Structure-based discovery of selective positive allosteric modulators of antagonists for the M2 muscarinic acetylcholine receptor

Significance The orthosteric binding sites of the five muscarinic acetylcholine receptor (mAChR) subtypes are highly conserved, making the development of selective antagonists challenging. The allosteric sites of these receptors are more variable, allowing one to imagine allosteric modulators that confer subtype selectivity, which would reduce the major off-target effects of muscarinic antagonists. Accordingly, a large library docking campaign was prosecuted seeking unique positive allosteric modulators (PAMs) for antagonists, ultimately revealing a PAM that substantially potentiates antagonist binding leading to subtype selectivity at the M2 mAChR. This study supports the feasibility of discovering PAMs that can convert an armamentarium of potent but nonselective G-protein–coupled receptor (GPCR) antagonist drugs into subtype-selective reagents. Subtype-selective antagonists for muscarinic acetylcholine receptors (mAChRs) have long been elusive, owing to the highly conserved orthosteric binding site. However, allosteric sites of these receptors are less conserved, motivating the search for allosteric ligands that modulate agonists or antagonists to confer subtype selectivity. Accordingly, a 4.6 million-molecule library was docked against the structure of the prototypical M2 mAChR, seeking molecules that specifically stabilized antagonist binding. This led us to identify a positive allosteric modulator (PAM) that potentiated the antagonist N-methyl scopolamine (NMS). Structure-based optimization led to compound ’628, which enhanced binding of NMS, and the drug scopolamine itself, with a cooperativity factor (α) of 5.5 and a KB of 1.1 μM, while sparing the endogenous agonist acetylcholine. NMR spectral changes determined for methionine residues reflected changes in the allosteric network. Moreover, ’628 slowed the dissociation rate of NMS from the M2 mAChR by 50-fold, an effect not observed at the other four mAChR subtypes. The specific PAM effect of ’628 on NMS antagonism was conserved in functional assays, including agonist stimulation of [35S]GTPγS binding and ERK 1/2 phosphorylation. Importantly, the selective allostery between ’628 and NMS was retained in membranes from adult rat hypothalamus and in neonatal rat cardiomyocytes, supporting the physiological relevance of this PAM/antagonist approach. This study supports the feasibility of discovering PAMs that confer subtype selectivity to antagonists; molecules like ’628 can convert an armamentarium of potent but nonselective GPCR antagonist drugs into subtype-selective reagents, thus reducing their off-target effects.

[1]  Ruben Abagyan,et al.  Towards a structural understanding of allosteric drugs at the human calcium-sensing receptor , 2016, Cell Research.

[2]  Peter Kolb,et al.  Structure-based discovery of β2-adrenergic receptor ligands , 2009, Proceedings of the National Academy of Sciences.

[3]  Ali Jazayeri,et al.  Intracellular allosteric antagonism of the CCR9 receptor , 2016, Nature.

[4]  P. Sexton,et al.  Probe Dependence in the Allosteric Modulation of a G Protein-Coupled Receptor: Implications for Detection and Validation of Allosteric Ligand Effects , 2012, Molecular Pharmacology.

[5]  A. Grover Use of Allosteric Targets in the Discovery of Safer Drugs , 2013, Medical Principles and Practice.

[6]  N. Birdsall,et al.  International Union of Pharmacology. XVII. Classification of muscarinic acetylcholine receptors. , 1998, Pharmacological reviews.

[7]  Jesús Giraldo,et al.  Helix 3 acts as a conformational hinge in Class A GPCR activation: An analysis of interhelical interaction energies in crystal structures. , 2015, Journal of structural biology.

[8]  F. Mitchelson,et al.  THE INHIBITORY EFFECT OF GALLAMINE ON MUSCARINIC RECEPTORS , 1976, British journal of pharmacology.

[9]  Jan Jakubík,et al.  Allosteric Modulation of Muscarinic Acetylcholine Receptors , 2010, Pharmaceuticals.

[10]  H. Colecraft,et al.  Signaling Mechanisms Underlying Muscarinic Receptor-mediated Increase in Contraction Rate in Cultured Heart Cells* , 1998, The Journal of Biological Chemistry.

[11]  Gareth J Waldron,et al.  Reducing safety-related drug attrition: the use of in vitro pharmacological profiling , 2012, Nature Reviews Drug Discovery.

[12]  Tudor I. Oprea,et al.  A comprehensive map of molecular drug targets , 2016, Nature Reviews Drug Discovery.

[13]  Tudor I. Oprea,et al.  Is There a Difference between Leads and Drugs? A Historical Perspective , 2001, J. Chem. Inf. Comput. Sci..

[14]  Gregory W. Kauffman,et al.  Discovery of the Potent and Selective M1 PAM-Agonist N-[(3R,4S)-3-Hydroxytetrahydro-2H-pyran-4-yl]-5-methyl-4-[4-(1,3-thiazol-4-yl)benzyl]pyridine-2-carboxamide (PF-06767832): Evaluation of Efficacy and Cholinergic Side Effects. , 2016, Journal of medicinal chemistry.

[15]  S. P. Andrews,et al.  Extra-helical binding site of a glucagon receptor antagonist , 2016, Nature.

[16]  Arthur Christopoulos,et al.  Novel Allosteric Modulators of G Protein-coupled Receptors* , 2015, The Journal of Biological Chemistry.

[17]  Brian K. Shoichet,et al.  Ligand Pose and Orientational Sampling in Molecular Docking , 2013, PloS one.

[18]  Ryan G. Coleman,et al.  ZINC: A Free Tool to Discover Chemistry for Biology , 2012, J. Chem. Inf. Model..

[19]  I. Kuntz,et al.  Automated docking with grid‐based energy evaluation , 1992 .

[20]  Henry Lin,et al.  Structure-based discovery of opioid analgesics with reduced side effects , 2016, Nature.

[21]  J M Blaney,et al.  A geometric approach to macromolecule-ligand interactions. , 1982, Journal of molecular biology.

[22]  P. Conn,et al.  Allosteric Modulation of GPCRs: New Insights and Potential Utility for Treatment of Schizophrenia and Other CNS Disorders , 2017, Neuron.

[23]  J. Wess,et al.  M1 muscarinic allosteric modulators slow prion neurodegeneration and restore memory loss , 2016, The Journal of clinical investigation.

[24]  Jonathan A. Javitch,et al.  Discovery of a Novel Selective Kappa-Opioid Receptor Agonist Using Crystal Structure-Based Virtual Screening , 2013, J. Chem. Inf. Model..

[25]  B. Shoichet,et al.  A common mechanism underlying promiscuous inhibitors from virtual and high-throughput screening. , 2002, Journal of medicinal chemistry.

[26]  Brian K. Shoichet,et al.  ZINC - A Free Database of Commercially Available Compounds for Virtual Screening , 2005, J. Chem. Inf. Model..

[27]  Peter A. Kollman,et al.  AMBER, a package of computer programs for applying molecular mechanics, normal mode analysis, molecular dynamics and free energy calculations to simulate the structural and energetic properties of molecules , 1995 .

[28]  E. El-Fakahany,et al.  The allosteric binding profile of himbacine: a comparison with other cardioselective muscarinic antagonists. , 1990, European journal of pharmacology.

[29]  John J. Irwin,et al.  ZINC 15 – Ligand Discovery for Everyone , 2015, J. Chem. Inf. Model..

[30]  Nathan Robertson,et al.  Article pubs.acs.org/jmc Identification of Novel Adenosine A 2A Receptor Antagonists by Virtual Screening , 2022 .

[31]  R. Duman,et al.  Emerging treatment mechanisms for depression: focus on glutamate and synaptic plasticity. , 2016, Drug discovery today.

[32]  Ruben Abagyan,et al.  Structure of CC Chemokine Receptor 2 with Orthosteric and Allosteric Antagonists , 2016, Nature.

[33]  A. Christopoulos,et al.  Characterization of the Novel Positive Allosteric Modulator, LY2119620, at the Muscarinic M2 and M4 Receptors , 2014, Molecular Pharmacology.

[34]  Hualiang Jiang,et al.  Two disparate ligand-binding sites in the human P2Y1 receptor , 2015, Nature.

[35]  C. Lindsley,et al.  Development of a Highly Potent, Novel M5 Positive Allosteric Modulator (PAM) Demonstrating CNS Exposure: 1-((1H-Indazol-5-yl)sulfoneyl)-N-ethyl-N-(2-(trifluoromethyl)benzyl)piperidine-4-carboxamide (ML380) , 2014, Journal of medicinal chemistry.

[36]  J. Wess,et al.  Activation and allosteric modulation of a muscarinic acetylcholine receptor , 2013, Nature.

[37]  J. Andrew McCammon,et al.  Accelerated structure-based design of chemically diverse allosteric modulators of a muscarinic G protein-coupled receptor , 2016, Proceedings of the National Academy of Sciences.

[38]  Gregory D. Hawkins,et al.  Extension of the platform of applicability of the SM5.42R universal solvation model , 1999 .

[39]  Maria F. Sassano,et al.  Conformation Guides Molecular Efficacy in Docking Screens of Activated β-2 Adrenergic G Protein Coupled Receptor , 2013, ACS chemical biology.

[40]  D. Price,et al.  Is there a rationale and role for long-acting anticholinergic bronchodilators in asthma? , 2014, npj Primary Care Respiratory Medicine.

[41]  M. Chancellor,et al.  Antimuscarinics for the treatment of overactive bladder: understanding the role of muscarinic subtype selectivity , 2011, International Urogynecology Journal.

[42]  B. Shoichet,et al.  Hierarchical docking of databases of multiple ligand conformations. , 2005, Current topics in medicinal chemistry.

[43]  K. Sharp,et al.  Electrostatic contributions to heat capacity changes of DNA-ligand binding. , 1998, Biophysical journal.

[44]  Patrik Johansson,et al.  Structural insight into allosteric modulation of protease-activated receptor 2 , 2017, Nature.

[45]  Jing Liu,et al.  In silico design of novel probes for the atypical opioid receptor MRGPRX2 , 2016, Nature chemical biology.

[46]  Andreas Plückthun,et al.  Ligand Discovery for a Peptide-Binding GPCR by Structure-Based Screening of Fragment- and Lead-Like Chemical Libraries. , 2017, ACS chemical biology.

[47]  Michael M. Mysinger,et al.  Automated Docking Screens: A Feasibility Study , 2009, Journal of medicinal chemistry.

[48]  Arthur Christopoulos,et al.  Crystal structures of the M1 and M4 muscarinic acetylcholine receptors , 2016, Nature.

[49]  J. Irwin,et al.  An Aggregation Advisor for Ligand Discovery. , 2015, Journal of medicinal chemistry.

[50]  K. Mohr,et al.  Interactions of orthosteric and allosteric ligands with [3H]dimethyl-W84 at the common allosteric site of muscarinic M2 receptors. , 2003, Molecular pharmacology.

[51]  R. Cumming Drugs affecting the autonomic nervous system. , 1951, Arizona medicine.

[52]  Chris de Graaf,et al.  Function-specific virtual screening for GPCR ligands using a combined scoring method , 2016, Scientific Reports.

[53]  Brian K. Shoichet,et al.  Structure-Based Discovery of A2A Adenosine Receptor Ligands , 2010, Journal of medicinal chemistry.

[54]  J. Richardson,et al.  Asparagine and glutamine: using hydrogen atom contacts in the choice of side-chain amide orientation. , 1999, Journal of molecular biology.

[55]  H. Hasselmann Scopolamine and depression: a role for muscarinic antagonism? , 2014, CNS & neurological disorders drug targets.

[56]  Albert C. Pan,et al.  Structural basis for modulation of a G-protein-coupled receptor by allosteric drugs , 2013, Nature.

[57]  Albert C. Pan,et al.  Structure and Dynamics of the M3 Muscarinic Acetylcholine Receptor , 2012, Nature.

[58]  R. Read,et al.  Decoding Corticotropin-Releasing Factor Receptor Type 1 Crystal 
Structures , 2017, Current molecular pharmacology.

[59]  K. Andersson Antimuscarinics for treatment of overactive bladder , 2004, The Lancet Neurology.

[60]  O. Wassermann,et al.  Inhibition of the actions of carbachol and DFP on guinea pig isolated atria by alkane-bis-ammonium compounds. , 1969, European journal of pharmacology.

[61]  J. Ellis,et al.  Site-directed mutagenesis reveals two epitopes involved in the subtype selectivity of the allosteric interactions of gallamine at muscarinic acetylcholine receptors. , 1999, Molecular pharmacology.

[62]  Xin Chen,et al.  Allosteric ligands for the pharmacologically dark receptors GPR68 and GPR65 , 2015, Nature.

[63]  Sujata Sharma,et al.  Structural basis for the cooperative allosteric activation of the free fatty acid receptor GPR40 , 2017, Nature Structural &Molecular Biology.

[64]  A. Kruse,et al.  Structure of the human M2 muscarinic acetylcholine receptor bound to an antagonist , 2011, Nature.

[65]  J. Irwin,et al.  Docking Screens for Novel Ligands Conferring New Biology. , 2016, Journal of medicinal chemistry.

[66]  P. Sexton,et al.  Positive and negative allosteric modulators promote biased signaling at the calcium-sensing receptor. , 2012, Endocrinology.

[67]  M. Yamada,et al.  Regional brain uptake of the muscarinic ligand, [18F]FP-TZTP, is greatly decreased in M2 receptor knockout mice but not in M1, M3 and M4 receptor knockout mice , 2003, Neuropharmacology.

[68]  Brian K. Shoichet,et al.  Rapid Context-Dependent Ligand Desolvation in Molecular Docking , 2010, J. Chem. Inf. Model..

[69]  Michael J. Keiser,et al.  Large Scale Prediction and Testing of Drug Activity on Side-Effect Targets , 2012, Nature.

[70]  Kim A. Sharp,et al.  Polyelectrolyte electrostatics: Salt dependence, entropic, and enthalpic contributions to free energy in the nonlinear Poisson–Boltzmann model , 1995 .

[71]  B. Shoichet,et al.  Colloidal aggregation and the in vitro activity of traditional Chinese medicines. , 2015, ACS chemical biology.

[72]  Arthur Christopoulos,et al.  Advances in G Protein-Coupled Receptor Allostery: From Function to Structure , 2014, Molecular Pharmacology.

[73]  S. Grzesiek,et al.  NMRPipe: A multidimensional spectral processing system based on UNIX pipes , 1995, Journal of biomolecular NMR.

[74]  B. Matthews,et al.  A model binding site for testing scoring functions in molecular docking. , 2002, Journal of molecular biology.

[75]  R. E. Higgs,et al.  M1 and M2 Muscarinic Receptor Subtypes Regulate Antidepressant-Like Effects of the Rapidly Acting Antidepressant Scopolamine , 2014, The Journal of Pharmacology and Experimental Therapeutics.

[76]  Arthur Christopoulos,et al.  A kinetic view of GPCR allostery and biased agonism. , 2017, Nature chemical biology.

[77]  J. Kavanagh,et al.  Anticholinergic activity in the nervous system: Consequences for visuomotor function , 2017, Physiology & Behavior.

[78]  A. W. Schüttelkopf,et al.  PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. , 2004, Acta crystallographica. Section D, Biological crystallography.

[79]  F. Schmäl,et al.  Neuronal Mechanisms and the Treatment of Motion Sickness , 2013, Pharmacology.