1
|
Horan WP, Sauder C, Harvey PD, Ramsay IS, Yohn SE, Keefe RSE, Davis VG, Paul SM, Brannan SK. The Impact of Xanomeline and Trospium Chloride on Cognitive Impairment in Acute Schizophrenia: Replication in Pooled Data From Two Phase 3 Trials. Am J Psychiatry 2024. [PMID: 39659157 DOI: 10.1176/appi.ajp.20240076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
OBJECTIVE Xanomeline and trospium chloride (formerly known as KarXT), a novel M1/M4 muscarinic receptor agonist, demonstrated efficacy across phase 2 and 3 trials as monotherapy for the treatment of inpatients with acute schizophrenia on the Positive and Negative Syndrome Scale total score primary endpoint. In the phase 2 trial, xanomeline/trospium improved performance on a cognitive outcome measure in the subgroup of participants with clinically significant baseline cognitive impairment. The authors sought to confirm this finding using data from two phase 3 trials. METHODS Data were pooled from two 5-week inpatient trials of xanomeline/trospium monotherapy in patients with acute schizophrenia. The statistical analysis plan prespecified comparisons of cognitive composite score changes between xanomeline/trospium and placebo in the full sample and the cognitively impaired (≤1 SD below norms at baseline) subgroup. RESULTS There was no significant xanomeline/trospium effect in the full sample (N=357); however, in the impaired subgroup, xanomeline/trospium (N=71) had a significantly greater benefit for cognition compared with placebo (N=66; least squares mean difference=0.31, SE=0.10; d=0.54). The xanomeline/trospium effect size increased significantly with a more stringent baseline impairment threshold (≤-1.5 SD; d=0.80). Improvements in cognition were minimally correlated with concurrent changes in total, positive, and negative symptoms in both treatment groups. CONCLUSIONS Participants with acute schizophrenia with prespecified impairments demonstrated significant cognitive improvement with xanomeline/trospium compared with placebo. This result directly confirms earlier findings. This benefit is not attributable to changes in symptoms, despite substantial evidence of efficacy for psychosis. Evaluation of xanomeline/trospium's potential for cognitive enhancement in a well-controlled trial of stable patients with cognitive impairment is warranted.
Collapse
Affiliation(s)
- William P Horan
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| | - Colin Sauder
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| | - Philip D Harvey
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| | - Ian S Ramsay
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| | - Samantha E Yohn
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| | - Richard S E Keefe
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| | - Vicki G Davis
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| | - Steven M Paul
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| | - Stephen K Brannan
- Bristol Myers Squibb, Boston (Horan, Sauder, Ramsay, Yohn, Paul, Brannan); University of Miami Miller School of Medicine, Miami (Harvey); Duke University School of Medicine, Durham (Keefe); Statistical Consultant, Chapel Hill (Davis)
| |
Collapse
|
2
|
Walker LC, Huckstep KL, Becker HC, Langmead CJ, Lawrence AJ. Targeting muscarinic receptors for the treatment of alcohol use disorders: Opportunities and hurdles for clinical development. Br J Pharmacol 2024; 181:4385-4398. [PMID: 37005377 DOI: 10.1111/bph.16081] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/04/2023] Open
Abstract
Emerging evidence suggests muscarinic acetylcholine receptors represent novel targets to treat alcohol use disorder. In this review, we draw from literature across medicinal chemistry, molecular biology, addiction and learning/cognition fields to interrogate the proposition for muscarinic receptor ligands in treating various aspects of alcohol use disorder, including cognitive dysfunction, motivation to consume alcohol and relapse. In support of this proposition, we describe cholinergic dysfunction in the pathophysiology of alcohol use disorder at a network level, including alcohol-induced adaptations present in both human post-mortem brains and reverse-translated rodent models. Preclinical behavioural pharmacology implicates specific muscarinic receptors, in particular, M4 and M5 receptors, as potential therapeutic targets worthy of further interrogation. We detail how these receptors can be selectively targeted in vivo by the use of subtype-selective allosteric modulators, a strategy that overcomes the issue of targeting a highly conserved orthosteric site bound by acetylcholine. Finally, we highlight the intense pharma interest in allosteric modulators of muscarinic receptors for other indications that provide an opportunity for repurposing into the alcohol use disorder space and provide some currently unanswered questions as a roadmap for future investigation.
Collapse
Affiliation(s)
- Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Kade L Huckstep
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
3
|
Paul SM, Yohn SE, Brannan SK, Neugebauer NM, Breier A. Muscarinic Receptor Activators as Novel Treatments for Schizophrenia. Biol Psychiatry 2024; 96:627-637. [PMID: 38537670 DOI: 10.1016/j.biopsych.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 05/26/2024]
Abstract
Achieving optimal treatment outcomes for individuals living with schizophrenia remains challenging, despite 70 years of drug development efforts. Many chemically distinct antipsychotics have been developed over the past 7 decades with improved safety and tolerability but with only slight variation in efficacy. All antipsychotics currently approved for the treatment of schizophrenia act as antagonists or partial agonists at the dopamine D2 receptor. With only a few possible exceptions, antipsychotic drugs have similar and modest efficacy for treating positive symptoms and are relatively ineffective in addressing the negative and cognitive symptoms of the disease. The development of novel treatments focused on targeting muscarinic acetylcholine receptors (mAChRs) has been of interest for more than 25 years following reports that treatment with a dual M1/M4-preferring mAChR agonist resulted in antipsychotic-like effects and procognitive properties in individuals living with Alzheimer's disease and schizophrenia; more recent clinical trials have confirmed these findings. In addition, advances in our understanding of the receptor binding and activation properties of xanomeline at specific mAChRs have the potential to inform future drug design targeting mAChRs.
Collapse
Affiliation(s)
- Steven M Paul
- Karuna Therapeutics, Boston, Massachusetts; Department of Psychiatry and Neurology, Washington University of St. Louis, St. Louis, Missouri.
| | | | | | | | - Alan Breier
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
4
|
Yohn SE, Harvey PD, Brannan SK, Horan WP. The potential of muscarinic M 1 and M 4 receptor activators for the treatment of cognitive impairment associated with schizophrenia. Front Psychiatry 2024; 15:1421554. [PMID: 39483736 PMCID: PMC11525114 DOI: 10.3389/fpsyt.2024.1421554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/03/2024] [Indexed: 11/03/2024] Open
Abstract
Cognitive impairment is a core symptom of schizophrenia and a major determinant of poor long-term functional outcomes. Despite considerable efforts, we do not yet have any approved pharmacological treatments for cognitive impairment associated with schizophrenia (CIAS). A combination of advances in pre-clinical research and recent clinical trial findings have led to a resurgence of interest in the cognition-enhancing potential of novel muscarinic acetylcholine receptor (mAChR) agonists in schizophrenia. This article provides an overview of the scientific rationale for targeting M1 and M4 mAChRs. We describe the evolution of neuroscience research on these receptors since early drug discovery efforts focused on the mAChR agonist xanomeline. This work has revealed that M1 and M4 mAChRs are highly expressed in brain regions that are implicated in cognition. The functional significance of M1 and M4 mAChRs has been extensively characterized in animal models via use of selective receptor subtype compounds through neuronal and non-neuronal mechanisms. Recent clinical trials of a dual M1/M4 mAChR agonist show promising, replicable evidence of potential pro-cognitive effects in schizophrenia, with several other mAChR agonists in clinical development.
Collapse
Affiliation(s)
| | - Phillip D. Harvey
- Division of Psychology, University of Miami, Miami, FL, United States
| | | | - William P. Horan
- Bristol Myers Squibb, Princeton, NJ, United States
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
5
|
Tobin AB. A golden age of muscarinic acetylcholine receptor modulation in neurological diseases. Nat Rev Drug Discov 2024; 23:743-758. [PMID: 39143241 DOI: 10.1038/s41573-024-01007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 08/16/2024]
Abstract
Over the past 40 years, the muscarinic acetylcholine receptor family, particularly the M1-receptor and M4-receptor subtypes, have emerged as validated targets for the symptomatic treatment of neurological diseases such as schizophrenia and Alzheimer disease. However, despite considerable effort and investment, no drugs have yet gained clinical approval. This is largely attributable to cholinergic adverse effects that have halted the majority of programmes and resulted in a waning of interest in these G-protein-coupled receptor targets. Recently, this trend has been reversed. Driven by advances in structure-based drug design and an appreciation of the optimal pharmacological properties necessary to deliver clinical efficacy while minimizing adverse effects, a new generation of M1-receptor and M4-receptor orthosteric agonists and positive allosteric modulators are now entering the clinic. These agents offer the prospect of novel therapeutic solutions for 'hard to treat' neurological diseases, heralding a new era of muscarinic drug discovery.
Collapse
Affiliation(s)
- Andrew B Tobin
- Centre for Translational Pharmacology, School of Molecular Biosciences, The Advanced Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
6
|
Burns JN, Jenkins AK, Xue X, Petersen KA, Ketchesin KD, Perez MS, Vadnie CA, Scott MR, Seney ML, Tseng GC, McClung CA. Comparative transcriptomic rhythms in the mouse and human prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615154. [PMID: 39386590 PMCID: PMC11463408 DOI: 10.1101/2024.09.26.615154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Alterations in multiple subregions of the human prefrontal cortex (PFC) have been heavily implicated in psychiatric diseases. Moreover, emerging evidence suggests that circadian rhythms in gene expression are present across the brain, including in the PFC, and that these rhythms are altered in disease. However, investigation into the potential circadian mechanisms underlying these diseases in animal models must contend with the fact that the human PFC is highly evolved and specialized relative to that of rodents. Here, we use RNA sequencing to lay the groundwork for translational studies of molecular rhythms through a sex-specific, cross species comparison of transcriptomic rhythms between the mouse medial PFC (mPFC) and two subregions of the human PFC, the anterior cingulate cortex (ACC) and the dorsolateral PFC (DLPFC). We find that while circadian rhythm signaling is conserved across species and subregions, there is a phase shift in the expression of core clock genes between the mouse mPFC and human PFC subregions that differs by sex. Furthermore, we find that the identity of rhythmic transcripts is largely unique between the mouse mPFC and human PFC subregions, with the most overlap (20%, 236 transcripts) between the mouse mPFC and the human ACC in females. Nevertheless, we find that basic biological processes are enriched for rhythmic transcripts across species, with key differences between regions and sexes. Together, this work highlights both the evolutionary conservation of transcriptomic rhythms and the advancement of the human PFC, underscoring the importance of considering cross-species differences when using animal models.
Collapse
Affiliation(s)
- Jennifer N. Burns
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Aaron K. Jenkins
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kaitlyn A. Petersen
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Megan S. Perez
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261
| | - Chelsea A. Vadnie
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH 43015
| | - Madeline R. Scott
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Marianne L. Seney
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
7
|
Cadeddu R, Braccagni G, Branca C, van Luik ER, Pittenger C, Thomsen MS, Bortolato M. Activation of M 4 muscarinic receptors in the striatum reduces tic-like behaviours in two distinct murine models of Tourette syndrome. Br J Pharmacol 2024; 181:3064-3081. [PMID: 38689378 DOI: 10.1111/bph.16392] [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] [Scholar Register] [Received: 08/30/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND AND PURPOSE Current pharmacotherapies for Tourette syndrome (TS) are often unsatisfactory and poorly tolerated, underscoring the need for novel treatments. Insufficient striatal acetylcholine has been suggested to contribute to tic ontogeny. Thus, we tested whether activating M1 and/or M4 receptors-the two most abundant muscarinic receptors in the striatum-reduced tic-related behaviours in mouse models of TS. EXPERIMENTAL APPROACH Studies were conducted using CIN-d and D1CT-7 mice, two TS models characterized by early-life depletion of striatal cholinergic interneurons and cortical neuropotentiation, respectively. First, we tested the effects of systemic and intrastriatal xanomeline, a selective M1/M4 receptor agonist, on tic-like and other TS-related responses. Then, we examined whether xanomeline effects were reduced by either M1 or M4 antagonists or mimicked by the M1/M3 agonist cevimeline or the M4 positive allosteric modulator (PAM) VU0467154. Finally, we measured striatal levels of M1 and M4 receptors and assessed the impact of VU0461754 on the striatal expression of the neural marker activity c-Fos. KEY RESULTS Systemic and intrastriatal xanomeline reduced TS-related behaviours in CIN-d and D1CT-7 mice. Most effects were blocked by M4, but not M1, receptor antagonists. VU0467154, but not cevimeline, elicited xanomeline-like ameliorative effects in both models. M4, but not M1, receptors were down-regulated in the striatum of CIN-d mice. Additionally, VU0467154 reduced striatal c-Fos levels in these animals. CONCLUSION AND IMPLICATIONS Activation of striatal M4, but not M1, receptors reduced tic-like manifestations in mouse models, pointing to xanomeline and M4 PAMs as novel putative therapeutic strategies for TS.
Collapse
Affiliation(s)
- Roberto Cadeddu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Giulia Braccagni
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Caterina Branca
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Easton R van Luik
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Christopher Pittenger
- Department of Psychiatry, School of Medicine, Yale University, New Haven, Connecticut, USA
- Department of Psychology, School of Arts and Sciences, Yale University, New Haven, Connecticut, USA
- Child Study Center, School of Medicine, Yale University, New Haven, Connecticut, USA
- Center for Brain and Mind Health, School of Medicine, Yale University, New Haven, Connecticut, USA
| | | | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
8
|
Dudzik P, Lustyk K, Pytka K. Beyond dopamine: Novel strategies for schizophrenia treatment. Med Res Rev 2024; 44:2307-2330. [PMID: 38653551 DOI: 10.1002/med.22042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Despite extensive research efforts aimed at discovering novel antipsychotic compounds, a satisfactory pharmacological strategy for schizophrenia treatment remains elusive. All the currently available drugs act by modulating dopaminergic neurotransmission, leading to insufficient management of the negative and cognitive symptoms of the disorder. Due to these challenges, several attempts have been made to design agents with innovative, non-dopaminergic mechanisms of action. Consequently, a number of promising compounds are currently progressing through phases 2 and 3 of clinical trials. This review aims to examine the rationale behind the most promising of these strategies while simultaneously providing a comprehensive survey of study results. We describe the versatility behind the cholinergic neurotransmission modulation through the activation of M1 and M4 receptors, exemplified by the prospective drug candidate KarXT. Our discussion extends to the innovative approach of activating TAAR1 receptors via ulotaront, along with the promising outcomes of iclepertin, a GlyT-1 inhibitor with the potential to become the first treatment option for cognitive impairment associated with schizophrenia. Finally, we evaluate the 5-HT2A antagonist paradigm, assessing two recently developed serotonergic agents, pimavanserin and roluperidone. We present the latest advancements in developing novel solutions to the complex challenges posed by schizophrenia, offering an additional perspective on the diverse investigated drug candidates.
Collapse
Affiliation(s)
- Paulina Dudzik
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Klaudia Lustyk
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
9
|
Butler CR, Popiolek M, McAllister LA, LaChapelle EA, Kramer M, Beck EM, Mente S, Brodney MA, Brown M, Gilbert A, Helal C, Ogilvie K, Starr J, Uccello D, Grimwood S, Edgerton J, Garst-Orozco J, Kozak R, Lotarski S, Rossi A, Smith D, O'Connor R, Lazzaro J, Steppan C, Steyn SJ. Design and Synthesis of Clinical Candidate PF-06852231 (CVL-231): A Brain Penetrant, Selective, Positive Allosteric Modulator of the M 4 Muscarinic Acetylcholine Receptor. J Med Chem 2024; 67:10831-10847. [PMID: 38888621 DOI: 10.1021/acs.jmedchem.4c00293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Selective activation of the M4 muscarinic acetylcholine receptor subtype offers a novel strategy for the treatment of psychosis in multiple neurological disorders. Although the development of traditional muscarinic activators has been stymied due to pan-receptor activation, muscarinic receptor subtype selectivity can be achieved through the utilization of a subtype of a unique allosteric site. A major challenge in capitalizing on this allosteric site to date has been achieving a balance of suitable potency and brain penetration. Herein, we describe the design of a brain penetrant series of M4 selective positive allosteric modulators (PAMs), ultimately culminating in the identification of 21 (PF-06852231, now CVL-231/emraclidine), which is under active clinical development as a novel mechanism and approach for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Christopher R Butler
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Michael Popiolek
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Laura A McAllister
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Erik A LaChapelle
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Melissa Kramer
- Medicine Design, Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Elizabeth M Beck
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Scot Mente
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Michael A Brodney
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Matthew Brown
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Adam Gilbert
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Chris Helal
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Kevin Ogilvie
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Jeremy Starr
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Daniel Uccello
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Sarah Grimwood
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Jeremy Edgerton
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | | | - Rouba Kozak
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Susan Lotarski
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Amie Rossi
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Deborah Smith
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Rebecca O'Connor
- Discovery Sciences, Primary Pharmacology, Pfizer Inc., Groton, Connecticut 06340, United States
| | - John Lazzaro
- Discovery Sciences, Primary Pharmacology, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Claire Steppan
- Discovery Sciences, Primary Pharmacology, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Stefanus J Steyn
- Medicine Design, Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| |
Collapse
|
10
|
Lawn T, Giacomel A, Martins D, Veronese M, Howard M, Turkheimer FE, Dipasquale O. Normative modelling of molecular-based functional circuits captures clinical heterogeneity transdiagnostically in psychiatric patients. Commun Biol 2024; 7:689. [PMID: 38839931 PMCID: PMC11153627 DOI: 10.1038/s42003-024-06391-3] [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] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/27/2024] [Indexed: 06/07/2024] Open
Abstract
Advanced methods such as REACT have allowed the integration of fMRI with the brain's receptor landscape, providing novel insights transcending the multiscale organisation of the brain. Similarly, normative modelling has allowed translational neuroscience to move beyond group-average differences and characterise deviations from health at an individual level. Here, we bring these methods together for the first time. We used REACT to create functional networks enriched with the main modulatory, inhibitory, and excitatory neurotransmitter systems and generated normative models of these networks to capture functional connectivity deviations in patients with schizophrenia, bipolar disorder (BPD), and ADHD. Substantial overlap was seen in symptomatology and deviations from normality across groups, but these could be mapped into a common space linking constellations of symptoms through to underlying neurobiology transdiagnostically. This work provides impetus for developing novel biomarkers that characterise molecular- and systems-level dysfunction at the individual level, facilitating the transition towards mechanistically targeted treatments.
Collapse
Affiliation(s)
- Timothy Lawn
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Alessio Giacomel
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Matthew Howard
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Department of Research & Development Advanced Applications, Olea Medical, La Ciotat, France.
| |
Collapse
|
11
|
Birgül Iyison N, Abboud C, Abboud D, Abdulrahman AO, Bondar AN, Dam J, Georgoussi Z, Giraldo J, Horvat A, Karoussiotis C, Paz-Castro A, Scarpa M, Schihada H, Scholz N, Güvenc Tuna B, Vardjan N. ERNEST COST action overview on the (patho)physiology of GPCRs and orphan GPCRs in the nervous system. Br J Pharmacol 2024. [PMID: 38825750 DOI: 10.1111/bph.16389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/09/2024] [Accepted: 02/24/2024] [Indexed: 06/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of cell surface receptors that play a critical role in nervous system function by transmitting signals between cells and their environment. They are involved in many, if not all, nervous system processes, and their dysfunction has been linked to various neurological disorders representing important drug targets. This overview emphasises the GPCRs of the nervous system, which are the research focus of the members of ERNEST COST action (CA18133) working group 'Biological roles of signal transduction'. First, the (patho)physiological role of the nervous system GPCRs in the modulation of synapse function is discussed. We then debate the (patho)physiology and pharmacology of opioid, acetylcholine, chemokine, melatonin and adhesion GPCRs in the nervous system. Finally, we address the orphan GPCRs, their implication in the nervous system function and disease, and the challenges that need to be addressed to deorphanize them.
Collapse
Affiliation(s)
- Necla Birgül Iyison
- Department of Molecular Biology and Genetics, University of Bogazici, Istanbul, Turkey
| | - Clauda Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | - Dayana Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | | | - Ana-Nicoleta Bondar
- Faculty of Physics, University of Bucharest, Magurele, Romania
- Forschungszentrum Jülich, Institute for Computational Biomedicine (IAS-5/INM-9), Jülich, Germany
| | - Julie Dam
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
| | - Zafiroula Georgoussi
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Christos Karoussiotis
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Alba Paz-Castro
- Molecular Pharmacology of GPCRs research group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago, Spain
| | - Miriam Scarpa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Nicole Scholz
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Bilge Güvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
12
|
Fu L, Luo Y, Niu L, Lin Y, Chen X, Zhang J, Tang W, Chen Y, Jiao Y. M 1/M 4 receptors as potential therapeutic treatments for schizophrenia: A comprehensive study. Bioorg Med Chem 2024; 105:117728. [PMID: 38640587 DOI: 10.1016/j.bmc.2024.117728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Muscarinic acetylcholine receptors (mAChRs) play a significant role in the pathophysiology of schizophrenia. Although activating mAChRs holds potential in addressing the full range of schizophrenia symptoms, clinical application of many non-selective mAChR agonists in cognitive deficits, positive and negative symptoms is hindered by peripheral side effects (gastrointestinal disturbances and cardiovascular effects) and dosage restrictions. Ligands binding to the allosteric sites of mAChRs, particularly the M1 and M4 subtypes, demonstrate activity in improving cognitive function and amelioration of positive and negative symptoms associated with schizophrenia, enhancing our understanding of schizophrenia. The article aims to critically examine current design concepts and clinical advancements in synthesizing and designing small molecules targeting M1/M4, providing theoretical insights and empirical support for future research in this field.
Collapse
Affiliation(s)
- Lingsheng Fu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yi Luo
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Longyan Niu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Ying Lin
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Xingru Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Junhao Zhang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Weifang Tang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China..
| | - Yadong Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China..
| | - Yu Jiao
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China..
| |
Collapse
|
13
|
Dean B. Muscarinic M1 and M4 receptor agonists for schizophrenia: promising candidates for the therapeutic arsenal. Expert Opin Investig Drugs 2023; 32:1113-1121. [PMID: 37994870 DOI: 10.1080/13543784.2023.2288074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/22/2023] [Indexed: 11/24/2023]
Abstract
INTRODUCTION Successful phase 3 trials of KarXT in people with schizophrenia herald a new era of treating the disorder with drugs that do not target the dopamine D2 receptor. The active component of KarXT is xanomeline, a muscarinic (CHRM) M1 and M4 agonist, making muscarinic receptors a viable target for treating schizophrenia. AREAS COVERED This review covers the process of taking drugs that activate the muscarinic M1 and M4 receptors from conceptualization to the clinic and details the mechanisms by which activating the CHRM1 and 4 can affect the broad spectrum of symptoms experienced by people with schizophrenia. EXPERT OPINION Schizophrenia is a syndrome which means drugs that activate muscarinic M1 and M4 receptors, as was the case for antipsychotic drugs acting on the dopamine D2 receptor, will not give optimal outcomes in everyone within the syndrome. Thus, it would be ideal to identify people who are responsive to drugs activating the CHRM1 and 4. Given knowledge of the actions of these receptors, it is possible treatment non-response could be restricted to sub-groups within the syndrome who have deficits in cortical CHRM1 or those with one of the cognitive endophenotypes that may be identifiable by changes in the blood transcriptome.
Collapse
Affiliation(s)
- Brian Dean
- The Synaptic Biology and Cognition Laboratory, The Florey, Parkville, Victoria, Australia
- Florey Department of Neuroscience and Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Chambers NE, Millett M, Moehle MS. The muscarinic M4 acetylcholine receptor exacerbates symptoms of movement disorders. Biochem Soc Trans 2023; 51:691-702. [PMID: 37013974 PMCID: PMC10212540 DOI: 10.1042/bst20220525] [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] [Scholar Register] [Received: 09/18/2022] [Revised: 12/31/2022] [Accepted: 03/14/2023] [Indexed: 04/05/2023]
Abstract
Barbeau's seesaw hypothesis of dopamine-acetylcholine balance has predominated movement disorders literature for years. Both the simplicity of the explanation and the matching efficacy of anticholinergic treatment in movement disorders seem to support this hypothesis. However, evidence from translational and clinical studies in movement disorders indicates that many features of this simple balance are lost, broken, or absent from movement disorders models or in imaging studies of patients with these disorders. This review reappraises the dopamine-acetylcholine balance hypothesis in light of recent evidence and describes how the Gαi/o coupled muscarinic M4 receptor acts in opposition to dopamine signaling in the basal ganglia. We highlight how M4 signaling can ameliorate or exacerbate movement disorders symptoms and physiological correlates of these symptoms in specific disease states. Furthermore, we propose future directions for investigation of this mechanisms to fully understand the potential efficacy of M4 targeting therapeutics in movement disorders. Overall, initial evidence suggest that M4 is a promising pharmaceutical target to ameliorate motor symptoms of hypo- and hyper-dopaminergic disorders.
Collapse
Affiliation(s)
- Nicole E. Chambers
- Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Michael Millett
- Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Mark S. Moehle
- Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| |
Collapse
|
15
|
Teal LB, Bubser M, Duncan E, Gould RW, Lindsley CW, Jones CK. Selective M 5 muscarinic acetylcholine receptor negative allosteric modulator VU6008667 blocks acquisition of opioid self-administration. Neuropharmacology 2023; 227:109424. [PMID: 36720403 DOI: 10.1016/j.neuropharm.2023.109424] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/04/2023] [Accepted: 01/16/2023] [Indexed: 01/30/2023]
Abstract
Recent evidence suggests that inhibition of the M5 muscarinic acetylcholine receptor (mAChR) may provide a novel non-opioid mechanism for the treatment of opioid use disorder (OUD). Previous studies from our group and others have demonstrated that acute administration of the long-acting M5 negative allosteric modulator (NAM) ML375 attenuates established self-administration of cocaine, ethanol, oxycodone, and remifentanil in rats. In the present study, we characterized the effects of acute and repeated administration of the novel, short-acting M5 NAM VU6008667 on the reinforcing effects of oxycodone and reinstatement of oxycodone-seeking behaviors in male Sprague-Dawley rats, as well as on physiological withdrawal from oxycodone. Acute VU6008667 decreased oxycodone self-administration under both fixed ratio 3 (FR3) and progressive ratio (PR) schedules of reinforcement and attenuated cue-induced reinstatement of lever pressing following extinction from oxycodone self-administration, a commonly used relapse model. When administered daily to opioid-naïve rats, VU6008667 prevented acquisition of oxycodone self-administration behavior. VU6008667 had minimal effects on naloxone-precipitated withdrawal. After acute administration, VU6008667 did not inhibit sucrose self-administration and, when given chronically, delayed but did not prevent acquisition of sucrose maintained self-administration. VU6008667 also did not impact oxycodone induced anti-nociception or motor coordination, but mildly decreased novelty exploration. Finally, acute or daily VU6008667 administration did not impair cued fear conditioning. Overall, these results suggest that inhibition of the M5 mAChR may provide a novel, non-opioid based treatment for distinct aspects of OUD by inhibiting opioid intake in established OUD, reducing relapse during abstinence, and by reducing the risk of developing OUD.
Collapse
Affiliation(s)
- Laura B Teal
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Michael Bubser
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Edith Duncan
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Robert W Gould
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA; Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA; Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Carrie K Jones
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
16
|
Dean B, Bakker G, Ueda HR, Tobin AB, Brown A, Kanaan RAA. A growing understanding of the role of muscarinic receptors in the molecular pathology and treatment of schizophrenia. Front Cell Neurosci 2023; 17:1124333. [PMID: 36909280 PMCID: PMC9992992 DOI: 10.3389/fncel.2023.1124333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Pre-clinical models, postmortem and neuroimaging studies all support a role for muscarinic receptors in the molecular pathology of schizophrenia. From these data it was proposed that activation of the muscarinic M1 and/or M4 receptor would reduce the severity of the symptoms of schizophrenia. This hypothesis is now supported by results from two clinical trials which indicate that activating central muscarinic M1 and M4 receptors can reduce the severity of positive, negative and cognitive symptoms of the disorder. This review will provide an update on a growing body of evidence that argues the muscarinic M1 and M4 receptors have critical roles in CNS functions that are dysregulated by the pathophysiology of schizophrenia. This realization has been made possible, in part, by the growing ability to visualize and quantify muscarinic M1 and M4 receptors in the human CNS using molecular neuroimaging. We will discuss how these advances have provided evidence to support the notion that there is a sub-group of patients within the syndrome of schizophrenia that have a unique molecular pathology driven by a marked loss of muscarinic M1 receptors. This review is timely, as drugs targeting muscarinic receptors approach clinical use for the treatment of schizophrenia and here we outline the background biology that supported development of such drugs to treat the disorder.
Collapse
Affiliation(s)
- Brian Dean
- Synaptic Biology and Cognition Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | | | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Andrew B Tobin
- Advanced Research Centre (ARC), School of Molecular Bioscience, University of Glasgow, Glasgow, United Kingdom
| | | | - Richard A A Kanaan
- Department of Psychiatry, Austin Health, The University of Melbourne, Heidelberg, VIC, Australia
| |
Collapse
|
17
|
Paul SM, Yohn SE, Popiolek M, Miller AC, Felder CC. Muscarinic Acetylcholine Receptor Agonists as Novel Treatments for Schizophrenia. Am J Psychiatry 2022; 179:611-627. [PMID: 35758639 DOI: 10.1176/appi.ajp.21101083] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Schizophrenia remains a challenging disease to treat effectively with current antipsychotic medications due to their limited efficacy across the entire spectrum of core symptoms as well as their often burdensome side-effect profiles and poor tolerability. An unmet need remains for novel, mechanistically unique, and better tolerated therapeutic agents for treating schizophrenia, especially those that treat not only positive symptoms but also the negative and cognitive symptoms of the disease. Almost 25 years ago, the muscarinic acetylcholine receptor (mAChR) agonist xanomeline was reported to reduce psychotic symptoms and improve cognition in patients with Alzheimer's disease. The antipsychotic and procognitive properties of xanomeline were subsequently confirmed in a small study of acutely psychotic patients with chronic schizophrenia. These unexpected clinical findings have prompted considerable efforts across academia and industry to target mAChRs as a new approach to potentially treat schizophrenia and other psychotic disorders. The authors discuss recent advances in mAChR biology and pharmacology and the current understanding of the relative roles of the various mAChR subtypes, their downstream cellular effectors, and key neural circuits mediating the reduction in the core symptoms of schizophrenia in patients treated with xanomeline. They also provide an update on the status of novel mAChR agonists currently in development for potential treatment of schizophrenia and other neuropsychiatric disorders.
Collapse
|
18
|
Cikowski J, Holt C, Arthur B, Smith M, Gonzalez S, Lindsley CW, Niswender CM, Gogliotti RG. Optimized Administration of the M 4 PAM VU0467154 Demonstrates Broad Efficacy, but Limited Effective Concentrations in Mecp2+/- Mice. ACS Chem Neurosci 2022; 13:1891-1901. [PMID: 35671352 PMCID: PMC9266622 DOI: 10.1021/acschemneuro.2c00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Hypofunction of cholinergic circuits and diminished cholinergic tone have been associated with the neurodevelopmental disorder Rett syndrome (RTT). Specifically, deletion of Mecp2 in cholinergic neurons evokes the same social and cognitive phenotypes in mice seen with global Mecp2 knockout, and decreased choline acetyltransferase activity and vesamicol binding have been reported in RTT autopsy samples. Further, we recently identified significant decreases in muscarinic acetylcholine receptor subtype 4 (M4) expression in both the motor cortex and cerebellum of RTT patient autopsies and established proof of concept that an acute dose of the positive allosteric modulator (PAM) VU0467154 (VU154) rescued phenotypes in Mecp2+/- mice. Here, we expand the assessment of M4 PAMs in RTT to address clinically relevant questions of tolerance, scope of benefit, dose response, chronic treatment, and mechanism. We show that VU154 has efficacy on anxiety, social preference, cognitive, and respiratory phenotypes in Mecp2+/- mice; however, the therapeutic range is narrow, with benefits seen at 3 mg/kg concentrations, but not 1 or 10 mg/kg. Further, sociability was diminished in VU154-treated Mecp2+/- mice, suggestive of a potential adverse effect. Compound efficacy on social, cognitive, and respiratory phenotypes was conserved with a 44-day treatment paradigm, with the caveat that breath rate was moderately decreased with chronic treatment in Mecp2+/+ and Mecp2+/- mice. VU154 effects on respiratory function correlated with an increase in Gsk3β inhibition in the brainstem. These results identify the core symptom domains where efficacy and adverse effects may present with M4 administration in RTT model mice and advocate for the continued evaluation as potential RTT therapeutics.
Collapse
Affiliation(s)
- Jakub Cikowski
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States
| | - Calista Holt
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States
| | - Bright Arthur
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States,Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Mackenzie Smith
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States
| | - Sonia Gonzalez
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States
| | - Craig W. Lindsley
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States,Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States,Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States,Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States,Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States,Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States,Vanderbilt
Brain Institute, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Rocco G. Gogliotti
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States,Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States,Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,. Phone: 708-216-9021. Fax: 708-216-8318
| |
Collapse
|
19
|
Inactivation of the cholinergic M4 receptor results in a disinhibited endophenotype predicting alcohol use. Behav Brain Res 2022; 430:113921. [DOI: 10.1016/j.bbr.2022.113921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/20/2022] [Accepted: 05/04/2022] [Indexed: 11/21/2022]
|
20
|
Spark DL, Fornito A, Langmead CJ, Stewart GD. Beyond antipsychotics: a twenty-first century update for preclinical development of schizophrenia therapeutics. Transl Psychiatry 2022; 12:147. [PMID: 35393394 PMCID: PMC8991275 DOI: 10.1038/s41398-022-01904-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/02/2022] [Accepted: 03/15/2022] [Indexed: 11/15/2022] Open
Abstract
Despite 50+ years of drug discovery, current antipsychotics have limited efficacy against negative and cognitive symptoms of schizophrenia, and are ineffective-with the exception of clozapine-against any symptom domain for patients who are treatment resistant. Novel therapeutics with diverse non-dopamine D2 receptor targets have been explored extensively in clinical trials, yet often fail due to a lack of efficacy despite showing promise in preclinical development. This lack of translation between preclinical and clinical efficacy suggests a systematic failure in current methods that determine efficacy in preclinical rodent models. In this review, we critically evaluate rodent models and behavioural tests used to determine preclinical efficacy, and look to clinical research to provide a roadmap for developing improved translational measures. We highlight the dependence of preclinical models and tests on dopamine-centric theories of dysfunction and how this has contributed towards a self-reinforcing loop away from clinically meaningful predictions of efficacy. We review recent clinical findings of distinct dopamine-mediated dysfunction of corticostriatal circuits in patients with treatment-resistant vs. non-treatment-resistant schizophrenia and suggest criteria for establishing rodent models to reflect such differences, with a focus on objective, translational measures. Finally, we review current schizophrenia drug discovery and propose a framework where preclinical models are validated against objective, clinically informed measures and preclinical tests of efficacy map onto those used clinically.
Collapse
Affiliation(s)
- Daisy L Spark
- Drug Discovery Biology, Neuroscience & Mental Health Therapeutic Program Area, and Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, Monash Biomedical Imaging, and School of Psychological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Christopher J Langmead
- Drug Discovery Biology, Neuroscience & Mental Health Therapeutic Program Area, and Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| | - Gregory D Stewart
- Drug Discovery Biology, Neuroscience & Mental Health Therapeutic Program Area, and Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
21
|
Thomsen M, Crittenden JR, Lindsley CW, Graybiel AM. Effects of acute and repeated administration of the selective M 4 PAM VU0152099 on cocaine versus food choice in male rats. Addict Biol 2022; 27:e13145. [PMID: 35229940 PMCID: PMC9162150 DOI: 10.1111/adb.13145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/29/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022]
Abstract
Ligands that stimulate muscarinic acetylcholine receptors 1 and 4 (M1 , M4 ) have shown promising effects as putative pharmacotherapy for cocaine use disorder in rodent assays. We have previously shown reductions in cocaine effects with acute M4 stimulation, as well as long-lasting, delayed reductions in cocaine taking and cocaine seeking with combined M1 /M4 receptor stimulation or with M1 stimulation alone. M4 stimulation opposes dopaminergic signalling acutely, but direct dopamine receptor antagonists have proved unhelpful in managing cocaine use disorder because they lose efficacy with long-term administration. It is therefore critical to determine whether M4 approaches themselves can remain effective with repeated or chronic dosing. We assessed the effects of repeated administration of the M4 positive allosteric modulator (PAM) VU0152099 in rats trained to choose between intravenous cocaine and a liquid food reinforcer to obtain quantitative measurement of whether M4 stimulation could produce delayed and lasting reduction in cocaine taking. VU0152099 produced progressively augmenting suppression of cocaine choice and cocaine intake, but produced neither rebound nor lasting effects after treatment ended. To compare and contrast effects of M1 versus M4 stimulation, we tested whether the M4 PAM VU0152100 suppressed cocaine self-administration in mice lacking CalDAG-GEFI signalling factor, required for M1 -mediated suppression of cocaine self-administration. CalDAG-GEFI ablation had no effect on M4 -mediated suppression of cocaine self-administration. These findings support the potential usefulness of M4 PAMs as pharmacotherapy to manage cocaine use disorder, alone or in combination with M1 -selective ligands, and show that M1 and M4 stimulation modulate cocaine-taking behaviour by distinct mechanisms.
Collapse
Affiliation(s)
- Morgane Thomsen
- Neuroscience and Behavioral Pharmacology Laboratory, Alcohol and Drug Abuse Research Center, Division of Basic Research, McLean Hospital/Harvard Medical School, Belmont, MA, USA
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services in the Capital Region of Denmark, Copenhagen, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jill R. Crittenden
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Craig W. Lindsley
- Vanderbilt Program in Drug Discovery, Vanderbilt Specialized Chemistry Center (Molecular Libraries Probe Production Centers Network; MLPCN), Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ann M. Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
22
|
Li W, Wang Y, Lohith TG, Zeng Z, Tong L, Mazzola R, Riffel K, Miller P, Purcell M, Holahan M, Haley H, Gantert L, Hesk D, Ren S, Morrow J, Uslaner J, Struyk A, Wai JMC, Rudd MT, Tellers DM, McAvoy T, Bormans G, Koole M, Van Laere K, Serdons K, de Hoon J, Declercq R, De Lepeleire I, Pascual MB, Zanotti-Fregonara P, Yu M, Arbones V, Masdeu JC, Cheng A, Hussain A, Bueters T, Anderson MS, Hostetler ED, Basile AS. The PET tracer [ 11C]MK-6884 quantifies M4 muscarinic receptor in rhesus monkeys and patients with Alzheimer's disease. Sci Transl Med 2022; 14:eabg3684. [PMID: 35020407 DOI: 10.1126/scitranslmed.abg3684] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Wenping Li
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Yuchuan Wang
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | - Zhizhen Zeng
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Ling Tong
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | - Kerry Riffel
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | - Mona Purcell
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | - Hyking Haley
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Liza Gantert
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - David Hesk
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Sumei Ren
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - John Morrow
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | - Arie Struyk
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | | | | | | | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, KU Leuven, 3001 Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, KU Leuven and University Hospital Leuven, 3001 Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, KU Leuven and University Hospital Leuven, 3001 Leuven, Belgium
| | - Kim Serdons
- Nuclear Medicine and Molecular Imaging, KU Leuven and University Hospital Leuven, 3001 Leuven, Belgium
| | - Jan de Hoon
- Center for Clinical Pharmacology, KU Leuven, 3001 Leuven, Belgium
| | - Ruben Declercq
- Translational Pharmacology Europe, MSD (Europe) Inc., 1200 Brussels, Belgium
| | - Inge De Lepeleire
- Translational Pharmacology Europe, MSD (Europe) Inc., 1200 Brussels, Belgium
| | - Maria B Pascual
- Nantz National Alzheimer Center, Houston Methodist Neurological Institute, Houston, TX 77030, USA.,Department of Neurology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center, Houston Methodist Neurological Institute, Houston, TX 77030, USA.,Department of Neurology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Meixiang Yu
- Nantz National Alzheimer Center, Houston Methodist Neurological Institute, Houston, TX 77030, USA.,Department of Neurology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Victoria Arbones
- Nantz National Alzheimer Center, Houston Methodist Neurological Institute, Houston, TX 77030, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Houston Methodist Neurological Institute, Houston, TX 77030, USA.,Department of Neurology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Amy Cheng
- MRL, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | | | | | | | | |
Collapse
|
23
|
Vaidya S, Guerin AA, Walker LC, Lawrence AJ. Clinical Effectiveness of Muscarinic Receptor-Targeted Interventions in Neuropsychiatric Disorders: A Systematic Review. CNS Drugs 2022; 36:1171-1206. [PMID: 36269510 PMCID: PMC9653329 DOI: 10.1007/s40263-022-00964-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND For decades, treatment of mood disorders, psychoses, anxiety and dementia have been confounded by limited efficacy and high rates of treatment resistance. Preclinical and clinical evidence have highlighted disruption of cholinergic signalling in several neuropsychiatric conditions and examined intervention strategies including acetylcholinesterase inhibitors and nicotinic receptor-targeted intervention. However, the effectiveness of these approaches is often curtailed by on-target side effects. Post mortem studies implicate muscarinic receptor dysregulation in neuropsychiatric pathophysiology; therefore, we conducted a systematic review and meta-analysis to investigate the therapeutic efficacy and safety of muscarinic receptor-targeted interventions in adults with neuropsychiatric disorders. METHODS PubMed, EMBASE, PsycINFO, EBSCO and Web of Science were searched using relevant keywords from database inception to 7 August 2022. Randomised, double-blind, placebo-controlled studies were included if they investigated the effect of muscarinic receptor-targeted intervention in adults with a diagnosis of a neuropsychiatric disorder and were published in English. A narrative synthesis approach was adopted to describe the findings. Wherever three or more studies with a similar intervention were available, effect sizes were calculated, and a meta-analysis was performed. Cochrane risk-of-bias-2 tool was utilised to assess the risk of bias, and sensitivity analyses were performed to identify publication bias. Certainty analysis (high, moderate, low and/or very low) was conducted using GRADE criteria. RESULTS Overall, 33 studies met the inclusion criteria and 5 were included in the meta-analysis. Despite a limited pool with several different interventions, we found therapeutic efficacy of xanomeline (M1/M4 agonist) in primary psychotic disorders plus behavioural and psychological symptoms of dementia. Scopolamine showed a significant antidepressant effect in a combined cohort of major depressive and bipolar disorders in the short-term outcome measure, but no effect following cessation of treatment. Results from bias assessments suggest "very low" certainty in the antidepressant effect of scopolamine. Critical limitations of the current literature included low power, high heterogeneity in the patient population and a lack of active comparators. CONCLUSION While the results are not definitive, findings on muscarinic receptor-targeted interventions in several mental disorders are promising in terms of efficacy and safety, specifically in treating schizophrenia, mood disorders, and behavioural and psychiatric symptoms of Alzheimer's disease. However, orthosteric muscarinic receptor-targeted interventions are associated with a range of peripheral adverse effects that are thought to be mediated via M2/M3 receptors. The orthosteric binding site of muscarinic acetylcholine receptors is remarkably conserved, posing a challenge for subtype-selective interventions; nonetheless allosteric ligands with biased signalling pathways are now in development. We conclude that adequately powered prospective studies with subtype-selective interventions are required to determine the clinical effectiveness of muscarinic-receptor targeted interventions for the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Shivani Vaidya
- Florey Institute of Neuroscience & Mental Health, Royal Parade, Parkville, VIC 3010 Australia ,Florey Department of Neuroscience & Mental Health, University of Melbourne, Parkville, VIC 3010 Australia
| | - Alexandre A. Guerin
- Centre for Youth Mental Health, University of Melbourne, 35 Poplar Rd, Parkville, VIC 3052 Australia ,Orygen, 35 Poplar Rd, Parkville, VIC 3052 Australia
| | - Leigh C. Walker
- Florey Institute of Neuroscience & Mental Health, Royal Parade, Parkville, VIC 3010 Australia ,Florey Department of Neuroscience & Mental Health, University of Melbourne, Parkville, VIC 3010 Australia
| | - Andrew J. Lawrence
- Florey Institute of Neuroscience & Mental Health, Royal Parade, Parkville, VIC 3010 Australia ,Florey Department of Neuroscience & Mental Health, University of Melbourne, Parkville, VIC 3010 Australia
| |
Collapse
|
24
|
Effects of a novel M4 muscarinic positive allosteric modulator on behavior and cognitive deficits relevant to Alzheimer's disease and schizophrenia in rhesus monkey. Neuropharmacology 2021; 197:108754. [PMID: 34389398 DOI: 10.1016/j.neuropharm.2021.108754] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/19/2021] [Accepted: 08/08/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a profoundly debilitating neurodegenerative disorder characterized most notably by progressive cognitive decline, but also agitation and behavioral disturbances that are extremely disruptive to patient and caregiver. Current pharmacological treatments for these symptoms have limited efficacy and significant side effects. We have recently reported the discovery of Compound 24, an M4 positive allosteric modulator (PAM) that is potent, highly selective, and devoid of cholinergic-like side effects in rats. In order to further evaluate the translatability of the effects of compound 24 in primates, here we describe the effect of Compound 24 on three behavioral and cognition assays in rhesus monkeys, the stimulant induced motor activity (SIMA) assay, the object retrieval detour task (ORD), and the visuo-spatial paired-associates learning (vsPAL) task. As far as we know, this is the first such characterization of an M4 PAM in non-human primate. Compound 24 and the clinical standard olanzapine attenuated amphetamine induced hyperactivity to a similar degree. In addition, Compound 24 demonstrated procognitive effects in scopolamine-impaired ORD and vsPAL, and these effects were of similar magnitude to donepezil. These findings suggest that M4 PAMs may be beneficial to diseases such as Alzheimer's disease and schizophrenia, which are marked by behavioral disturbances as well as deficits in cognitive function.
Collapse
|
25
|
Moehle MS, Bender AM, Dickerson JW, Foster DJ, Qi A, Cho HP, Donsante Y, Peng W, Bryant Z, Stillwell KJ, Bridges TM, Chang S, Watson KJ, O’Neill JC, Engers JL, Peng L, Rodriguez AL, Niswender CM, Lindsley CW, Hess EJ, Conn PJ, Rook JM. Discovery of the First Selective M 4 Muscarinic Acetylcholine Receptor Antagonists with in Vivo Antiparkinsonian and Antidystonic Efficacy. ACS Pharmacol Transl Sci 2021; 4:1306-1321. [PMID: 34423268 PMCID: PMC8369681 DOI: 10.1021/acsptsci.0c00162] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Indexed: 11/30/2022]
Abstract
Nonselective antagonists of muscarinic acetylcholine receptors (mAChRs) that broadly inhibit all five mAChR subtypes provide an efficacious treatment for some movement disorders, including Parkinson's disease and dystonia. Despite their efficacy in these and other central nervous system disorders, antimuscarinic therapy has limited utility due to severe adverse effects that often limit their tolerability by patients. Recent advances in understanding the roles that each mAChR subtype plays in disease pathology suggest that highly selective ligands for individual subtypes may underlie the antiparkinsonian and antidystonic efficacy observed with the use of nonselective antimuscarinic therapeutics. Our recent work has indicated that the M4 muscarinic acetylcholine receptor has several important roles in opposing aberrant neurotransmitter release, intracellular signaling pathways, and brain circuits associated with movement disorders. This raises the possibility that selective antagonists of M4 may recapitulate the efficacy of nonselective antimuscarinic therapeutics and may decrease or eliminate the adverse effects associated with these drugs. However, this has not been directly tested due to lack of selective antagonists of M4. Here, we utilize genetic mAChR knockout animals in combination with nonselective mAChR antagonists to confirm that the M4 receptor activation is required for the locomotor-stimulating and antiparkinsonian efficacy in rodent models. We also report the synthesis, discovery, and characterization of the first-in-class selective M4 antagonists VU6013720, VU6021302, and VU6021625 and confirm that these optimized compounds have antiparkinsonian and antidystonic efficacy in pharmacological and genetic models of movement disorders.
Collapse
Affiliation(s)
- Mark S. Moehle
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Department
of Pharmacology & Therapeutics, Center for Translational Research
in Neurodegeneration, University of Florida, Gainesville, Florida 32610, United States
| | - Aaron M. Bender
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jonathan W. Dickerson
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Daniel J. Foster
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Aidong Qi
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Hyekyung P. Cho
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Yuping Donsante
- Department
of Pharmacology & Chemical Biology, Emory University, Atlanta, Georgia 30322, United States
| | - Weimin Peng
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Zoey Bryant
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Kaylee J. Stillwell
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Sichen Chang
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Katherine J. Watson
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jordan C. O’Neill
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Julie L. Engers
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Li Peng
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Alice L. Rodriguez
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Ellen J. Hess
- Department
of Pharmacology & Chemical Biology, Emory University, Atlanta, Georgia 30322, United States
| | - P. Jeffrey Conn
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232, United States,E-mail:
| | - Jerri M. Rook
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,E-mail:
| |
Collapse
|
26
|
Holter KM, Lekander AD, LaValley CM, Bedingham EG, Pierce BE, Sands LP, Lindsley CW, Jones CK, Gould RW. Partial mGlu 5 Negative Allosteric Modulator M-5MPEP Demonstrates Antidepressant-Like Effects on Sleep Without Affecting Cognition or Quantitative EEG. Front Neurosci 2021; 15:700822. [PMID: 34276300 PMCID: PMC8283128 DOI: 10.3389/fnins.2021.700822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/03/2021] [Indexed: 01/28/2023] Open
Abstract
Selective negative allosteric modulators (NAMs) targeting the metabotropic glutamate receptor subtype 5 (mGlu5) demonstrate anxiolytic-like and antidepressant-like effects yet concern regarding adverse effect liability remains. Functional coupling of mGlu5 with ionotropic N-methyl-D-aspartate receptors (NMDARs) represents a potential mechanism through which full inhibition leads to adverse effects, as NMDAR inhibition can induce cognitive impairments and psychotomimetic-like effects. Recent development of "partial" mGlu5 NAMs, characterized by submaximal but saturable levels of blockade, may represent a novel development approach to broaden the therapeutic index of mGlu5 NAMs. This study compared the partial mGlu5 NAM, M-5MPEP, with the full mGlu5 NAM, VU0424238 on sleep, cognition, and brain function alone and in combination with a subthreshold dose of the NMDAR antagonist, MK-801, using a paired-associates learning (PAL) cognition task and electroencephalography (EEG) in rats. M-5MPEP and VU0424238 decreased rapid eye movement (REM) sleep and increased REM sleep latency, both putative biomarkers of antidepressant-like activity. Neither compound alone affected accuracy, but 30 mg/kg VU0424238 combined with MK-801 decreased accuracy on the PAL task. Using quantitative EEG, VU0424238, but not M-5MPEP, prolonged arousal-related elevations in high gamma power, and, in combination, VU0424238 potentiated effects of MK-801 on high gamma power. Together, these studies further support a functional interaction between mGlu5 and NMDARs that may correspond with cognitive impairments. Present data support further development of partial mGlu5 NAMs given their potentially broader therapeutic index than full mGlu5 NAMs and use of EEG as a translational biomarker to titrate doses aligning with therapeutic versus adverse effects.
Collapse
Affiliation(s)
- Kimberly M. Holter
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Alex D. Lekander
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Christina M. LaValley
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | | | - Bethany E. Pierce
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - L. Paul Sands
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Craig W. Lindsley
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
- Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Carrie K. Jones
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Robert W. Gould
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
27
|
Walker LC, Huckstep KL, Chen NA, Hand LJ, Lindsley CW, Langmead CJ, Lawrence AJ. Muscarinic M 4 and M 5 receptors in the ventral subiculum differentially modulate alcohol seeking versus consumption in male alcohol-preferring rats. Br J Pharmacol 2021; 178:3730-3746. [PMID: 33942300 DOI: 10.1111/bph.15513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Muscarinic acetylcholine receptors mediate alcohol consumption and seeking in rats. While M4 and M5 receptors have recently been implicated to mediate these behaviours in the striatum, their role in other brain regions remain unknown. The ventral tegmental area (VTA) and ventral subiculum (vSub) both densely express M4 and M5 receptors and modulate alcohol-seeking, via their projections to the nucleus accumbens shell (AcbSh). EXPERIMENTAL APPROACH In Indiana alcohol-preferring (iP) male rats, we examined Chrm4 (M4 ) and Chrm5 (M5 ) expression in the VTA and vSub following long-term alcohol consumption and abstinence using RT-qPCR. Using a combination of retrograde tracing and RNAscope, we examined the localisation of Chrm4 and Chrm5 on vSub cells that project to the AcbSh. Using selective allosteric modulators, we examined the functional role of M4 and M5 receptors within the vSub in alcohol consumption, context-induced alcohol-seeking, locomotor activity, and food/water consumption. KEY RESULTS Long-term alcohol and abstinence dysregulated the expression of genes for muscarinic receptors in the vSub, not in the VTA. Chrm4 was down-regulated following long-term alcohol and abstinence, while Chrm5 was up-regulated following long-term alcohol consumption. Consistent with these data, a positive allosteric modulator (VU0467154) of intra-vSub M4 receptors reduced context-induced alcohol-seeking, but not motivation for alcohol self-administration, while M5 receptor negative allosteric modulator (ML375) reduced initial motivation for alcohol self-administration, but not context-induced alcohol-seeking. CONCLUSION AND IMPLICATIONS Collectively, our data highlight alcohol-induced cholinergic dysregulation in the vSub and distinct roles for M4 and M5 receptor allosteric modulators to reduce alcohol consumption or seeking.
Collapse
Affiliation(s)
- Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Kate L Huckstep
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Nicola A Chen
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Lexi J Hand
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
28
|
Foster DJ, Bryant ZK, Conn PJ. Targeting muscarinic receptors to treat schizophrenia. Behav Brain Res 2021; 405:113201. [PMID: 33647377 PMCID: PMC8006961 DOI: 10.1016/j.bbr.2021.113201] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/02/2021] [Accepted: 02/18/2021] [Indexed: 11/23/2022]
Abstract
Schizophrenia is a severe neuropsychiatric disorder characterized by a diverse range of symptoms that can have profound impacts on the lives of patients. Currently available antipsychotics target dopamine receptors, and while they are useful for ameliorating the positive symptoms of the disorder, this approach often does not significantly improve negative and cognitive symptoms. Excitingly, preclinical and clinical research suggests that targeting specific muscarinic acetylcholine receptor subtypes could provide more comprehensive symptomatic relief with the potential to ameliorate numerous symptom domains. Mechanistic studies reveal that M1, M4, and M5 receptor subtypes can modulate the specific brain circuits and physiology that are disrupted in schizophrenia and are thought to underlie positive, negative, and cognitive symptoms. Novel therapeutic strategies for targeting these receptors are now advancing in clinical and preclinical development and expand upon the promise of these new treatment strategies to potentially provide more comprehensive relief than currently available antipsychotics.
Collapse
Affiliation(s)
- Daniel J Foster
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States
| | - Zoey K Bryant
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States.
| |
Collapse
|
29
|
Walker LC, Berizzi AE, Chen NA, Rueda P, Perreau VM, Huckstep K, Srisontiyakul J, Govitrapong P, Xiaojian J, Lindsley CW, Jones CK, Riddy DM, Christopoulos A, Langmead CJ, Lawrence AJ. Acetylcholine Muscarinic M 4 Receptors as a Therapeutic Target for Alcohol Use Disorder: Converging Evidence From Humans and Rodents. Biol Psychiatry 2020; 88:898-909. [PMID: 32331824 PMCID: PMC11390032 DOI: 10.1016/j.biopsych.2020.02.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/03/2020] [Accepted: 02/19/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a major socioeconomic burden on society, and current pharmacotherapeutic treatment options are inadequate. Aberrant alcohol use and seeking alters frontostriatal function. METHODS We performed genome-wide RNA sequencing and subsequent quantitative polymerase chain reaction and receptor binding validation in the caudate-putamen of human AUD samples to identify potential therapeutic targets. We then back-translated our top candidate targets into a rodent model of long-term alcohol consumption to assess concordance of molecular adaptations in the rat striatum. Finally, we adopted rat behavioral models of alcohol intake and seeking to validate a potential therapeutic target. RESULTS We found that G protein-coupled receptors were the top canonical pathway differentially regulated in individuals with AUD. The M4 muscarinic acetylcholine receptor (mAChR) was downregulated at the gene and protein levels in the putamen, but not in the caudate, of AUD samples. We found concordant downregulation of the M4 mAChR, specifically on dopamine D1 receptor-expressing medium spiny neurons in the rat dorsolateral striatum. Systemic administration of the selective M4 mAChR positive allosteric modulator, VU0467154, reduced home cage and operant alcohol self-administration, motivation to obtain alcohol, and cue-induced reinstatement of alcohol seeking in rats. Local microinjections of VU0467154 in the rat dorsolateral striatum reduced alcohol self-administration and cue-induced reinstatement of alcohol seeking. CONCLUSIONS Collectively, these results identify the M4 mAChR as a potential therapeutic target for the treatment of AUD and the D1 receptor-positive medium spiny neurons in the dorsolateral striatum as a key site mediating the actions of M4 mAChR in relation to alcohol consumption and seeking.
Collapse
Affiliation(s)
- Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Alice E Berizzi
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Nicola A Chen
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Patricia Rueda
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Victoria M Perreau
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Katherine Huckstep
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Jirawoot Srisontiyakul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Jia Xiaojian
- Shenzhen Kangning Hospital, Shenzhen University Health Science Center, Shenzhen, China; Shenzhen Mental Health Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt Center for Neuroscience and Drug Discovery, Vanderbilt University, Nashville, Tennessee; Department of Chemistry, Vanderbilt Center for Neuroscience and Drug Discovery, Vanderbilt University, Nashville, Tennessee
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt Center for Neuroscience and Drug Discovery, Vanderbilt University, Nashville, Tennessee; Department of Chemistry, Vanderbilt Center for Neuroscience and Drug Discovery, Vanderbilt University, Nashville, Tennessee
| | - Darren M Riddy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
30
|
Deng X, Zhang Y, Rong J, Kumata K, Shao T, Wang G, Hatori A, Mori W, Yu Q, Hu K, Fujinaga M, Shao Y, Josephson L, Sun S, Zhang MR, Liang S. Synthesis and preliminary evaluation of 18F-labeled 1-(6,7-dimethyl-4-(methylamino)-1,3-dihydro-2H-pyrrolo[3,4-c]pyridin-2-yl)-2-(trans-2-(6-fluoropyridin-3-yl)cyclopropyl)ethan-1-one for imaging muscarinic acetylcholine receptor subtype 4. Tetrahedron Lett 2020. [DOI: 10.1016/j.tetlet.2020.152060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
31
|
Bakker G, Vingerhoets C, Bloemen OJN, Sahakian BJ, Booij J, Caan MWA, van Amelsvoort TAMJ. The muscarinic M 1 receptor modulates associative learning and memory in psychotic disorders. NEUROIMAGE-CLINICAL 2020; 27:102278. [PMID: 32563036 PMCID: PMC7305431 DOI: 10.1016/j.nicl.2020.102278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 11/25/2022]
Abstract
Psychosis characterised by different M1 sensitivity in learning and memory. Greater limbic-temporal hyperactivity in response to biperiden in psychosis. Hippocampal M1 binding predicted limbic-temporal hyperactivation underlying learning. M1 agonist may normalise functional response underlying learning & memory in psychosis.
Background Psychotic disorders are characterized by prominent deficits in associative learning and memory for which there are currently no effective treatments. Functional magnetic resonance imaging (fMRI) studies in psychotic disorders have identified deficits in fronto-temporal activation during associative learning and memory. The underlying pathology of these findings remains unclear. Postmortem data have suggested these deficits may be related to loss of muscarinic M1 receptor mediated signaling. This is supported by an in-vivo study showing improvements in these symptoms after treatment with the experimental M1/4 receptor agonist xanomeline. The current study tests whether reported deficits in fronto-temporal activation could be mediated by loss of M1 receptor signaling in psychotic disorders. Methods Twenty-six medication-free subjects diagnosed with a psychotic disorder and 29 age-, gender-, and IQ-matched healthy controls underwent two functional magnetic resonance imaging (fMRI) sessions, one under placebo and one under selective M1 antagonist biperiden, while performing the paired associated learning task. M1 binding potentials (BPND) were measured in the dorsolateral prefrontal cortex (DLPFC) and hippocampus using 123I-IDEX single photon emission computed tomography. Results In the subjects with psychotic disorders DLPFC hypoactivation was only found in the memory phase of the task. In both learning and memory phases of the task, M1 antagonism by biperiden elicited significantly greater hyperactivation of the parahippocampal gyrus and superior temporal gyrus in subjects with a psychotic disorders compared to controls. Greater hyperactivation of these areas after biperiden was associated with greater hippocampal M1 receptor binding during learning, with no association found with M1 receptor binding in the DLPFC. M1 receptor binding in the DLPFC was related to greater functional sensitivity to biperiden of the cingulate gyrus during the memory phase. Conclusion The current study is the first to show differences in M1 receptor mediated functional sensitivity between subjects with a psychotic disorder and controls during a paired associate learning and memory task. Results point to subjects with psychotic disorders having a loss of M1 receptor reserve in temporal-limbic areas.
Collapse
Affiliation(s)
- Geor Bakker
- Department of Psychiatry and Psychology, University of Maastricht, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Claudia Vingerhoets
- Department of Psychiatry and Psychology, University of Maastricht, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Oswald J N Bloemen
- Department of Psychiatry and Psychology, University of Maastricht, Maastricht, The Netherlands; GGZ Centraal, Center for Mental Health Care Innova, Amersfoort, The Netherlands
| | - Barbara J Sahakian
- Department of Psychiatry, University of Cambridge, Cambridgeshire, United Kingdom
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Matthan W A Caan
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centres, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
32
|
Walker LC, Lawrence AJ. Allosteric modulation of muscarinic receptors in alcohol and substance use disorders. FROM STRUCTURE TO CLINICAL DEVELOPMENT: ALLOSTERIC MODULATION OF G PROTEIN-COUPLED RECEPTORS 2020; 88:233-275. [DOI: 10.1016/bs.apha.2020.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
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: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar 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
|
34
|
Chopko TC, Han C, Gregro AR, Engers DW, Felts AS, Poslusney MS, Bollinger KA, Morrison RD, Bubser M, Lamsal A, Luscombe VB, Cho HP, Schnetz-Boutaud NC, Rodriguez AL, Chang S, Daniels JS, Stec DF, Niswender CM, Jones CK, Wood MR, Wood MW, Duggan ME, Brandon NJ, Conn PJ, Bridges TM, Lindsley CW, Melancon BJ. SAR inspired by aldehyde oxidase (AO) metabolism: Discovery of novel, CNS penetrant tricyclic M 4 PAMs. Bioorg Med Chem Lett 2019; 29:2224-2228. [PMID: 31248774 PMCID: PMC6690795 DOI: 10.1016/j.bmcl.2019.06.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 01/18/2023]
Abstract
This letter describes progress towards an M4 PAM preclinical candidate inspired by an unexpected aldehyde oxidase (AO) metabolite of a novel, CNS penetrant thieno[2,3-c]pyridine core to an equipotent, non-CNS penetrant thieno[2,3-c]pyrdin-7(6H)-one core. Medicinal chemistry design efforts yielded two novel tricyclic cores that enhanced M4 PAM potency, regained CNS penetration, displayed favorable DMPK properties and afforded robust in vivo efficacy in reversing amphetamine-induced hyperlocomotion in rats.
Collapse
Affiliation(s)
- Trevor C Chopko
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Changho Han
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Alison R Gregro
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Darren W Engers
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Andrew S Felts
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Mike S Poslusney
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Katrina A Bollinger
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Ryan D Morrison
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Michael Bubser
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Atin Lamsal
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Vincent B Luscombe
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Hyekyung P Cho
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Nathalie C Schnetz-Boutaud
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alice L Rodriguez
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sichen Chang
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - J Scott Daniels
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Donald F Stec
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Carrie K Jones
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael R Wood
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael W Wood
- AstraZeneca Neuroscience, IMED Biotech Unit, R&D, Boston, MA 02451, USA
| | - Mark E Duggan
- AstraZeneca Neuroscience, IMED Biotech Unit, R&D, Boston, MA 02451, USA
| | | | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Thomas M Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Bruce J Melancon
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
35
|
Teal LB, Gould RW, Felts AS, Jones CK. Selective allosteric modulation of muscarinic acetylcholine receptors for the treatment of schizophrenia and substance use disorders. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 86:153-196. [PMID: 31378251 DOI: 10.1016/bs.apha.2019.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Muscarinic acetylcholine receptor (mAChRs) subtypes represent exciting new targets for the treatment of schizophrenia and substance use disorder (SUD). Recent advances in the development of subtype-selective allosteric modulators have revealed promising effects in preclinical models targeting the different symptoms observed in schizophrenia and SUD. M1 PAMs display potential for addressing the negative and cognitive symptoms of schizophrenia, while M4 PAMs exhibit promise in treating preclinical models predictive of antipsychotic-like activity. In SUD, there is increasing support for modulation of mesocorticolimbic dopaminergic circuitry involved in SUD with selective M4 mAChR PAMs or M5 mAChR NAMs. Allosteric modulators of these mAChR subtypes have demonstrated efficacy in rodent models of cocaine and ethanol seeking, with indications that these ligand may also be useful for other substances of abuse, as well as in various stages in the cycle of addiction. Importantly, allosteric modulators of the different mAChR subtypes may provide viable treatment options, while conferring greater subtype specificity and corresponding enhanced therapeutic index than orthosteric muscarinic ligands and maintaining endogenous temporo-spatial ACh signaling. Overall, subtype specific mAChR allosteric modulators represent important novel therapeutic mechanisms for schizophrenia and SUD.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Robert W Gould
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Andrew S Felts
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.
| |
Collapse
|
36
|
Engers DW, Melancon BJ, Gregro AR, Bertron JL, Bollinger SR, Felts AS, Konkol LC, Wood MR, Bollinger KA, Luscombe VB, Rodriguez AL, Jones CK, Bubser M, Yohn SE, Wood MW, Brandon NJ, Dugan ME, Niswender CM, Conn PJ, Bridges TM, Lindsley CW. VU6005806/AZN-00016130, an advanced M 4 positive allosteric modulator (PAM) profiled as a potential preclinical development candidate. Bioorg Med Chem Lett 2019; 29:1714-1718. [PMID: 31113706 DOI: 10.1016/j.bmcl.2019.05.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/24/2022]
Abstract
This letter describes progress towards an M4 PAM preclinical candidate that resulted in the discovery of VU6005806/AZN-00016130. While the thieno[2,3-c]pyridazine core has been a consistent feature of key M4 PAMs, no work had previously been reported with respect to alternate functionality at the C3 position of the pyridazine ring. Here, we detail new chemistry and analogs that explored this region, and quickly led to VU6005806/AZN-00016130, which was profiled as a putative candidate. While, the β-amino carboxamide moiety engendered solubility limited absorption in higher species precluding advancement (or requiring extensive pharmaceutical sciences formulation), VU6005806/AZN-00016130 represents a new, high quality preclinical in vivo probe.
Collapse
Affiliation(s)
- Darren W Engers
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Bruce J Melancon
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Allison R Gregro
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jeanette L Bertron
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sean R Bollinger
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Andrew S Felts
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Leah C Konkol
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael R Wood
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Katrina A Bollinger
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Vincent B Luscombe
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alice L Rodriguez
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Carrie K Jones
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael Bubser
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Samantha E Yohn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael W Wood
- AstraZeneca Neuroscience, IMED Biotech Unit, R&D Boston, MA 02451, USA
| | | | - Mark E Dugan
- AstraZeneca Neuroscience, IMED Biotech Unit, R&D Boston, MA 02451, USA
| | - Colleen M Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Thomas M Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
37
|
Thorn CA, Moon J, Bourbonais CA, Harms J, Edgerton JR, Stark E, Steyn SJ, Butter CR, Lazzaro JT, O’Connor RE, Popiolek M. Striatal, Hippocampal, and Cortical Networks Are Differentially Responsive to the M4- and M1-Muscarinic Acetylcholine Receptor Mediated Effects of Xanomeline. ACS Chem Neurosci 2019; 10:1753-1764. [PMID: 30480428 DOI: 10.1021/acschemneuro.8b00625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Preclinical and clinical data suggest that muscarinic acetylcholine receptor activation may be therapeutically beneficial for the treatment of schizophrenia and Alzheimer's diseases. This is best exemplified by clinical observations with xanomeline, the efficacy of which is thought to be mediated through co-activation of the M1 and M4 muscarinic acetylcholine receptors (mAChRs). Here we examined the impact of treatment with xanomeline and compared it to the actions of selective M1 and M4 mAChR activators on in vivo intracellular signaling cascades in mice, including 3'-5'-cyclic adenosine monophosphate response element binding protein (CREB) phosphorylation and inositol phosphate-1 (IP1) accumulation in the striatum, hippocampus, and prefrontal cortex. We additionally assessed the effects of xanomeline on hippocampal electrophysiological signatures in rats using ex vivo recordings from CA1 (Cornu Ammonis 1) as well as in vivo hippocampal theta. As expected, xanomeline's effects across these readouts were consistent with activation of both M1 and M4 mAChRs; however, differences were observed across different brain regions, suggesting non-uniform activation of these receptor subtypes in the central nervous system. Interestingly, despite having nearly equal in vitro potency at the M1 and the M4 mAChRs, during in vivo assays xanomeline produced M4-like effects at significantly lower brain exposures than those at which M1-like effects were observed. Our results raise the possibility that clinical efficacy observed with xanomeline was driven, in part, through its non-uniform activation of mAChR subtypes in the central nervous system and, at lower doses, through preferential agonism of the M4 mAChR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - John T. Lazzaro
- Primary Pharmacology Group, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Rebecca E. O’Connor
- Primary Pharmacology Group, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | | |
Collapse
|
38
|
Poslusney MS, Salovich JM, Wood MR, Melancon BJ, Bollinger KA, Luscombe VB, Rodriguez AL, Engers DW, Bridges TM, Niswender CM, Conn PJ, Lindsley CW. Novel M 4 positive allosteric modulators derived from questioning the role and impact of a presumed intramolecular hydrogen-bonding motif in β-amino carboxamide-harboring ligands. Bioorg Med Chem Lett 2019; 29:362-366. [PMID: 30580918 PMCID: PMC6690489 DOI: 10.1016/j.bmcl.2018.12.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 12/25/2022]
Abstract
This letter describes a focused exercise to explore the role of the β-amino carboxamide moiety found in all of the first generation M4 PAMs and question if the NH2 group served solely to stabilize an intramolecular hydrogen bond (IMHB) and enforce planarity. To address this issue (and to potentially find a substitute for the β-amino carboxamide that engendered P-gp and contributed to solubility liabilities), we removed the NH2, generating des-amino congeners and surveyed other functional groups in the β-position. These modifications led to weak M4 PAMs with poor DMPK properties. Cyclization of the β-amino carboxamide moiety by virtue of a pyrazole ring re-enforced the IMHB, led to potent (and patented) M4 PAMs, many as potent as the classical bicyclic β-amino carboxamide analogs, but with significant CYP1A2 inhibition. Overall, this exercise indicated that the β-amino carboxamide moiety most likely facilitates an IMHB, and is essential for M4 PAM activity within classical bicyclic M4 PAM scaffolds.
Collapse
Affiliation(s)
- Michael S Poslusney
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - James M Salovich
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Michael R Wood
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Bruce J Melancon
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Katrina A Bollinger
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Vincent B Luscombe
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Alice L Rodriguez
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Darren W Engers
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Thomas M Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
39
|
Zhou D, Lv D, Wang Z, Zhang Y, Chen Z, Wang C. GLYX-13 Ameliorates Schizophrenia-Like Phenotype Induced by MK-801 in Mice: Role of Hippocampal NR2B and DISC1. Front Mol Neurosci 2018; 11:121. [PMID: 29695955 PMCID: PMC5904356 DOI: 10.3389/fnmol.2018.00121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/28/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Evidence supports that the hypofunction of N-methyl-D-aspartate receptor (NMDAR) and downregulation of disrupted-in-schizophrenia 1 (DISC1) contribute to the pathophysiology of schizophrenia. N-Methyl D-aspartate receptor subtype 2B (NR2B)-containing NMDAR are associated with cognitive dysfunction in schizophrenia. GLYX-13 is an NMDAR glycine-site functional partial agonist and cognitive enhancer that does not induce psychotomimetic side effects. However, it remains unclear whether NR2B plays a critical role in the GLYX-13-induced alleviation of schizophrenia-like behaviors in mice. Methods: The effect of GLYX-13 was tested by observing changes in locomotor activity, novel object recognition ability, and prepulse inhibition (PPI) induced by dizocilpine (known as MK-801) in mice. Lentivirus-mediated NR2B knockdown in the hippocampus was assessed to confirm the role of NR2B in GLYX-13 pathophysiology, using Western blots and immunohistochemistry. Results: The systemic administration of GLYX-13 (0.5 and 1 mg/kg, i.p.) ameliorates MK-801 (0.5 mg/kg, i.p.)-induced hyperlocomotion, deficits in memory, and PPI in mice. Additionally, GLYX-13 normalized the MK-801-induced alterations in signaling molecules, including NR2B and DISC1 in the hippocampus. Furthermore, we found that NR2B knockdown produced memory and PPI deficits without any changes in locomotor activity. Notably, DISC1 levels significantly decreased by NR2B knockdown. However, the effective dose of GLYX-13 did not alleviate the memory and PPI dysfunctions or downregulation of DISC1 induced by NR2B knockdown. Conclusion: Our results suggest GLYX-13 as a candidate for schizophrenia treatment, and NR2B and DISC1 in the hippocampus may account for the molecular mechanisms of GLYX-13.
Collapse
Affiliation(s)
- Dongsheng Zhou
- Ningbo Kangning Hospital, Ningbo, China.,Ningbo Key Laboratory of Behavioral Neuroscience, School of Medicine, Ningbo University, Ningbo, China
| | - Dan Lv
- Ningbo Key Laboratory of Behavioral Neuroscience, School of Medicine, Ningbo University, Ningbo, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China.,Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China
| | - Zhen Wang
- Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yanhua Zhang
- Ningbo Key Laboratory of Behavioral Neuroscience, School of Medicine, Ningbo University, Ningbo, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China.,Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China
| | | | - Chuang Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, School of Medicine, Ningbo University, Ningbo, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China.,Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
40
|
Effects of muscarinic M 1 and M 4 acetylcholine receptor stimulation on extinction and reinstatement of cocaine seeking in male mice, independent of extinction learning. Psychopharmacology (Berl) 2018; 235:815-827. [PMID: 29250738 PMCID: PMC6472894 DOI: 10.1007/s00213-017-4797-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/20/2017] [Indexed: 01/16/2023]
Abstract
RATIONALE Stimulating muscarinic M1/M4 receptors can blunt reinforcing and other effects of cocaine. A hallmark of addiction is continued drug seeking/craving after abstinence and relapse. OBJECTIVES We tested whether stimulating M1 and/or M4 receptors could facilitate extinction of cocaine seeking, and whether this was mediated via memory consolidation. METHODS Experimentally naïve C57BL/6J mice were allowed to acquire self-administration of intravenous cocaine (1 mg/kg/infusion) under a fixed-ratio 1 schedule of reinforcement. Then, saline was substituted for cocaine until responding extinguished to ≤30% of cocaine-reinforced responding. Immediately after each extinction session, mice received saline, the M1/M4 receptor-preferring agonist xanomeline, the M1 receptor-selective allosteric agonist VU0357017, the M4 receptor-selective positive allosteric modulator VU0152100, or VU0357017 + VU0152100. In additional experiments, xanomeline was administered delayed after the session or in the home cage before extinction training began. In the latter group, reinstatement of responding by a 10-mg/kg cocaine injection was also tested. RESULTS Stimulating M1 + M4 receptors significantly expedited extinction from 17.2 sessions to 8.3 using xanomeline or 7.8 using VU0357017 + VU0152100. VU0357017 alone and VU0152100 alone did not significantly modify rates of extinction (12.6 and 14.6 sessions). The effect of xanomeline was fully preserved when administered delayed after or unpaired from extinction sessions (7.5 and 6.4 sessions). Xanomeline-treated mice showed no cocaine-induced reinstatement. CONCLUSIONS These findings show that M1/M4 receptor stimulation can decrease cocaine seeking in mice. The effect lasted beyond treatment duration and was not dependent upon extinction learning. This suggests that M1/M4 receptor stimulation modulated or reversed some neurochemical effects of cocaine exposure.
Collapse
|