokinetic properties. Final refinement by addressing cardiovascular safety liabilities led to the discovery of compound 48. Besides good potency, selectivity, and ADME properties, compound 48 displayed robust in vivo activity in a sleep-wake electroencephalogram (sw-EEG) assay consistent with mGlu2 receptor activation, in accordance with previous work from our laboratories.
Collapse
Affiliation(s)
- Jose María Cid
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S.A., Jarama 75, 45007-Toledo, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pacey LKK, Tharmalingam S, Hampson DR. Subchronic administration and combination metabotropic glutamate and GABAB receptor drug therapy in fragile X syndrome. J Pharmacol Exp Ther 2011; 338:897-905. [PMID: 21636656 PMCID: PMC3164344 DOI: 10.1124/jpet.111.183327] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 05/31/2011] [Indexed: 12/31/2022] Open
Abstract
The most common cause of inherited mental retardation, fragile X syndrome, results from a triplet repeat expansion in the FMR1 gene and loss of the mRNA binding protein, fragile X mental retardation protein (FMRP). In the absence of FMRP, signaling through group I metabotropic glutamate receptors (mGluRs) is enhanced. We previously proposed a mechanism whereby the audiogenic seizures exhibited by FMR1 null mice result from an imbalance in excitatory mGluR and inhibitory GABA(B) receptor (GABA(B)R) signaling (Mol Pharmacol 76:18-24, 2009). Here, we tested the mGluR5-positive allosteric modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB), the mGluR5 inverse agonist 2-methyl-6-(phenylethynyl)pyridine (MPEP), and GABA(B) receptor agonists, alone and in combination on receptor protein expression and audiogenic seizures in FMR1 mice. Single doses of MPEP (30 mg/kg), the GABA(B)R orthosteric agonist R-baclofen (1 mg/kg), or the GABA(B)R-positive allosteric modulator N,N'-dicyclopentyl-2-(methylthio)-5-nitro-4,6-pyrimidine diamine (GS-39783) (30 mg/kg), reduced the incidence of seizures. However, when administered subchronically (daily injections for 6 days), MPEP retained its anticonvulsant activity, whereas R-baclofen and GS-39783 did not. When administered at lower doses that had no effect when given alone, a single injection of MPEP plus R-baclofen also reduced seizures, but the effect was lost after subchronic administration. We were surprised to find that subchronic treatment with R-baclofen also induced tolerance to a single high dose of MPEP. These data demonstrate that tolerance develops rapidly to the antiseizure properties of R-baclofen alone and R-baclofen coadministered with MPEP, but not with MPEP alone. Our findings suggest that cross-talk between the G-protein signaling pathways of these receptors affects drug efficacy after repeated treatment.
Collapse
Affiliation(s)
- Laura K K Pacey
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
42
|
Zhang L, Brodney MA, Candler J, Doran AC, Duplantier AJ, Efremov IV, Evrard E, Kraus K, Ganong AH, Haas JA, Hanks AN, Jenza K, Lazzaro JT, Maklad N, McCarthy SA, Qian W, Rogers BN, Rottas MD, Schmidt CJ, Siuciak JA, Tingley FD, Zhang AQ. 1-[(1-Methyl-1H-imidazol-2-yl)methyl]-4-phenylpiperidines as mGluR2 Positive Allosteric Modulators for the Treatment of Psychosis. J Med Chem 2011; 54:1724-39. [DOI: 10.1021/jm101414h] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Lei Zhang
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Michael A. Brodney
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - John Candler
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Angela C. Doran
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Allen J. Duplantier
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ivan V. Efremov
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Edel Evrard
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kenneth Kraus
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Alan H. Ganong
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jessica A. Haas
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ashley N. Hanks
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Keith Jenza
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - John T. Lazzaro
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Noha Maklad
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sheryl A. McCarthy
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Weimin Qian
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Bruce N. Rogers
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Melinda D. Rottas
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Christopher J. Schmidt
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Judith A. Siuciak
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - F. David Tingley
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andy Q. Zhang
- Pfizer Global Research and Development, Groton Laboratories, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
43
|
Urwyler S. Allosteric modulation of family C G-protein-coupled receptors: from molecular insights to therapeutic perspectives. Pharmacol Rev 2011; 63:59-126. [PMID: 21228259 DOI: 10.1124/pr.109.002501] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Allosteric receptor modulation is an attractive concept in drug targeting because it offers important potential advantages over conventional orthosteric agonism or antagonism. Allosteric ligands modulate receptor function by binding to a site distinct from the recognition site for the endogenous agonist. They often have no effect on their own and therefore act only in conjunction with physiological receptor activation. This article reviews the current status of allosteric modulation at family C G-protein coupled receptors in the light of their specific structural features on the one hand and current concepts in receptor theory on the other hand. Family C G-protein-coupled receptors are characterized by a large extracellular domain containing the orthosteric agonist binding site known as the "venus flytrap module" because of its bilobal structure and the dynamics of its activation mechanism. Mutational analysis and chimeric constructs have revealed that allosteric modulators of the calcium-sensing, metabotropic glutamate and GABA(B) receptors bind to the seven transmembrane domain, through which they modify signal transduction after receptor activation. This is in contrast to taste-enhancing molecules, which bind to different parts of sweet and umami receptors. The complexity of interactions between orthosteric and allosteric ligands is revealed by a number of adequate biochemical and electrophysiological assay systems. Many allosteric family C GPCR modulators show in vivo efficacy in behavioral models for a variety of clinical indications. The positive allosteric calcium sensing receptor modulator cinacalcet is the first drug of this type to enter the market and therefore provides proof of principle in humans.
Collapse
Affiliation(s)
- Stephan Urwyler
- Department of Chemistry and Biochemistry, University of Berne, P/A Weissensteinweg 3, CH-3303 Jegenstorf, Berne, Switzerland.
| |
Collapse
|
44
|
Dhanya RP, Sidique S, Sheffler DJ, Nickols HH, Herath A, Yang L, Dahl R, Ardecky R, Semenova S, Markou A, Conn PJ, Cosford NDP. Design and synthesis of an orally active metabotropic glutamate receptor subtype-2 (mGluR2) positive allosteric modulator (PAM) that decreases cocaine self-administration in rats. J Med Chem 2011; 54:342-53. [PMID: 21155570 PMCID: PMC3071440 DOI: 10.1021/jm1012165] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The modification of 3'-((2-cyclopentyl-6,7-dimethyl-1-oxo-2,3-dihydro-1H-inden-5-yloxy)methyl)biphenyl-4-carboxylic acid (BINA, 1) by incorporating heteroatoms into the structure and replacing the cyclopentyl moiety led to the development of new mGluR2 positive allosteric modulators (PAMs) with optimized potency and superior druglike properties. These analogues are more potent than 1 in vitro and are highly selective for mGluR2 vs other mGluR subtypes. They have significantly improved pharmacokinetic (PK) properties, with excellent oral bioavailability and brain penetration. The benzisothiazol-3-one derivative 14 decreased cocaine self-administration in rats, providing proof-of-concept for the use of mGluR2 PAMs for the treatment of cocaine dependence.
Collapse
Affiliation(s)
- Raveendra-Panickar Dhanya
- Apoptosis and Cell Death Research Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037
| | - Shyama Sidique
- Apoptosis and Cell Death Research Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037
| | - Douglas J. Sheffler
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232
| | | | - Ananda Herath
- Apoptosis and Cell Death Research Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037
| | - Li Yang
- Apoptosis and Cell Death Research Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037
| | - Russell Dahl
- Apoptosis and Cell Death Research Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037
| | - Robert Ardecky
- Apoptosis and Cell Death Research Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037
| | - Svetlana Semenova
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Athina Markou
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA 92093
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Nicholas D. P. Cosford
- Apoptosis and Cell Death Research Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037
| |
Collapse
|
45
|
|
46
|
Froestl W. Chemistry and Pharmacology of GABAB Receptor Ligands. GABABRECEPTOR PHARMACOLOGY - A TRIBUTE TO NORMAN BOWERY 2010; 58:19-62. [DOI: 10.1016/s1054-3589(10)58002-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
47
|
Scaffold hopping from pyridones to imidazo[1,2-a]pyridines. New positive allosteric modulators of metabotropic glutamate 2 receptor. Bioorg Med Chem Lett 2009; 20:175-9. [PMID: 19932615 DOI: 10.1016/j.bmcl.2009.11.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 11/03/2009] [Accepted: 11/03/2009] [Indexed: 11/21/2022]
Abstract
Imidazo[1,2-a]pyridines were identified via their shape and electrostatic similarity as novel positive allosteric modulators of the metabotropic glutamate 2 receptor. The subsequent synthesis and SAR are described. Potent, selective and metabolically stable compounds were found representing a promising avenue for current further studies.
Collapse
|
48
|
Duplantier AJ, Efremov I, Candler J, Doran AC, Ganong AH, Haas JA, Hanks AN, Kraus KG, Lazzaro JT, Lu J, Maklad N, McCarthy SA, O'Sullivan TJ, Rogers BN, Siuciak JA, Spracklin DK, Zhang L. 3-Benzyl-1,3-oxazolidin-2-ones as mGluR2 positive allosteric modulators: Hit-to lead and lead optimization. Bioorg Med Chem Lett 2009; 19:2524-9. [PMID: 19328692 DOI: 10.1016/j.bmcl.2009.03.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Revised: 03/09/2009] [Accepted: 03/10/2009] [Indexed: 11/15/2022]
Abstract
The discovery, synthesis and SAR of a novel series of 3-benzyl-1,3-oxazolidin-2-ones as positive allosteric modulators (PAMs) of mGluR2 is described. Expedient hit-to-lead work on a single HTS hit led to the identification of a ligand-efficient and structurally attractive series of mGluR2 PAMs. Human microsomal clearance and suboptimal physicochemical properties of the initial lead were improved to give potent, metabolically stable and orally available mGluR2 PAMs.
Collapse
Affiliation(s)
- Allen J Duplantier
- CNS Chemistry, Pfizer Global Research and Development, Eastern Point Road, Groton, CT 06340, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gjoni T, Urwyler S. Changes in the properties of allosteric and orthosteric GABAB receptor ligands after a continuous, desensitizing agonist pretreatment. Eur J Pharmacol 2008; 603:37-41. [PMID: 19109945 DOI: 10.1016/j.ejphar.2008.12.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 11/24/2008] [Accepted: 12/09/2008] [Indexed: 11/27/2022]
Abstract
It has been estimated that only 15% of the compounds classified as silent G protein-coupled receptor antagonists are indeed devoid of either positive or negative intrinsic efficacy. Considering that 40% of all drugs on the market target G protein-coupled receptors mainly as orthosteric ligands, elucidating their intrinsic properties is becoming increasingly important. While agonism can be demonstrated using appropriately sensitive experimental setups, the detection of inverse agonism can be limited by a low degree of constitutive activity in many assay systems. In this study, changes in ligand behavior upon a lasting pretreatment with gamma-aminobutyric acid (GABA), that induced receptor desensitization, were observed, measuring the second messenger cyclic AMP (cAMP) in a GABA(B) receptor-expressing recombinant cell line. The GABA(B) receptor partial agonist 2-OH-saclofen lost its ability to inhibit 7beta-forskolin-induced cAMP production upon GABA-pretreatment. The "silent" receptor antagonists CGP62349, CGP52432, CGP56999 and SCH50911, on the other hand, stimulated 7beta-forskolin-induced cAMP production under these conditions. The inverse agonism of CGP56999 was inhibited by the efficacy-deficient 2-OH-saclofen, proving it was truly mediated through the orthosteric site of the GABA(B) receptor. Finally, the positive allosteric modulator GS39783, which previously only marginally inhibited cAMP production, suppressed it by 60% both alone and in the presence of the competitive receptor antagonist 2-OH-saclofen, thus GS39783 became an allosteric receptor agonist at desensitized GABA(B) receptors. These changes likely reflect adaptations in the mechanisms of GABA(B) receptor function following desensitization and may be important in the elucidation of intrinsic ligand efficacies as well as for the consequences of continuous drug treatment.
Collapse
Affiliation(s)
- Tina Gjoni
- Novartis Institutes for BioMedical Research, Neuroscience, Basel, Switzerland
| | | |
Collapse
|