1
|
Wang J, Wu M, Chen Z, Wu L, Wang T, Cao D, Wang H, Liu S, Xu Y, Li F, Liu J, Chen N, Zhao S, Cheng J, Wang S, Hua T. The unconventional activation of the muscarinic acetylcholine receptor M4R by diverse ligands. Nat Commun 2022; 13:2855. [PMID: 35606397 PMCID: PMC9126879 DOI: 10.1038/s41467-022-30595-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 05/04/2022] [Indexed: 01/22/2023] Open
Abstract
Muscarinic acetylcholine receptors (mAChRs) respond to the neurotransmitter acetylcholine and play important roles in human nervous system. Muscarinic receptor 4 (M4R) is a promising drug target for treating neurological and mental disorders, such as Alzheimer's disease and schizophrenia. However, the lack of understanding on M4R's activation by subtype selective agonists hinders its therapeutic applications. Here, we report the structural characterization of M4R selective allosteric agonist, compound-110, as well as agonist iperoxo and positive allosteric modulator LY2119620. Our cryo-electron microscopy structures of compound-110, iperoxo or iperoxo-LY2119620 bound M4R-Gi complex reveal their different interaction modes and activation mechanisms of M4R, and the M4R-ip-LY-Gi structure validates the cooperativity between iperoxo and LY2119620 on M4R. Through the comparative structural and pharmacological analysis, compound-110 mostly occupies the allosteric binding pocket with vertical binding pose. Such a binding and activation mode facilitates its allostersic selectivity and agonist profile. In addition, in our schizophrenia-mimic mouse model study, compound-110 shows antipsychotic activity with low extrapyramidal side effects. Thus, this study provides structural insights to develop next-generation antipsychotic drugs selectively targeting on mAChRs subtypes.
Collapse
Affiliation(s)
- Jingjing Wang
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Meng Wu
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Zhangcheng Chen
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Lijie Wu
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Tian Wang
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Dongmei Cao
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Huan Wang
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Shenhui Liu
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Yueming Xu
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Fei Li
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Junlin Liu
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Na Chen
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.
| | - Sheng Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| |
Collapse
|
2
|
Son JH, Phuan PW, Zhu JS, Lipman E, Cheung A, Tsui KY, Tantillo DJ, Verkman AS, Haggie PM, Kurth MJ. 1-BENZYLSPIRO[PIPERIDINE-4,1'-PYRIDO[3,4-b]indole] 'co-potentiators' for minimal function CFTR mutants. Eur J Med Chem 2021; 209:112888. [PMID: 33092904 PMCID: PMC7744356 DOI: 10.1016/j.ejmech.2020.112888] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/19/2020] [Accepted: 09/23/2020] [Indexed: 11/25/2022]
Abstract
We previously identified a spiro [piperidine-4,1-pyrido [3,4-b]indole] class of co-potentiators that function in synergy with existing CFTR potentiators such as VX-770 or GLGP1837 to restore channel activity of a defined subset of minimal function cystic fibrosis transmembrane conductance regulator (CFTR) mutants. Here, structure-activity studies were conducted to improve their potency over the previously identified compound, 20 (originally termed CP-A01). Targeted synthesis of 37 spiro [piperidine-4,1-pyrido [3,4-b]indoles] was generally accomplished using versatile two or three step reaction protocols with each step having high efficiency. Structure-activity relationship studies established that analog 2i, with 6'-methoxyindole and 2,4,5-trifluorobenzyl substituents, had the greatest potency for activation of N1303K-CFTR, with EC50 ∼600 nM representing an ∼17-fold improvement over the original compound identified in a small molecule screen.
Collapse
Affiliation(s)
- Jung-Ho Son
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Puay-Wah Phuan
- Departments of Medicine & Physiology, University of California San Francisco, CA 94143, USA
| | - Jie S Zhu
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Elena Lipman
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Amy Cheung
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Ka Yi Tsui
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Dean J Tantillo
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Alan S Verkman
- Departments of Medicine & Physiology, University of California San Francisco, CA 94143, USA
| | - Peter M Haggie
- Departments of Medicine & Physiology, University of California San Francisco, CA 94143, USA.
| | - Mark J Kurth
- Department of Chemistry, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
3
|
The operational model of allosteric modulation of pharmacological agonism. Sci Rep 2020; 10:14421. [PMID: 32879329 PMCID: PMC7468285 DOI: 10.1038/s41598-020-71228-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022] Open
Abstract
Proper determination of agonist efficacy is indispensable in the evaluation of agonist selectivity and bias to activation of specific signalling pathways. The operational model (OM) of pharmacological agonism is a useful means for achieving this goal. Allosteric ligands bind to receptors at sites that are distinct from those of endogenous agonists that interact with the orthosteric domain on the receptor. An allosteric modulator and an orthosteric agonist bind simultaneously to the receptor to form a ternary complex, where the allosteric modulator affects the binding affinity and operational efficacy of the agonist. Allosteric modulators are an intensively studied group of receptor ligands because of their selectivity and preservation of physiological space-time pattern of the signals they modulate. We analysed the operational model of allosterically-modulated agonism (OMAM) including modulation by allosteric agonists. Similar to OM, several parameters of OMAM are inter-dependent. We derived equations describing mutual relationships among parameters of the functional response and OMAM. We present a workflow for the robust fitting of OMAM to experimental data using derived equations.
Collapse
|
4
|
She X, Pegoli A, Gruber CG, Wifling D, Carpenter J, Hübner H, Chen M, Wan J, Bernhardt G, Gmeiner P, Holliday ND, Keller M. Red-Emitting Dibenzodiazepinone Derivatives as Fluorescent Dualsteric Probes for the Muscarinic Acetylcholine M 2 Receptor. J Med Chem 2020; 63:4133-4154. [PMID: 32233403 DOI: 10.1021/acs.jmedchem.9b02172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Fluorescently labeled dibenzodiazepinone-type muscarinic acetylcholine receptor (MR) antagonists, including dimeric ligands, were prepared using red-emitting cyanine dyes. Probes containing a fluorophore with negative charge showed high M2R affinities (pKi (radioligand competition binding): 9.10-9.59). Binding studies at M1 and M3-M5 receptors indicated a M2R preference. Flow cytometric and high-content imaging saturation and competition binding (M1R, M2R, and M4R) confirmed occupation of the orthosteric site. Confocal microscopy revealed that fluorescence was located mainly at the cell membrane (CHO-hM2R cells). Results from dissociation and saturation binding experiments (M2R) in the presence of allosteric M2R modulators (dissociation: W84, LY2119620, and alcuronium; saturation binding: W84) were consistent with a competitive mode of action between the fluorescent probes and the allosteric ligands. Taken together, these lines of evidence indicate that these ligands are useful fluorescent molecular tools to label the M2R in imaging and binding studies and suggest that they have a dualsteric mode of action.
Collapse
Affiliation(s)
- Xueke She
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Andrea Pegoli
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Corinna G Gruber
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - David Wifling
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Jessica Carpenter
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, U.K
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Mengya Chen
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Jianfei Wan
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Nicholas D Holliday
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Derby Road, Nottingham NG7 2UH, U.K
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| |
Collapse
|
5
|
Wang Y, Zhao S, Xue Q, Chen X. Enantioselective benzylation of methyl 4-oxo-3-piperidinecarboxylate with cinchona alkaloids phase-transfer catalysts. SYNTHETIC COMMUN 2018. [DOI: 10.1080/00397911.2018.1496259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Yaomin Wang
- School of Pharmaceutical and Life Sciences, Changzhou University, Changzhou, Jiangsu, P. R. China
| | - Shuai Zhao
- School of Pharmaceutical and Life Sciences, Changzhou University, Changzhou, Jiangsu, P. R. China
| | - Qiaoyun Xue
- School of Pharmaceutical and Life Sciences, Changzhou University, Changzhou, Jiangsu, P. R. China
| | - Xin Chen
- School of Pharmaceutical and Life Sciences, Changzhou University, Changzhou, Jiangsu, P. R. China
| |
Collapse
|
6
|
She X, Pegoli A, Mayr J, Hübner H, Bernhardt G, Gmeiner P, Keller M. Heterodimerization of Dibenzodiazepinone-Type Muscarinic Acetylcholine Receptor Ligands Leads to Increased M 2R Affinity and Selectivity. ACS OMEGA 2017; 2:6741-6754. [PMID: 30023530 PMCID: PMC6044897 DOI: 10.1021/acsomega.7b01085] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/05/2017] [Indexed: 05/13/2023]
Abstract
In search for selective ligands for the muscarinic acetylcholine receptor (MR) subtype M2, the dimeric ligand approach, that is combining two pharmacophores in one and the same molecule, was pursued. Different types (agonists, antagonists, orthosteric, and allosteric) of monomeric MR ligands were combined by various linkers with a dibenzodiazepinone-type MR antagonist, affording five types of heterodimeric compounds ("DIBA-xanomeline," "DIBA-TBPB," "DIBA-77-LH-28-1," "DIBA-propantheline," and "DIBA-4-DAMP"), which showed high M2R affinities (pKi > 8.3). The heterodimeric ligand UR-SK75 (46) exhibited the highest M2R affinity and selectivity [pKi (M1R-M5R): 8.84, 10.14, 7.88, 8.59, and 7.47]. Two tritium-labeled dimeric derivatives ("DIBA-xanomeline"-type: [3H]UR-SK71 ([3H]44) and "DIBA-TBPB"-type: [3H]UR-SK59 ([3H]64)) were prepared to investigate their binding modes at hM2R. Saturation-binding experiments showed that these compounds address the orthosteric binding site of the M2R. The investigation of the effect of various allosteric MR modulators [gallamine (13), W84 (14), and LY2119620 (15)] on the equilibrium (13-15) or saturation (14) binding of [3H]64 suggested a competitive mechanism between [3H]64 and the investigated allosteric ligands, and consequently a dualsteric binding mode of 64 at the M2R.
Collapse
Affiliation(s)
- Xueke She
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Andrea Pegoli
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Judith Mayr
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Harald Hübner
- Department
of Chemistry and Pharmacy, Emil Fischer Center, Friedrich Alexander University, Schuhstr. 19, D-91052 Erlangen, Germany
| | - Günther Bernhardt
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Peter Gmeiner
- Department
of Chemistry and Pharmacy, Emil Fischer Center, Friedrich Alexander University, Schuhstr. 19, D-91052 Erlangen, Germany
| | - Max Keller
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| |
Collapse
|
7
|
Chintamaneni PK, Krishnamurthy PT, Rao PV, Pindiprolu SS. Surface modified nano-lipid drug conjugates of positive allosteric modulators of M1 muscarinic acetylcholine receptor for the treatment of Alzheimer’s disease. Med Hypotheses 2017; 101:17-22. [DOI: 10.1016/j.mehy.2017.01.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 10/31/2016] [Accepted: 01/02/2017] [Indexed: 12/24/2022]
|
8
|
Liu B, Song RJ, Ouyang XH, Li Y, Hu M, Li JH. Palladium-catalyzed oxidative 6-exo-trig cyclization of 1,6-enynes: facile synthesis of bicyclo[4.1.0]heptan-5-ones. Chem Commun (Camb) 2015; 51:12819-22. [DOI: 10.1039/c5cc04300j] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We here describe a new palladium-catalyzed oxidative 6-exo-trig cyclization of 1,6-enynes at room temperature using tBuONO as an oxidant for the synthesis of 3-bicyclo[4.1.0]heptan-5-ones.
Collapse
Affiliation(s)
- Bang Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
- China
| | - Ren-Jie Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
- China
| | - Xuan-Hui Ouyang
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
- China
| | - Yang Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
- China
| | - Ming Hu
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
- China
| | - Jin-Heng Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
- China
| |
Collapse
|
9
|
Keov P, López L, Devine SM, Valant C, Lane JR, Scammells PJ, Sexton PM, Christopoulos A. Molecular mechanisms of bitopic ligand engagement with the M1 muscarinic acetylcholine receptor. J Biol Chem 2014; 289:23817-37. [PMID: 25006252 DOI: 10.1074/jbc.m114.582874] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
TBPB and 77-LH-28-1 are selective agonists of the M1 muscarinic acetylcholine receptor (mAChR) that may gain their selectivity through a bitopic mechanism, interacting concomitantly with the orthosteric site and part of an allosteric site. The current study combined site-directed mutagenesis, analytical pharmacology,and molecular modeling to gain further insights into the structural basis underlying binding and signaling by these agonists. Mutations within the orthosteric binding site caused similar reductions in affinity and signaling efficacy for both selective and prototypical orthosteric ligands. In contrast, the mutation of residues within transmembrane helix (TM) 2 and the second extracellular loop (ECL2) discriminated between the different classes of ligand. In particular, ECL2 appears to be involved in the selective binding of bitopic ligands and in coordinating biased agonism between intracellular calcium mobilization and ERK1/2 phosphorylation. Molecular modeling of the interaction between TBPB and the M1 mAChR revealed a binding pose predicted to extend from the orthosteric site up toward a putative allosteric site bordered by TM2, TM3, and TM7, thus consistent with a bitopic mode of binding. Overall, these findings provide valuable structural and mechanistic insights into bitopic ligand actions and receptor activation and support a role for ECL2 in dictating the active states that can be adopted by a G protein-coupled receptor. This may enable greater selective ligand design and development for mAChRs and facilitate improved identification of bitopic ligands.
Collapse
Affiliation(s)
- Peter Keov
- From the Drug Discovery Biology Theme and Department of Pharmacology and
| | - Laura López
- From the Drug Discovery Biology Theme and Department of Pharmacology and
| | - Shane M Devine
- the Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Celine Valant
- From the Drug Discovery Biology Theme and Department of Pharmacology and
| | - J Robert Lane
- From the Drug Discovery Biology Theme and Department of Pharmacology and
| | - Peter J Scammells
- the Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Patrick M Sexton
- From the Drug Discovery Biology Theme and Department of Pharmacology and
| | | |
Collapse
|
10
|
Melancon BJ, Tarr JC, Panarese JD, Wood MR, Lindsley CW. Allosteric modulation of the M1 muscarinic acetylcholine receptor: improving cognition and a potential treatment for schizophrenia and Alzheimer's disease. Drug Discov Today 2013; 18:1185-99. [PMID: 24051397 PMCID: PMC3876030 DOI: 10.1016/j.drudis.2013.09.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/02/2013] [Accepted: 09/09/2013] [Indexed: 12/13/2022]
Abstract
Allosteric modulation of AMPA, NR2B, mGlu2, mGlu5 and M1, targeting glutamatergic dysfunction, represents a significant area of research for the treatment of schizophrenia. Of these targets, clinical promise has been demonstrated using muscarinic activators for the treatment of Alzheimer's disease (AD) and schizophrenia. These diseases have inspired researchers to determine the effects of modulating cholinergic transmission in the forebrain, which is primarily regulated by one of five subtypes of muscarinic acetylcholine receptor (mAChR), a subfamily of G-protein-coupled receptors (GPCRs). Of these five subtypes, M1 is highly expressed in brain regions responsible for learning, cognition and memory. Xanomeline, an orthosteric muscarinic agonist with modest selectivity, was one of the first compounds that displayed improvements in behavioral disturbances in AD patients and efficacy in schizophrenics. Since these initial clinical results, many scientists, including those in our laboratories, have strived to elucidate the role of M1 with compounds that display improved selectivity for this receptor by targeting allosteric modes of receptor activation. A survey of selected compounds in this area will be presented.
Collapse
Affiliation(s)
- Bruce J Melancon
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center, 1205 Light Hall, Nashville, TN 37232-6600, USA
| | | | | | | | | |
Collapse
|
11
|
Keov P, Valant C, Devine SM, Lane JR, Scammells PJ, Sexton PM, Christopoulos A. Reverse engineering of the selective agonist TBPB unveils both orthosteric and allosteric modes of action at the M₁ muscarinic acetylcholine receptor. Mol Pharmacol 2013; 84:425-37. [PMID: 23798605 DOI: 10.1124/mol.113.087320] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Recent interest in the M₁ muscarinic acetylcholine (ACh) receptor (mAChR) has led to the discovery of various selective agonists for the receptor. The novel selective agonist 1-(1'-(2-methylbenzyl)-1,4'-bipiperidin-4-yl)-1H-benzo[d]imidazol-2(3H)-1 (TBPB) displays unprecedented functional selectivity at the M₁ mAChR. This functional selectivity has been described to stem from sole interaction with an allosteric site, although the evidence for such a mechanism is equivocal. To delineate TBPB's mechanism of action, several truncated variants of TBPB were synthesized and characterized. Binding experiments with [³H]N-methylscopolamine at the M₁, M₂, M₃, and M₄ mAChRs revealed radioligand displacement in a manner consistent with a competitive binding mode at the orthosteric site by TBPB and fragment derivatives. Cell-based functional assays of fragment derivatives of TBPB identified both agonistic and antagonistic moieties, one of which, 1-(1-cyclohexylpiperidin-4-yl)-1H-benzo[d]imidazol-2(3H)-1 (VCP794), lost agonistic selectivity for the M₁ mAChR. Further interaction experiments between TBPB or its antagonist fragments with ACh also indicated a mechanism consistent with competitive binding at mAChRs. However, interaction with an allosteric site by an antagonist fragment of TBPB was demonstrated via its ability to retard radioligand dissociation. To reconcile this dual orthosteric/allosteric pharmacological behavior, we propose that TBPB is a bitopic ligand, interacting with both the orthosteric site and an allosteric site, at the M₁ mAChR. This mechanism may also be the case for other selective agonists for mAChRs, and should be taken into consideration in the profiling and classification of new novel selective agonists for this receptor family.
Collapse
Affiliation(s)
- Peter Keov
- Drug Discovery Biology & Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
12
|
Davie BJ, Christopoulos A, Scammells PJ. Development of M1 mAChR allosteric and bitopic ligands: prospective therapeutics for the treatment of cognitive deficits. ACS Chem Neurosci 2013; 4:1026-48. [PMID: 23659787 PMCID: PMC3715844 DOI: 10.1021/cn400086m] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 05/09/2013] [Indexed: 12/22/2022] Open
Abstract
Since the cholinergic hypothesis of memory dysfunction was first reported, extensive research efforts have focused on elucidating the mechanisms by which this intricate system contributes to the regulation of processes such as learning, memory, and higher executive function. Several cholinergic therapeutic targets for the treatment of cognitive deficits, psychotic symptoms, and the underlying pathophysiology of neurodegenerative disorders, such as Alzheimer's disease and schizophrenia, have since emerged. Clinically approved drugs now exist for some of these targets; however, they all may be considered suboptimal therapeutics in that they produce undesirable off-target activity leading to side effects, fail to address the wide variety of symptoms and underlying pathophysiology that characterize these disorders, and/or afford little to no therapeutic effect in subsets of patient populations. A promising target for which there are presently no approved therapies is the M1 muscarinic acetylcholine receptor (M1 mAChR). Despite avid investigation, development of agents that selectively activate this receptor via the orthosteric site has been hampered by the high sequence homology of the binding site between the five muscarinic receptor subtypes and the wide distribution of this receptor family in both the central nervous system (CNS) and the periphery. Hence, a plethora of ligands targeting less structurally conserved allosteric sites of the M1 mAChR have been investigated. This Review aims to explain the rationale behind allosterically targeting the M1 mAChR, comprehensively summarize and critically evaluate the M1 mAChR allosteric ligand literature to date, highlight the challenges inherent in allosteric ligand investigation that are impeding their clinical advancement, and discuss potential methods for resolving these issues.
Collapse
Affiliation(s)
- Briana J. Davie
- Medicinal
Chemistry and Drug Discovery Biology, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal
Parade, Parkville VIC 3052, Australia
| | - Arthur Christopoulos
- Medicinal
Chemistry and Drug Discovery Biology, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal
Parade, Parkville VIC 3052, Australia
| | - Peter J. Scammells
- Medicinal
Chemistry and Drug Discovery Biology, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal
Parade, Parkville VIC 3052, Australia
| |
Collapse
|
13
|
Sheffler DJ, Sevel C, Le U, Lovell KM, Tarr JC, Carrington SJS, Cho HP, Digby GJ, Niswender CM, Conn PJ, Hopkins CR, Wood MR, Lindsley CW. Further exploration of M₁ allosteric agonists: subtle structural changes abolish M₁ allosteric agonism and result in pan-mAChR orthosteric antagonism. Bioorg Med Chem Lett 2013; 23:223-7. [PMID: 23200253 PMCID: PMC3525729 DOI: 10.1016/j.bmcl.2012.10.132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/18/2012] [Accepted: 10/29/2012] [Indexed: 01/22/2023]
Abstract
This letter describes the further exploration of two series of M(1) allosteric agonists, TBPB and VU0357017, previously reported from our lab. Within the TPBP scaffold, either electronic or steric perturbations to the central piperidine ring led to a loss of selective M(1) allosteric agonism and afforded pan-mAChR antagonism, which was demonstrated to be mediated via the orthosteric site. Additional SAR around a related M(1) allosteric agonist family (VU0357017) identified similar, subtle 'molecular switches' that modulated modes of pharmacology from allosteric agonism to pan-mAChR orthosteric antagonism. Therefore, all of these ligands are best classified as bi-topic ligands that possess high affinity binding at an allosteric site to engender selective M(1) activation, but all bind, at higher concentrations, to the orthosteric ACh site, leading to non-selective orthosteric site binding and mAChR antagonism.
Collapse
Affiliation(s)
- Douglas J. Sheffler
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
| | - Christian Sevel
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Uyen Le
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
| | - Kimberly M. Lovell
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
| | - James C. Tarr
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
| | - Sheridan J. S. Carrington
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hyekyung P. Cho
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
| | - Gregory J. Digby
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Colleen M. Niswender
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
| | - Corey R. Hopkins
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Michael R. Wood
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W. Lindsley
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
14
|
Abstract
INTRODUCTION There is substantial evidence from preclinical and early proof-of-concept studies suggesting that selective modulation of the M(1) muscarinic receptor is efficacious in cognitive models of Alzheimer's disease (AD) and antipsychotic models of schizophrenia. For example, a number of nonselective M(1) muscarinic agonists have previously shown positive effects on cognitive function in AD patients, but were limited due to cholinergic adverse events thought to be mediated by pan activation of the M(2) to M(5) subtypes. Thus, there is a need to identify selective activators of the M(1) receptor to evaluate their potential in cognitive disorders. One strategy to confer selectivity for M(1) is the identification of allosteric agonists or positive allosteric modulators, which would target an allosteric site on the M(1) receptor rather than the highly conserved orthosteric acetylcholine binding site. AREAS COVERED This review discusses the M(1) muscarinic receptor and its potential therapeutic value in the treatment of CNS disorders such as AD and schizophrenia. Specifically, novel allosteric ligands that activate or positively modulate the M(1) receptor are examined and peer-reviewed articles associated with these patents publications are also described. EXPERT OPINION There is substantial evidence supporting activation of the M(1) receptor might be effective in treating symptoms of AD and schizophrenia, but therapeutic success has been elusive and is hypothesized to be due to the lack of selectivity among orthosteric agonists. During the past decade, allosteric modulation of GPCRs has evolved as a viable strategy toward generating subtype selective molecules. A number of novel, selective ligands in the form of allosteric agonists and positive allosteric modulators of the M(1) receptor have been identified offering the potential for clinical evaluation of M(1)-specific receptor activation.
Collapse
Affiliation(s)
- Scott D Kuduk
- Department of Medicinal Chemistry, Merck Research Laboratories , Sumneytown Pike, PO Box 4, West Point, PA 19486, USA.
| | | |
Collapse
|
15
|
Kumar N, Khan SI, Rawat DS. Synthesis and Antimalarial-Activity Evaluation of TetraoxaneTriazine Hybrids and Spiro[piperidine-4,3′-tetraoxanes]. Helv Chim Acta 2012. [DOI: 10.1002/hlca.201200015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
16
|
Allosteric modulators of rhodopsin-like G protein-coupled receptors: opportunities in drug development. Pharmacol Ther 2012; 135:292-315. [PMID: 22728155 DOI: 10.1016/j.pharmthera.2012.06.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 06/07/2012] [Indexed: 11/21/2022]
Abstract
Rhodopsin-like (class A) G protein-coupled receptors (GPCRs) are one of the most important classes of drug targets. The discovery that these GPCRs can be allosterically modulated by small drug molecules has opened up new opportunities in drug development. It will allow the drugability of "difficult targets", such as GPCRs activated by large (glyco)proteins, or by very polar or highly lipophilic physiological agonists. Receptor subtype selectivity should be more easily achievable with allosteric than with orthosteric ligands. Allosteric modulation will allow a broad spectrum of pharmacological effects largely expanding that of orthosteric ligands. Furthermore, allosteric modulators may show an improved safety profile as compared to orthosteric ligands. Only recently, the explicit search for allosteric modulators has been started for only a few rhodopsin-like GPCRs. The first negative allosteric modulators (allosteric antagonists) of chemokine receptors, maraviroc (CCR5 receptor), used in HIV therapy, and plerixafor (CXCR4 receptor) for stem cell mobilization, have been approved as drugs. The development of allosteric modulators for rhodopsin-like GPCRs as novel drugs is still at an early stage; it appears highly promising.
Collapse
|
17
|
A quantitative structure-activity relationship study of tetrabutylphosphonium bromide analogs as muscarinic acetylcholine receptors. JOURNAL OF THE SERBIAN CHEMICAL SOCIETY 2011. [DOI: 10.2298/jsc101122102s] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Quantitative structure-activity relationship (QSAR) of tetrabutylphosphonium
bromide (TBPB) analogs as muscarinic acetylcholine receptors (mAChRs) was
studied. A suitable set of molecular descriptors was calculated and stepwise
multiple linear regression (SW-MLR) was employed to select those descriptors
that resulted in the best fitted models. A MLR model with three selected
descriptors was obtained. Furthermore, the MLR model was validated using the
leave-one-out (LOO) and leave-group-out (LGO) crossvalidation, and the
Y-randomization test. This model, with high statistical significance (R2
train = 0.982, F = 388.715, Q2 LOO = 0.973, Q2 LGO = 0.977 and R2 test =
0.986) could predict the activity of the molecules with a percentage
prediction error lower than 5 %.
Collapse
|
18
|
Sams AG, Hentzer M, Mikkelsen GK, Larsen K, Bundgaard C, Plath N, Christoffersen CT, Bang-Andersen B. Discovery of N-{1-[3-(3-oxo-2,3-dihydrobenzo[1,4]oxazin-4-yl)propyl]piperidin-4-yl}-2-phenylacetamide (Lu AE51090): an allosteric muscarinic M1 receptor agonist with unprecedented selectivity and procognitive potential. J Med Chem 2010; 53:6386-97. [PMID: 20684563 DOI: 10.1021/jm100697g] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The discovery and structure-activity relationship (SAR) of a series of allosteric muscarinic M(1) receptor agonists are described. Compound 17 (Lu AE51090) was identified as a representative compound from the series, based on its high selectivity as an agonist at the muscarinic M(1) receptor across a panel of muscarinic receptor subtypes. Furthermore, 17 displayed a high degree of selectivity when tested in a broad panel of G-protein-coupled receptors, ion channels, transporters, and enzymes, and 17 showed an acceptable pharmacokinetic profile and sufficient brain exposure in rodents in order to characterize the compound in vivo. Hence, in a rodent model of learning and memory, 17 reversed delay-induced natural forgetting, suggesting a procognitive potential of 17.
Collapse
Affiliation(s)
- Anette G Sams
- Medicinal Chemistry Research, Lundbeck Research Denmark, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Budzik B, Garzya V, Shi D, Walker G, Woolley-Roberts M, Pardoe J, Lucas A, Tehan B, Rivero RA, Langmead CJ, Watson J, Wu Z, Forbes IT, Jin J. Novel N-Substituted Benzimidazolones as Potent, Selective, CNS-Penetrant, and Orally Active M1 mAChR Agonists. ACS Med Chem Lett 2010; 1:244-8. [PMID: 24900202 DOI: 10.1021/ml100105x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Accepted: 06/02/2010] [Indexed: 12/16/2022] Open
Abstract
Virtual screening of the corporate compound collection yielded compound 1 as a subtype selective muscarinic M1 receptor agonist hit. Initial optimization of the N-capping group of the central piperidine ring resulted in compounds 2 and 3 with significantly improved potency and selectivity. Subsequent optimization of substituents on the phenyl ring of the benzimidazolone moiety led to the discovery of novel muscarinic M1 receptor agonists 4 and 5 with excellent potency, general and subtype selectivity, and pharmacokinetic (PK) properties including good central nervous system (CNS) penetration and oral bioavailability. Compound 5 showed robust in vivo activities in animal models of cognition enhancement. The combination of high potency, excellent selectivity, and good PK properties makes compounds 4 and 5 valuable tool compounds for investigating and validating potential therapeutic benefits resulting from selective M1 activation.
Collapse
Affiliation(s)
- Brian Budzik
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Vincenzo Garzya
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Dongchuan Shi
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Graham Walker
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | | | - Joanne Pardoe
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Adam Lucas
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Ben Tehan
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Ralph A. Rivero
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | | | - Jeannette Watson
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Zining Wu
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Ian T. Forbes
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| | - Jian Jin
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
| |
Collapse
|
20
|
The discovery of a series of N-substituted 3-(4-piperidinyl)-1,3-benzoxazolinones and oxindoles as highly brain penetrant, selective muscarinic M1 agonists. Bioorg Med Chem Lett 2010; 20:5434-8. [DOI: 10.1016/j.bmcl.2010.07.097] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 07/22/2010] [Accepted: 07/23/2010] [Indexed: 01/02/2023]
|
21
|
Davis AA, Heilman CJ, Brady AE, Miller NR, Fuerstenau-Sharp M, Hanson BJ, Lindsley CW, Conn PJ, Lah JJ, Levey AI. Differential effects of allosteric M(1) muscarinic acetylcholine receptor agonists on receptor activation, arrestin 3 recruitment, and receptor downregulation. ACS Chem Neurosci 2010; 1:542-551. [PMID: 20835371 DOI: 10.1021/cn100011e] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Muscarinic acetylcholine receptors (mAChRs) are drug targets for multiple neurodegenerative and neuropsychiatric disorders, but the full therapeutic potential of mAChR-targeted drugs has not been realized, mainly because of a lack of subtype-selective agonists. Recent advances have allowed the development of highly selective agonists that bind to an allosteric site on the M(1) mAChR that is spatially distinct from the orthosteric acetylcholine binding site, but less is known about the profile of intracellular signals activated by orthosteric versus allosteric M(1) mAChR agonists. We investigated the activation and regulatory mechanisms of two structurally distinct allosteric M(1) mAChR agonists, AC260584 and TBPB. We show that allosteric agonists potently activate multiple signal transduction pathways linked to the M(1) mAChR receptor but, compared to orthosteric agonists, much less efficiently recruit arrestin 3, a protein involved in regulation of G-protein coupled receptor signaling. Consistent with decreased arrestin recruitment, both allosteric agonists showed blunted responses in measurements of receptor desensitization, internalization, and downregulation. These results advance the understanding of mAChR biology and may shed light on unanticipated differences in the pharmacology of orthosteric vs. allosteric agonists that might be capitalized upon for drug development for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Albert A. Davis
- Center for Neurodegenerative Disease and Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Craig J. Heilman
- Center for Neurodegenerative Disease and Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Ashley E. Brady
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Nicole R. Miller
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | | | | | - Craig W. Lindsley
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Chemistry, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt Program in Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Chemistry, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - James J. Lah
- Center for Neurodegenerative Disease and Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Allan I. Levey
- Center for Neurodegenerative Disease and Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
22
|
Bridges TM, Kennedy JP, Hopkins CR, Conn PJ, Lindsley CW. Heterobiaryl and heterobiaryl ether derived M5 positive allosteric modulators. Bioorg Med Chem Lett 2010; 20:5617-22. [PMID: 20801651 DOI: 10.1016/j.bmcl.2010.08.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 08/05/2010] [Accepted: 08/09/2010] [Indexed: 10/19/2022]
Abstract
This Letter describes a chemical lead optimization campaign directed at VU0238429, the first M(5)-preferring positive allosteric modulator (PAM), discovered through analog work around VU0119498, a pan G(q) mAChR M(1), M(3), M(5) PAM. An iterative parallel synthesis approach was employed to incorporate basic heterocycles to improve physiochemical properties.
Collapse
Affiliation(s)
- Thomas M Bridges
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | |
Collapse
|
23
|
Moustafa MMAR, Pagenkopf BL. Synthesis of 5-Azaindoles via a Cycloaddition Reaction between Nitriles and Donor−Acceptor Cyclopropanes. Org Lett 2010; 12:3168-71. [DOI: 10.1021/ol101078z] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Brian L. Pagenkopf
- Department of Chemistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada
| |
Collapse
|
24
|
Budzik B, Garzya V, Shi D, Walker G, Lauchart Y, Lucas AJ, Rivero RA, Langmead CJ, Watson J, Wu Z, Forbes IT, Jin J. 2′ Biaryl amides as novel and subtype selective M1 agonists. Part II: Further optimization and profiling. Bioorg Med Chem Lett 2010; 20:3545-9. [DOI: 10.1016/j.bmcl.2010.04.127] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 04/26/2010] [Accepted: 04/27/2010] [Indexed: 11/28/2022]
|
25
|
Budzik B, Garzya V, Shi D, Foley JJ, Rivero RA, Langmead CJ, Watson J, Wu Z, Forbes IT, Jin J. 2′ Biaryl amides as novel and subtype selective M1 agonists. Part I: Identification, synthesis, and initial SAR. Bioorg Med Chem Lett 2010; 20:3540-4. [DOI: 10.1016/j.bmcl.2010.04.128] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 04/26/2010] [Accepted: 04/27/2010] [Indexed: 10/19/2022]
|
26
|
Bridges TM, LeBois EP, Hopkins CR, Wood MR, Jones CK, Conn PJ, Lindsley CW. The antipsychotic potential of muscarinic allosteric modulation. DRUG NEWS & PERSPECTIVES 2010; 23:229-40. [PMID: 20520852 PMCID: PMC4780339 DOI: 10.1358/dnp.2010.23.4.1416977] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The cholinergic hypothesis of schizophrenia emerged over 50 years ago based on clinical observations with both anticholinergics and pan-muscarinic agonists. Not until the 1990s did the cholinergic hypothesis of schizophrenia receive renewed enthusiasm based on clinical data with xanomeline, a muscarinic acetylcholine receptor M(1)/M(4)-preferring orthosteric agonist. In a clinical trial with Alzheimer's patients, xanomeline not only improved cognitive performance, but also reduced psychotic behaviors. This encouraging data spurred a second clinical trial in schizophrenic patients, wherein xanomeline significantly improved the positive, negative and cognitive symptom clusters. However, the question remained: Was the antipsychotic efficacy due to activation of M(1), M(4) or both M(1)/M(4)? Classical orthosteric ligands lacked the muscarinic receptor subtype selectivity required to address this key question. More recently, functional assays have allowed for the discovery of ligands that bind at allosteric sites, binding sites distinct from the orthosteric (acetylcholine) site, which are structurally less conserved and thereby afford high levels of receptor subtype selectivity. Recently, allosteric ligands, with unprecedented selectivity for either M(1) or M(4), have been discovered and have demonstrated comparable efficacy to xanomeline in preclinical antipsychotic and cognition models. These data suggest that selective allosteric activation of either M(1) or M(4) has antipsychotic potential through distinct, yet complimentary mechanisms.
Collapse
Affiliation(s)
- Thomas M. Bridges
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Evan P. LeBois
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Corey R. Hopkins
- Department of Pharmacology, Vanderbilt Program in Drug Discovery and Vanderbilt Specialized Chemistry Center (MLPCN), Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael R. Wood
- Department of Pharmacology, Vanderbilt Program in Drug Discovery and Vanderbilt Specialized Chemistry Center (MLPCN), Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carrie K. Jones
- Department of Pharmacology, Vanderbilt Program in Drug Discovery and Vanderbilt Specialized Chemistry Center (MLPCN), Vanderbilt University Medical Center, and U.S. Department of Veterans Affairs, Tennessee Valley Healthcare System (TVHS), Nashville, Tennessee, USA
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt Program in Drug Discovery and Vanderbilt Specialized Chemistry Center (MLPCN), Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig W. Lindsley
- Department of Pharmacology, Vanderbilt Program in Drug Discovery and Vanderbilt Specialized Chemistry Center (MLPCN), Vanderbilt University Medical Center, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
27
|
Bridges TM, Phillip Kennedy J, Noetzel MJ, Breininger ML, Gentry PR, Conn PJ, Lindsley CW. Chemical lead optimization of a pan Gq mAChR M1, M3, M5 positive allosteric modulator (PAM) lead. Part II: development of a potent and highly selective M1 PAM. Bioorg Med Chem Lett 2010; 20:1972-5. [PMID: 20156687 PMCID: PMC2834874 DOI: 10.1016/j.bmcl.2010.01.109] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 01/15/2010] [Accepted: 01/20/2010] [Indexed: 01/24/2023]
Abstract
This Letter describes a chemical lead optimization campaign directed at VU0119498, a pan G(q) mAChR M(1), M(3), M(5) positive allosteric modulator (PAM) with the goal of developing a selective M(1) PAM. An iterative library synthesis approach delivered a potent (M(1) EC(50)=830 nM) and highly selective M(1) PAM (>30 microM vs M(2)-M(5)).
Collapse
Affiliation(s)
- Thomas M Bridges
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Bridges TM, Kennedy JP, Cho HP, Breininger ML, Gentry PR, Hopkins CR, Conn PJ, Lindsley CW. Chemical lead optimization of a pan G(q) mAChR M(1), M(3), M(5) positive allosteric modulator (PAM) lead. Part I: Development of the first highly selective M(5) PAM. Bioorg Med Chem Lett 2010; 20:558-62. [PMID: 20004578 PMCID: PMC3177601 DOI: 10.1016/j.bmcl.2009.11.089] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 11/17/2009] [Accepted: 11/18/2009] [Indexed: 11/26/2022]
Abstract
This Letter describes a chemical lead optimization campaign directed at VU0238429, the first M(5)-preferring positive allosteric modulator (PAM), discovered through analog work around VU0119498, a pan G(q) mAChR M(1), M(3), M(5) PAM. An iterative library synthesis approach delivered the first selective M(5) PAM (no activity at M(1)-M(4) @ 30microM), and an important tool compound to study the role of M(5) in the CNS.
Collapse
Affiliation(s)
- Thomas M Bridges
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Subtype-selective allosteric modulators of muscarinic receptors for the treatment of CNS disorders. Trends Pharmacol Sci 2009; 30:148-55. [PMID: 19201489 DOI: 10.1016/j.tips.2008.12.002] [Citation(s) in RCA: 225] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 12/01/2008] [Accepted: 12/03/2008] [Indexed: 01/21/2023]
Abstract
Muscarinic acetylcholine receptors (mAChRs) have long been viewed as viable targets for novel therapeutic agents for the treatment of Alzheimer's disease (AD) and other disorders involving impaired cognitive function. More recent evidence indicates that mAChR activators might also have utility in treating psychosis and other symptoms associated with schizophrenia and other central nervous system (CNS) disorders. Efforts to develop mAChR subtype-selective agonists have been hampered by difficulty in achieving high selectivity for individual mAChR subtypes important for CNS function (M(1) and M(4)) and adverse effects due to activation of peripheral mAChRs (especially M(2) and M(3)). Major advances have now been achieved in the discovery of allosteric agonists and positive allosteric modulators of M(1) and M(4) that show greater selectivity for individual mAChR subtypes than do previous mAChR agonists. Early studies indicate that these allosteric mAChR activators have properties needed for optimization as potential clinical candidates and have robust effects in animal models that predict efficacy in the treatment of AD, schizophrenia and related disorders.
Collapse
|
30
|
Miller NR, Daniels RN, Bridges TM, Brady AE, Conn PJ, Lindsley CW. Synthesis and SAR of analogs of the M1 allosteric agonist TBPB. Part II: Amides, sulfonamides and ureas--the effect of capping the distal basic piperidine nitrogen. Bioorg Med Chem Lett 2008; 18:5443-7. [PMID: 18829311 PMCID: PMC3177607 DOI: 10.1016/j.bmcl.2008.09.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 09/05/2008] [Accepted: 09/08/2008] [Indexed: 11/17/2022]
Abstract
This letter describes the further synthesis and SAR, developed through an iterative analog library approach, of analogs of the highly selective M1 allosteric agonist TBPB by deletion of the distal basic piperidine nitrogen by the formation of amides, sulfonamides and ureas. Despite the large change in basicity and topology, M1 selectivity was maintained.
Collapse
Affiliation(s)
- Nicole R Miller
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|