1
|
Poslunsey M, Wood MR, Han C, Stauffer SR, Panarese JD, Melancon BJ, Engers JL, Dickerson JW, Peng W, Noetzel MJ, Cho HP, Rodriguez AL, Hopkins CR, Morrison R, Crouch RD, Bridges TM, Blobaum AL, Boutaud O, Daniels JS, Kates MJ, Castelhano A, Rook JM, Niswender CM, Jones CK, Conn PJ, Lindsley CW. Discovery of VU0467319: an M 1 Positive Allosteric Modulator Candidate That Advanced into Clinical Trials. ACS Chem Neurosci 2025; 16:95-107. [PMID: 39660766 PMCID: PMC11697341 DOI: 10.1021/acschemneuro.4c00769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/13/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024] Open
Abstract
Herein we detail the first disclosure of VU0467319 (VU319), an M1 Positive Allosteric Modulator (PAM) clinical candidate that successfully completed a Phase I Single Ascending Dose (SAD) clinical trial. VU319 (16) is a moderately potent M1 PAM (M1 PAM EC50 = 492 nM ± 2.9 nM, 71.3 ± 9.9% ACh Max), with minimal M1 agonism (EC50 > 30 μM), that displayed high CNS penetration (Kps > 0.67 and Kp,uus > 0.9) and multispecies pharmacokinetics permissive of further development. Based on robust efficacy in multiple preclinical models of cognition, an ancillary pharmacology profile devoid of appreciable off-target activities, and a lack of cholinergic adverse effects (AEs) in rats, dogs and nonhuman primates, VU319 advanced into IND-enabling studies. After completing 4-week rat and dog GLP toxicology without AEs, including absence of cholinergic effects, the first in human Phase I SAD clinical trial of VU319 (NCT03220295) was performed at Vanderbilt, where a similar lack of adverse effects, including absence of cholinergic effects was noted. Moreover, signals of target engagement were seen at the highest dose tested. Thus, VU319 demonstrated the feasibility of achieving selective targeting of central M1 muscarinic receptors without eliciting cholinergic AEs that have plagued other drugs targeting CNS cholinergic neurotransmission.
Collapse
Affiliation(s)
- Michael
S. Poslunsey
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Michael R. Wood
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Changho Han
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Shaun R. Stauffer
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Joseph D. Panarese
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Bruce J. Melancon
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Julie L. Engers
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Jonathan W. Dickerson
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Weimin Peng
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Meredith J. Noetzel
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Hyekyung P. Cho
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Alice L. Rodriguez
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Corey R. Hopkins
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Ryan Morrison
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Rachel D. Crouch
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Anna L. Blobaum
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Olivier Boutaud
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - J. Scott Daniels
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Michael J. Kates
- Davos
Pharma, Upper Saddle River, New Jersey 07458, United States
| | | | - Jerri M. Rook
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Brain Institute, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States
| | - Carrie K. Jones
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Department
of Chemistry, Vanderbilt University, Nashville Tennessee 37232, United States
- Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
2
|
Engers JL, Bollinger KA, Capstick RA, Long MF, Bender AM, Dickerson JW, Peng W, Presley CC, Cho HP, Rodriguez AL, Niswender CM, Moran SP, Xiang Z, Blobaum AL, Boutaud O, Rook JM, Engers DW, Conn PJ, Lindsley CW. Discovery of VU6007496: Challenges in the Development of an M 1 Positive Allosteric Modulator Backup Candidate. ACS Chem Neurosci 2024; 15:3421-3433. [PMID: 39197083 DOI: 10.1021/acschemneuro.4c00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 08/30/2024] Open
Abstract
Herein we report progress toward a backup clinical candidate to the M1 positive allosteric modulator (PAM) VU319/ACP-319. Scaffold-hopping from the pyrrolo[2,3-b]pyridine-based M1 PAM VU6007477 to isomeric pyrrolo[3,2-b]pyridine and thieno[3,2-b]pyridine congeners identified several backup contenders. Ultimately, VU6007496, a pyrrolo[3,2-b]pyridine, advanced into late stage profiling, only to be plagued with unanticipated, species-specific metabolism and active/toxic metabolites which were identified in our phenotypic seizure liability in vivo screen, preventing further development. However, VU6007496 proved to be a highly selective and CNS penetrant M1 PAM, with minimal agonism, that displayed excellent multispecies IV/PO pharmacokinetics (PK), CNS penetration, no induction of long-term depression (or cholinergic toxicity) and robust efficacy in novel object recognition (minimum effective dose = 3 mg/kg p.o.). Thus, VU6007496 can serve as another valuable in vivo tool compound in rats and nonhuman primates, but not mouse, to study selective M1 activation.
Collapse
Affiliation(s)
- Julie L Engers
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Katrina A Bollinger
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Rory A Capstick
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Madeline F Long
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Aaron M Bender
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Jonathan W Dickerson
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Weimin Peng
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Christopher C Presley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Hyekyung P Cho
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Alice L Rodriguez
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Colleen M Niswender
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Sean P Moran
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Zixiu Xiang
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Anna L Blobaum
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Olivier Boutaud
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Jerri M Rook
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Darren W Engers
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - P Jeffrey Conn
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
3
|
Dong HW, Weiss K, Baugh K, Meadows MJ, Niswender CM, Neul JL. Potentiation of the muscarinic acetylcholine receptor 1 modulates neurophysiological features in a mouse model of Rett syndrome. Neurotherapeutics 2024; 21:e00384. [PMID: 38880672 PMCID: PMC11284553 DOI: 10.1016/j.neurot.2024.e00384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/17/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder primarily caused by mutations in the X chromosome-linked gene Methyl-CpG Binding Protein 2 (MECP2). Restoring MeCP2 expression after disease onset in a mouse model of RTT reverses phenotypes, providing hope for development of treatments for RTT. Translatable biomarkers of improvement and treatment responses have the potential to accelerate both preclinical and clinical evaluation of targeted therapies in RTT. Studies in people with and mouse models of RTT have identified neurophysiological features, such as auditory event-related potentials, that correlate with disease severity, suggesting that they could be useful as biomarkers of disease improvement or early treatment response. We recently demonstrated that treatment of RTT mice with a positive allosteric modulator (PAM) of muscarinic acetylcholine subtype 1 receptor (M1) improved phenotypes, suggesting that modulation of M1 activity is a potential therapy in RTT. To evaluate whether neurophysiological features could be useful biomarkers to assess the effects of M1 PAM treatment, we acutely administered the M1 PAM VU0486846 (VU846) at doses of 1, 3, 10 and 30 mg/kg in wildtype and RTT mice. This resulted in an inverted U-shaped dose response with maximal improvement of AEP features at 3 mg/kg but with no marked effect on basal EEG power or epileptiform discharges in RTT mice and no significant changes in wildtype mice. These findings suggest that M1 potentiation can improve neural circuit synchrony to auditory stimuli in RTT mice and that neurophysiological features have potential as pharmacodynamic or treatment-responsive biomarkers for preclinical and clinical evaluation of putative therapies in RTT.
Collapse
Affiliation(s)
- Hong-Wei Dong
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA; Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kelly Weiss
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Kathryn Baugh
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA
| | - Mac J Meadows
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Colleen M Niswender
- Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Chemical Biology, Nashville, TN, USA; Vanderbilt Brain Institute, Nashville, TN, USA.
| | - Jeffrey L Neul
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA; Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Nashville, TN, USA.
| |
Collapse
|
4
|
Nguyen HTM, van der Westhuizen ET, Langmead CJ, Tobin AB, Sexton PM, Christopoulos A, Valant C. Opportunities and challenges for the development of M 1 muscarinic receptor positive allosteric modulators in the treatment for neurocognitive deficits. Br J Pharmacol 2024; 181:2114-2142. [PMID: 36355830 DOI: 10.1111/bph.15982] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/06/2022] [Revised: 09/22/2022] [Accepted: 10/18/2022] [Indexed: 11/12/2022] Open
Abstract
Targeting allosteric sites of M1 muscarinic acetylcholine receptors (M1 receptors) is a promising strategy to treat neurocognitive disorders, such as Alzheimer's disease and schizophrenia. Indeed, the last two decades have seen an impressive body of work focussing on the design and development of positive allosteric modulators (PAMs) for the M1 receptor. This has led to the identification of a structurally diverse range of highly selective M1 PAMs. In preclinical models, M1 PAMs have shown rescue of cognitive deficits and improvement of endpoints predictive of symptom domains of schizophrenia. Yet, to date only a few M1 PAMs have reached early-stage clinical trials, with many of them failing to progress further due to on-target mediated cholinergic adverse effects that have plagued the development of this class of ligand. This review covers the recent preclinical and clinical studies in the field of M1 receptor drug discovery for the treatment of Alzheimer's disease and schizophrenia, with a specific focus on M1 PAM, highlighting both the undoubted potential but also key challenges for the successful translation of M1 PAMs from bench-side to bedside. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Huong T M Nguyen
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- Department of Biochemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | | | - Christopher J Langmead
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Melbourne, VIC, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, Melbourne, VIC, Australia
| | - Andrew B Tobin
- Centre for Translational Pharmacology, University of Glasgow, Glasgow, UK
| | - Patrick M Sexton
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, Melbourne, VIC, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Melbourne, VIC, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, Melbourne, VIC, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash University, Parkville, Melbourne, VIC, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Luscombe VB, Baena-López LA, Bataille CJR, Russell AJ, Greaves DR. Kinetic insights into agonist-dependent signalling bias at the pro-inflammatory G-protein coupled receptor GPR84. Eur J Pharmacol 2023; 956:175960. [PMID: 37543157 PMCID: PMC10804997 DOI: 10.1016/j.ejphar.2023.175960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/02/2022] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
GPR84 is an orphan G-protein coupled receptor (GPCR) linked to inflammation. Strategies targeting GPR84 to prevent excessive inflammation in disease are hampered by a lack of understanding of its precise functional role. We have developed heterologous cell lines with low GPR84 expression levels that phenocopy the response of primary cells in a label-free cell electrical impedance (CEI) sensing system that measures cell morphology and adhesion. We then investigated the signalling profile and membrane localisation of GPR84 upon treatment with 6-OAU and DL-175, two agonists known to differentially influence immune cell function. When compared to 6-OAU, DL-175 was found to exhibit a delayed impedance response, a delayed and suppressed activation of Akt, which together correlated with an impaired ability to internalise GPR84 from the plasma membrane. The signalling differences were transient and occurred only at early time points in the low expressing cell lines, highlighting the importance of receptor number and kinetic readouts when evaluating signalling bias. Our findings open new ways to understand GPR84 signalling and evaluate the effect of newly developed agonists.
Collapse
Affiliation(s)
- Vincent B Luscombe
- Sir William Dunn School of Pathology, South Parks Rd, University of Oxford, Oxford, Oxfordshire, OX1 3RE, United Kingdom
| | - Luis Alberto Baena-López
- Sir William Dunn School of Pathology, South Parks Rd, University of Oxford, Oxford, Oxfordshire, OX1 3RE, United Kingdom
| | - Carole J R Bataille
- Department of Chemistry, Mansfield Rd, University of Oxford, Oxford, Oxfordshire, OX1 3TA, United Kingdom
| | - Angela J Russell
- Department of Chemistry, Mansfield Rd, University of Oxford, Oxford, Oxfordshire, OX1 3TA, United Kingdom; Department of Pharmacology, Mansfield Rd, University of Oxford, Oxford, Oxfordshire, OX1 3TA, United Kingdom
| | - David R Greaves
- Sir William Dunn School of Pathology, South Parks Rd, University of Oxford, Oxford, Oxfordshire, OX1 3RE, United Kingdom.
| |
Collapse
|
6
|
Mizuguchi T, Miyano K, Yamauchi R, Yoshida Y, Takahashi H, Yamazaki A, Ono H, Inagaki M, Nonaka M, Uezono Y, Fujii H. The first structure-activity relationship study of oxytocin as a positive allosteric modulator for the µ opioid receptor. Peptides 2023; 159:170901. [PMID: 36347314 DOI: 10.1016/j.peptides.2022.170901] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/04/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/07/2022]
Abstract
Positive allosteric modulators (PAMs) of G protein-coupled receptors (GPCRs) have drawn attention as novel drug candidates. PAMs can enhance the activities of endogenous agonists which are not only secreted at appropriate times and in parts of the body, but also are immediately metabolized. Therefore, they are expected to show fewer side effects than exogeneous orthosteric ligands. Recently, we have reported that oxytocin (OT) functioned as a PAM of the μ opioid receptor (MOR) which was one of the most potent targets for analgesics. OT is thus thought to be a useful compound for the development of novel analgesics. In this study, several OT analogs were synthesized and evaluated with an intact cell-based assay to investigate the crucial structures of OT for exerting the PAM activity. The assay results indicated that the cyclic structure formed by an intramolecular disulfide bond and the three C-terminal residues containing a small Gly residue of OT were essential for their function as a MOR-PAM. Intriguingly, two analogs having an amide or an ethylene tether instead of the intramolecular disulfide bridge did not have any PAM effects. The results suggested that the disulfide linkage of OT would be a key structure for exerting the PAM activity at the MOR.
Collapse
Affiliation(s)
- Takaaki Mizuguchi
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan; Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kanako Miyano
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Risa Yamauchi
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yuki Yoshida
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hideki Takahashi
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Ami Yamazaki
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Haruka Ono
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Miku Inagaki
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Miki Nonaka
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yasuhito Uezono
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Hideaki Fujii
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan; Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
7
|
Smith M, Arthur B, Cikowski J, Holt C, Gonzalez S, Fisher NM, Vermudez SAD, Lindsley CW, Niswender CM, Gogliotti RG. Clinical and Preclinical Evidence for M 1 Muscarinic Acetylcholine Receptor Potentiation as a Therapeutic Approach for Rett Syndrome. Neurotherapeutics 2022; 19:1340-1352. [PMID: 35670902 PMCID: PMC9587166 DOI: 10.1007/s13311-022-01254-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is characterized by developmental regression, loss of communicative ability, stereotyped hand wringing, cognitive impairment, and central apneas, among many other symptoms. RTT is caused by loss-of-function mutations in a methyl-reader known as methyl-CpG-binding protein 2 (MeCP2), a protein that links epigenetic changes on DNA to larger chromatin structure. Historically, target identification for RTT has relied heavily on Mecp2 knockout mice; however, we recently adopted the alternative approach of performing transcriptional profiling in autopsy samples from RTT patients. Through this mechanism, we identified muscarinic acetylcholine receptors (mAChRs) as potential therapeutic targets. Here, we characterized a cohort of 40 temporal cortex samples from individuals with RTT and quantified significantly decreased levels of the M1, M2, M3, and M5 mAChRs subtypes relative to neurotypical controls. Of these four subtypes, M1 expression demonstrated a linear relationship with MeCP2 expression, such that M1 levels were only diminished in contexts where MeCP2 was also significantly decreased. Further, we show that M1 potentiation with the positive allosteric modulator (PAM) VU0453595 (VU595) rescued social preference, spatial memory, and associative memory deficits, as well as decreased apneas in Mecp2+/- mice. VU595's efficacy on apneas in Mecp2+/- mice was mediated by the facilitation of the transition from inspiration to expiration. Molecular analysis correlated rescue with normalized global gene expression patterns in the brainstem and hippocampus, as well as increased Gsk3β inhibition and NMDA receptor trafficking. Together, these data suggest that M1 PAMs could represent a new class of RTT therapeutics.
Collapse
Affiliation(s)
- Mackenzie Smith
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Bright Arthur
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jakub Cikowski
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Calista Holt
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Sonia Gonzalez
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sheryl Anne D Vermudez
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Rocco G Gogliotti
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA.
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
8
|
Spearing PK, Cho HP, Luscombe VB, Blobaum AL, Boutaud O, Engers DW, Rodriguez AL, Niswender CM, Jeffrey Conn P, Lindsley CW, Bender AM. Discovery of a novel class of heteroaryl-pyrrolidinones as positive allosteric modulators of the muscarinic acetylcholine receptor M 1. Bioorg Med Chem Lett 2021; 47:128193. [PMID: 34118412 DOI: 10.1016/j.bmcl.2021.128193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/09/2021] [Revised: 05/31/2021] [Accepted: 06/06/2021] [Indexed: 11/25/2022]
Abstract
This Letter describes the synthesis and optimization of a series of heteroaryl-pyrrolidinone positive allosteric modulators (PAMs) of the muscarinic acetylcholine receptor M1 (mAChR M1). Through the continued optimization of M1 PAM tool compound VU0453595, with a focus on replacement of the 6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-5-one with a wide variety of alternative 4,5-dihydropyrrolo-fused heteroaromatics, the generation of M1 PAMs with structurally novel chemotypes is disclosed. Two compounds from these subseries, 8b (VU6005610) and 20a (VU6005852), show robust selectivity for the M1 mAChR, and no M1 agonism. Both compounds have favorable preliminary PK profiles in vitro;8b additionally demonstrates high brain exposure in a rodent IV cassette model.
Collapse
Affiliation(s)
- Paul K Spearing
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Hyekyung P Cho
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Vincent B Luscombe
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Anna L Blobaum
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Olivier Boutaud
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Darren W Engers
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Alice L Rodriguez
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Colleen M Niswender
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - P Jeffrey Conn
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States; Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - Aaron M Bender
- Warren Center for Neuroscience Drug Discovery, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| |
Collapse
|
9
|
|
10
|
Mandai T, Sako Y, Kurimoto E, Shimizu Y, Nakamura M, Fushimi M, Maeda R, Miyamoto M, Kimura H. T-495, a novel low cooperative M 1 receptor positive allosteric modulator, improves memory deficits associated with cholinergic dysfunction and is characterized by low gastrointestinal side effect risk. Pharmacol Res Perspect 2020; 8:e00560. [PMID: 31990455 PMCID: PMC6986443 DOI: 10.1002/prp2.560] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 12/20/2022] Open
Abstract
M1 muscarinic acetylcholine receptor (M1 R) activation can be a new therapeutic approach for the treatment of cognitive deficits associated with cholinergic hypofunction. However, M1 R activation causes gastrointestinal (GI) side effects in animals. We previously found that an M1 R positive allosteric modulator (PAM) with lower cooperativity (α-value) has a limited impact on ileum contraction and can produce a wider margin between cognitive improvement and GI side effects. In fact, TAK-071, a novel M1 R PAM with low cooperativity (α-value of 199), improved scopolamine-induced cognitive deficits with a wider margin against GI side effects than a high cooperative M1 R PAM, T-662 (α-value of 1786), in rats. Here, we describe the pharmacological characteristics of a novel low cooperative M1 R PAM T-495 (α-value of 170), using the clinically tested higher cooperative M1 R PAM MK-7622 (α-value of 511) as a control. In rats, T-495 caused diarrhea at a 100-fold higher dose than that required for the improvement of scopolamine-induced memory deficits. Contrastingly, MK-7622 showed memory improvement and induction of diarrhea at an equal dose. Combination of T-495, but not of MK-7622, and donepezil at each sub-effective dose improved scopolamine-induced memory deficits. Additionally, in mice with reduced acetylcholine levels in the forebrain via overexpression of A53T α-synuclein (ie, a mouse model of dementia with Lewy bodies and Parkinson's disease with dementia), T-495, like donepezil, reversed the memory deficits in the contextual fear conditioning test and Y-maze task. Thus, low cooperative M1 R PAMs are promising agents for the treatment of memory deficits associated with cholinergic dysfunction.
Collapse
Affiliation(s)
- Takao Mandai
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuu Sako
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Emi Kurimoto
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuji Shimizu
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan.,Biomolecular Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Minoru Nakamura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Makoto Fushimi
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Ryouta Maeda
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Maki Miyamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Haruhide Kimura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
11
|
Scarpa M, Hesse S, Bradley SJ. M1 muscarinic acetylcholine receptors: A therapeutic strategy for symptomatic and disease-modifying effects in Alzheimer's disease? ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 88:277-310. [PMID: 32416870 DOI: 10.1016/bs.apha.2019.12.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022]
Abstract
The M1 muscarinic acetylcholine receptor (mAChR) plays a crucial role in learning and memory processes and has long been identified as a promising therapeutic target for the improvement of cognitive decline in Alzheimer's disease (AD). As such, clinical trials with xanomeline, a mAChR orthosteric agonist, showed an improvement in cognitive and behavioral symptoms associated with AD. Despite this, the clinical utility of xanomeline was hampered by a lack of M1 receptor selectivity and adverse cholinergic responses attributed to activation of peripheral M2 and M3 mAChRs. More recently, efforts have focused on developing more selective M1 compounds via targeting the less-conserved allosteric binding pockets. As such, positive allosteric modulators (PAMs) have emerged as an exciting strategy to achieve exquisite selectivity for the M1 mAChR in order to deliver improvements in cognitive function in AD. Furthermore, over recent years it has become increasingly apparent that M1 therapeutics may also offer disease-modifying effects in AD, due to the modulation of pathogenic amyloid processing. This article will review the progress made in the development of M1 selective ligands for the treatment of cognitive decline in AD, and will discuss the current evidence that selective targeting of the M1 mAChR could also have the potential to modify AD progression.
Collapse
Affiliation(s)
- Miriam Scarpa
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sarah Hesse
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sophie J Bradley
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
12
|
Moran SP, Xiang Z, Doyle CA, Maksymetz J, Lv X, Faltin S, Fisher NM, Niswender CM, Rook JM, Lindsley CW, Conn PJ. Biased M 1 receptor-positive allosteric modulators reveal role of phospholipase D in M 1-dependent rodent cortical plasticity. Sci Signal 2019; 12:12/610/eaax2057. [PMID: 31796631 DOI: 10.1126/scisignal.aax2057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
Highly selective, positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have emerged as an exciting new approach to potentially improve cognitive function in patients suffering from Alzheimer's disease and schizophrenia. Discovery programs have produced a structurally diverse range of M1 receptor PAMs with distinct pharmacological properties, including different extents of agonist activity and differences in signal bias. This includes biased M1 receptor PAMs that can potentiate coupling of the receptor to activation of phospholipase C (PLC) but not phospholipase D (PLD). However, little is known about the role of PLD in M1 receptor signaling in native systems, and it is not clear whether biased M1 PAMs display differences in modulating M1-mediated responses in native tissue. Using PLD inhibitors and PLD knockout mice, we showed that PLD was necessary for the induction of M1-dependent long-term depression (LTD) in the prefrontal cortex (PFC). Furthermore, biased M1 PAMs that did not couple to PLD not only failed to potentiate orthosteric agonist-induced LTD but also blocked M1-dependent LTD in the PFC. In contrast, biased and nonbiased M1 PAMs acted similarly in potentiating M1-dependent electrophysiological responses that were PLD independent. These findings demonstrate that PLD plays a critical role in the ability of M1 PAMs to modulate certain central nervous system (CNS) functions and that biased M1 PAMs function differently in brain regions implicated in cognition.
Collapse
Affiliation(s)
- Sean P Moran
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Zixiu Xiang
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Catherine A Doyle
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - James Maksymetz
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Xiaohui Lv
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Sehr Faltin
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Jerri M Rook
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA. .,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| |
Collapse
|
13
|
Adenosine Receptor Profiling Reveals an Association between the Presence of Spare Receptors and Cardiovascular Disorders. Int J Mol Sci 2019; 20:ijms20235964. [PMID: 31783510 PMCID: PMC6928742 DOI: 10.3390/ijms20235964] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/24/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023] Open
Abstract
Adenosine and its receptors exert a potent control on the cardiovascular system. This review aims to present emerging experimental evidence supporting the existence and implication in cardiovascular disorders of specific adenosinergic pharmacological profiles, conforming to the concept of "receptor reserve", also known as "spare receptors". This kind of receptors allow agonists to achieve their maximal effect without occupying all of the relevant cell receptors. In the cardiovascular system, spare adenosine receptors appear to compensate for a low extracellular adenosine level and/or a low adenosine receptor number, such as in coronary artery disease or some kinds of neurocardiogenic syncopes. In both cases, the presence of spare receptors appears to be an attempt to overcome a weak interaction between adenosine and its receptors. The identification of adenosine spare receptors in cardiovascular disorders may be helpful for diagnostic purposes.
Collapse
|
14
|
Moran SP, Maksymetz J, Conn PJ. Targeting Muscarinic Acetylcholine Receptors for the Treatment of Psychiatric and Neurological Disorders. Trends Pharmacol Sci 2019; 40:1006-1020. [PMID: 31711626 DOI: 10.1016/j.tips.2019.10.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/05/2019] [Revised: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022]
Abstract
Muscarinic acetylcholine receptors (mAChR) play important roles in regulating complex behaviors such as cognition, movement, and reward, making them ideally situated as potential drug targets for the treatment of several brain disorders. Recent advances in the discovery of subtype-selective allosteric modulators for mAChRs has provided an unprecedented opportunity for highly specific modulation of signaling by individual mAChR subtypes in the brain. Recently, mAChR allosteric modulators have entered clinical development for Alzheimer's disease (AD) and schizophrenia, and have potential utility for other brain disorders. However, mAChR allosteric modulators can display a diverse array of pharmacological properties, and a more nuanced understanding of the mAChR will be necessary to best translate preclinical findings into successful clinical treatments.
Collapse
Affiliation(s)
- Sean P Moran
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - James Maksymetz
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
15
|
Miura S, Fukuda K, Masada S, Usutani H, Kanematsu M, Cork DG, Kawamoto T. Rapid and efficient synthesis of a novel cholinergic muscarinic M 1 receptor positive allosteric modulator using flash chemistry. Org Biomol Chem 2019; 17:8166-8174. [PMID: 31464336 DOI: 10.1039/c9ob01718f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022]
Abstract
Continuous flow-flash synthesis of a 2-bromobenzaldehyde derivative 18 as a key intermediate of a novel cholinergic muscarinic M1 positive allosteric modulator 1 bearing an isoindolin-1-one ring system as a pharmacophore has been achieved using flow microreactors through selective I/Li exchange of 1-bromo-2-iodobenzene derivative 17 with BuLi and subsequent formylation at -40 °C of the highly reactive 2-bromophenyllithium intermediate using DMF, which is difficult to achieve by a conventional batch process due to the conversion of the highly reactive 2-bromophenyllithium intermediate into benzyne even at -78 °C. Late-stage cyclization to give the isoindolin-1-one ring system, through reductive amination of 18 followed by palladium-catalyzed carbonylation with carbon monoxide and intramolecular cyclization, efficiently afforded 1 for its further research and development.
Collapse
Affiliation(s)
- Shotaro Miura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Ltd, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Koichiro Fukuda
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Ltd, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Shinichi Masada
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Ltd, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Hirotsugu Usutani
- Process Chemistry, Pharmaceutical Sciences, Takeda Pharmaceutical Company Ltd, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Makoto Kanematsu
- Process Chemistry, Pharmaceutical Sciences, Takeda Pharmaceutical Company Ltd, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - David G Cork
- Process Chemistry, Pharmaceutical Sciences, Takeda Pharmaceutical Company Ltd, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tetsuji Kawamoto
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Ltd, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
16
|
Engers JL, Bender AM, Kalbfleisch JJ, Cho HP, Lingenfelter KS, Luscombe VB, Han C, Melancon BJ, Blobaum AL, Dickerson JW, Rook JM, Niswender CM, Emmitte KA, Conn PJ, Lindsley CW. Discovery of Tricyclic Triazolo- and Imidazopyridine Lactams as M 1 Positive Allosteric Modulators. ACS Chem Neurosci 2019; 10:1035-1042. [PMID: 30086237 DOI: 10.1021/acschemneuro.8b00311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022] Open
Abstract
This Letter describes the chemical optimization of a new series of muscarinic acetylcholine receptor subtype 1 (M1) positive allosteric modulators (PAMs) based on novel tricyclic triazolo- and imidazopyridine lactam cores, devoid of M1 agonism, e.g., no M1 ago-PAM activity, in high expressing recombinant cell lines. While all the new tricyclic congeners afforded excellent rat pharmacokinetic (PK) properties (CLp < 8 mL/min/kg and t1/2 > 5 h), regioisomeric triazolopyridine analogues were uniformly not CNS penetrant ( Kp < 0.05), despite a lack of hydrogen bond donors. However, removal of a single nitrogen atom to afford imidazopyridine derivatives proved to retain the excellent rat PK and provide high CNS penetration ( Kp > 2), despite inclusion of a basic nitrogen. Moreover, 24c was devoid of M1 agonism in high expressing recombinant cell lines and did not induce cholinergic seizures in vivo in mice. Interestingly, all of the new M1 PAMs across the diverse tricyclic heterocyclic cores possessed equivalent CNS MPO scores (>4.5), highlighting the value of both "medicinal chemist's eye" and experimental data, e.g., not sole reliance (or decision bias) on in silico calculated properties, for parameters as complex as CNS penetration.
Collapse
Affiliation(s)
- Julie L. Engers
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Aaron M. Bender
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Jacob J. Kalbfleisch
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Hyekyung P. Cho
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kaelyn S. Lingenfelter
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Vincent B. Luscombe
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Changho Han
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Bruce J. Melancon
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Warren Family Research Center for Drug Discovery & Development, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Anna L. Blobaum
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Jonathan W. Dickerson
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Jerri M. Rook
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kyle A. Emmitte
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
17
|
Rook JM, Bertron JL, Cho HP, Garcia-Barrantes PM, Moran SP, Maksymetz JT, Nance KD, Dickerson JW, Remke DH, Chang S, Harp JM, Blobaum AL, Niswender CM, Jones CK, Stauffer SR, Conn PJ, Lindsley CW. A Novel M 1 PAM VU0486846 Exerts Efficacy in Cognition Models without Displaying Agonist Activity or Cholinergic Toxicity. ACS Chem Neurosci 2018; 9:2274-2285. [PMID: 29701957 PMCID: PMC6146057 DOI: 10.1021/acschemneuro.8b00131] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022] Open
Abstract
Selective activation of the M1 subtype of muscarinic acetylcholine receptor, via positive allosteric modulation (PAM), is an exciting strategy to improve cognition in schizophrenia and Alzheimer's disease patients. However, highly potent M1 ago-PAMs, such as MK-7622, PF-06764427, and PF-06827443, can engender excessive activation of M1, leading to agonist actions in the prefrontal cortex (PFC) that impair cognitive function, induce behavioral convulsions, and result in other classic cholinergic adverse events (AEs). Here, we report a fundamentally new and highly selective M1 PAM, VU0486846. VU0486846 possesses only weak agonist activity in M1-expressing cell lines with high receptor reserve and is devoid of agonist actions in the PFC, unlike previously reported ago-PAMs MK-7622, PF-06764427, and PF-06827443. Moreover, VU0486846 shows no interaction with antagonist binding at the orthosteric acetylcholine (ACh) site (e.g., neither bitopic nor displaying negative cooperativity with [3H]-NMS binding at the orthosteric site), no seizure liability at high brain exposures, and no cholinergic AEs. However, as opposed to ago-PAMs, VU0486846 produces robust efficacy in the novel object recognition model of cognitive function. Importantly, we show for the first time that an M1 PAM can reverse the cognitive deficits induced by atypical antipsychotics, such as risperidone. These findings further strengthen the argument that compounds with modest in vitro M1 PAM activity (EC50 > 100 nM) and pure-PAM activity in native tissues display robust procognitive efficacy without AEs mediated by excessive activation of M1. Overall, the combination of compound assessment with recombinant in vitro assays (mindful of receptor reserve), native tissue systems (PFC), and phenotypic screens (behavioral convulsions) is essential to fully understand and evaluate lead compounds and enhance success in clinical development.
Collapse
Affiliation(s)
- Jerri M. Rook
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Jeanette L. Bertron
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Hyekyung P. Cho
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Pedro M. Garcia-Barrantes
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Sean P. Moran
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - James T. Maksymetz
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Kellie D. Nance
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Jonathan W. Dickerson
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Daniel H. Remke
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Sichen Chang
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Joel M. Harp
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Anna L. Blobaum
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Colleen M. Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Carrie K. Jones
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Shaun R. Stauffer
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| | - Craig W. Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| |
Collapse
|
18
|
Engers JL, Childress ES, Long MF, Capstick RA, Luscombe VB, Cho HP, Dickerson JW, Rook JM, Blobaum AL, Niswender CM, Engers DW, Conn PJ, Lindsley CW. VU6007477, a Novel M 1 PAM Based on a Pyrrolo[2,3- b]pyridine Carboxamide Core Devoid of Cholinergic Adverse Events. ACS Med Chem Lett 2018; 9:917-922. [PMID: 30258541 DOI: 10.1021/acsmedchemlett.8b00261] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/07/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
Herein, we report the chemical optimization of a new series of M1 positive allosteric modulators (PAMs) based on a novel pyrrolo[2,3-b]pyridine core, developed via scaffold hopping and iterative parallel synthesis. The vast majority of analogs in this series proved to display robust cholinergic seizure activity. However, by removal of the secondary hydroxyl group, VU6007477 resulted with good rat M1 PAM potency (EC50 = 230 nM, 93% ACh max), minimal M1 agonist activity (agonist EC50 > 10 μM), good CNS penetration (rat brain/plasma K p = 0.28, K p,uu = 0.32; mouse K p = 0.16, K p,uu = 0.18), and no cholinergic adverse events (AEs, e.g., seizures). This work demonstrates that within a chemical series prone to robust M1 ago-PAM activity, SAR can result, which affords pure M1 PAMs, devoid of cholinergic toxicity/seizure liability.
Collapse
Affiliation(s)
- Julie L. Engers
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Elizabeth S. Childress
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Madeline F. Long
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Rory A. Capstick
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Vincent B. Luscombe
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Hekyung P. Cho
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jonathan W. Dickerson
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jerri M. Rook
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Anna L. Blobaum
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Darren W. Engers
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, ‡Department of Pharmacology, §Department of Chemistry, ∥Department of Biochemistry, and ⊥Vanderbilt Kennedy Center, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|