A kinetic view of GPCR allostery and biased agonism.

G-protein-coupled receptors (GPCRs) are one of the most tractable classes of drug targets. These dynamic proteins can adopt multiple active states that are linked to distinct functional outcomes. Such states can be differentially stabilized by ligands interacting with the endogenous agonist-binding orthosteric site and/or by ligands acting via spatially distinct allosteric sites, leading to the phenomena of 'biased agonism' or 'biased modulation'. These paradigms are having a major impact on modern drug discovery, but it is becoming increasingly apparent that 'kinetic context', at the level of both ligand-receptor and receptor-signal pathway kinetics, can have a profound impact on the observation and quantification of these phenomena. The concept of kinetic context thus represents an important new consideration that should be routinely incorporated into contemporary chemical biology and drug discovery studies of GPCR bias and allostery.

[1]  Leigh A. Stoddart,et al.  Allosteric interactions across native adenosine-A3 receptor homodimers: quantification using single-cell ligand-binding kinetics , 2011, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[2]  A. T. Nguyen,et al.  Extracellular Loop 2 of the Adenosine A1 Receptor Has a Key Role in Orthosteric Ligand Affinity and Agonist Efficacy , 2016, Molecular Pharmacology.

[3]  M. J. Chalmers,et al.  Ligand-dependent perturbation of the conformational ensemble for the GPCR β2 adrenergic receptor revealed by HDX. , 2011, Structure.

[4]  Arthur Christopoulos,et al.  Signalling bias in new drug discovery: detection, quantification and therapeutic impact , 2012, Nature Reviews Drug Discovery.

[5]  R. Lefkowitz,et al.  Beta-adrenergic receptors: evidence for negative cooperativity. , 1975, Biochemical and biophysical research communications.

[6]  B. Disse,et al.  Ba 679 BR, a novel long-acting anticholinergic bronchodilator. , 1993, Life sciences.

[7]  P. Strange,et al.  Co‐operativity in agonist binding at the D2 dopamine receptor: evidence from agonist dissociation kinetics , 2010, Journal of neurochemistry.

[8]  Terry Kenakin,et al.  Ligand-selective receptor conformations revisited: the promise and the problem. , 2003, Trends in pharmacological sciences.

[9]  S. Charlton,et al.  Elusive equilibrium: the challenge of interpreting receptor pharmacology using calcium assays , 2010, British journal of pharmacology.

[10]  S. Charlton,et al.  Long‐lasting target binding and rebinding as mechanisms to prolong in vivo drug action , 2010, British journal of pharmacology.

[11]  Ralf C. Kling,et al.  Functionally selective dopamine D2/D3 receptor agonists comprising an enyne moiety. , 2013, Journal of medicinal chemistry.

[12]  Robert J. Lefkowitz,et al.  β-Adrenergic receptors: Evidence for negative cooperativity , 1975 .

[13]  P. Seeman Targeting the dopamine D2 receptor in schizophrenia , 2006, Expert opinion on therapeutic targets.

[14]  A. Christopoulos,et al.  Structure-activity relationships of privileged structures lead to the discovery of novel biased ligands at the dopamine D₂ receptor. , 2014, Journal of medicinal chemistry.

[15]  M. Burghammer,et al.  Crystal structure of the human β2 adrenergic G-protein-coupled receptor , 2007, Nature.

[16]  P. Leff,et al.  The two-state model of receptor activation. , 1995, Trends in pharmacological sciences.

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

[18]  A. IJzerman,et al.  Agonists for the adenosine A1 receptor with tunable residence time. A Case for nonribose 4-amino-6-aryl-5-cyano-2-thiopyrimidines. , 2014, Journal of medicinal chemistry.

[19]  A. IJzerman,et al.  Functional efficacy of adenosine A2A receptor agonists is positively correlated to their receptor residence time , 2012, British journal of pharmacology.

[20]  N. Birdsall,et al.  Modification of the binding properties of muscarinic receptors by gallamine. , 1983, Molecular pharmacology.

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

[22]  R. Stevens,et al.  Structure of an Agonist-Bound Human A2A Adenosine Receptor , 2011, Science.

[23]  R. Lefkowitz,et al.  A ternary complex model explains the agonist-specific binding properties of the adenylate cyclase-coupled beta-adrenergic receptor. , 1980, The Journal of biological chemistry.

[24]  Stephen M. Husbands,et al.  Structural insights into μ-opioid receptor activation , 2015, Nature.

[25]  P. Gmeiner,et al.  Histidine 6.55 Is a Major Determinant of Ligand-Biased Signaling in Dopamine D2L Receptor , 2011, Molecular Pharmacology.

[26]  Arthur Christopoulos,et al.  Emerging paradigms in GPCR allostery: implications for drug discovery , 2013, Nature Reviews Drug Discovery.

[27]  J. Violin,et al.  Biased ligands: pathway validation for novel GPCR therapeutics. , 2014, Current opinion in pharmacology.

[28]  Sudarshan Rajagopal,et al.  Quantifying Ligand Bias at Seven-Transmembrane Receptors , 2011, Molecular Pharmacology.

[29]  J. Black,et al.  An operational model of pharmacological agonism: the effect of E/[A] curve shape on agonist dissociation constant estimation , 1985, British journal of pharmacology.

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

[31]  P Ghanouni,et al.  Functionally Different Agonists Induce Distinct Conformations in the G Protein Coupling Domain of the β2Adrenergic Receptor* , 2001, The Journal of Biological Chemistry.

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

[33]  C. Altenbach,et al.  High-resolution distance mapping in rhodopsin reveals the pattern of helix movement due to activation , 2008, Proceedings of the National Academy of Sciences.

[34]  A. IJzerman,et al.  Allosteric modulation of A(2A) adenosine receptors by amiloride analogues and sodium ions. , 2000, Biochemical pharmacology.

[35]  R. Lefkowitz,et al.  A mutation-induced activated state of the beta 2-adrenergic receptor. Extending the ternary complex model. , 1993, The Journal of biological chemistry.

[36]  Jonathan R. Tomshine,et al.  Conformational biosensors reveal GPCR signalling from endosomes , 2013, Nature.

[37]  D. E. Nichols,et al.  Crystal Structure of an LSD-Bound Human Serotonin Receptor , 2017, Cell.

[38]  A. IJzerman,et al.  Structure-kinetics relationships of Capadenoson derivatives as adenosine A1 receptor agonists. , 2015, European journal of medicinal chemistry.

[39]  Adriaan P IJzerman,et al.  Drug‐Target Residence Time—A Case for G Protein‐Coupled Receptors , 2014, Medicinal research reviews.

[40]  Jennifer J Linderman,et al.  Modeling of G-protein-coupled Receptor Signaling Pathways* , 2009, Journal of Biological Chemistry.

[41]  Arthur Christopoulos,et al.  Biased Agonism at G Protein‐Coupled Receptors: The Promise and the Challenges—A Medicinal Chemistry Perspective , 2014, Medicinal research reviews.

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

[43]  T. Kenakin,et al.  G Protein-Coupled Receptor Allosterism and Complexing , 2002, Pharmacological Reviews.

[44]  S. Nuber,et al.  β-Arrestin biosensors reveal a rapid, receptor-dependent activation/deactivation cycle , 2016, Nature.

[45]  P. Sexton,et al.  Glucagon-like peptide-1 receptor dimerization differentially regulates agonist signaling but does not affect small molecule allostery , 2012, Proceedings of the National Academy of Sciences.

[46]  P. Sexton,et al.  Ligand-Dependent Modulation of G Protein Conformation Alters Drug Efficacy , 2016, Cell.

[47]  Arthur Christopoulos,et al.  A simple method for quantifying functional selectivity and agonist bias. , 2012, ACS chemical neuroscience.

[48]  O. Nosjean,et al.  Prolonged Calcitonin Receptor Signaling by Salmon, but Not Human Calcitonin, Reveals Ligand Bias , 2014, PloS one.

[49]  J. Fung,et al.  FRET-based measurement of GPCR conformational changes. , 2009, Methods in molecular biology.

[50]  Ryan T. Strachan,et al.  Divergent Transducer-specific Molecular Efficacies Generate Biased Agonism at a G Protein-coupled Receptor (GPCR)* , 2014, The Journal of Biological Chemistry.

[51]  Arthur Christopoulos,et al.  Endogenous Allosteric Modulators of G Protein–Coupled Receptors , 2015, The Journal of Pharmacology and Experimental Therapeutics.

[52]  J. Violin,et al.  Structure-activity relationships and discovery of a G protein biased μ opioid receptor ligand, [(3-methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro-[4.5]decan-9-yl]ethyl})amine (TRV130), for the treatment of acute severe pain. , 2013, Journal of medicinal chemistry.

[53]  P. Sexton,et al.  Application of a Kinetic Model to the Apparently Complex Behavior of Negative and Positive Allosteric Modulators of Muscarinic Acetylcholine Receptors , 2004, Journal of Pharmacology and Experimental Therapeutics.

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

[55]  W. Paton,et al.  A theory of drug action based on the rate of drug-receptor combination , 1961, Proceedings of the Royal Society of London. Series B. Biological Sciences.

[56]  S. Rasmussen,et al.  Allosteric coupling from G protein to the agonist binding pocket in GPCRs , 2016, Nature.

[57]  R. Stevens,et al.  The 2.6 Angstrom Crystal Structure of a Human A2A Adenosine Receptor Bound to an Antagonist , 2008, Science.

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

[59]  S. Swillens,et al.  Glycoprotein hormone receptors: link between receptor homodimerization and negative cooperativity , 2005, The EMBO journal.

[60]  J. Fergus,et al.  Allosteric enhancement of adenosine A1 receptor binding and function by 2-amino-3-benzoylthiophenes. , 1990, Molecular pharmacology.

[61]  S. Rasmussen,et al.  Crystal Structure of the β2Adrenergic Receptor-Gs protein complex , 2011, Nature.

[62]  J. Changeux,et al.  International Union of Basic and Clinical Pharmacology. XC. Multisite Pharmacology: Recommendations for the Nomenclature of Receptor Allosterism and Allosteric Ligands , 2014, Pharmacological Reviews.

[63]  Hualiang Jiang,et al.  Structural Basis for Molecular Recognition at Serotonin Receptors , 2013, Science.

[64]  Eric Trinquet,et al.  Structural insights into biased G protein-coupled receptor signaling revealed by fluorescence spectroscopy , 2012, Proceedings of the National Academy of Sciences.

[65]  A. Christopoulos,et al.  A structure-activity analysis of biased agonism at the dopamine D2 receptor. , 2013, Journal of medicinal chemistry.

[66]  Weston B. Struwe,et al.  The role of interfacial lipids in stabilizing membrane protein oligomers , 2017, Nature.

[67]  T. Kenakin,et al.  Analytical pharmacology: the impact of numbers on pharmacology. , 2011, Trends in pharmacological sciences.

[68]  J. Vilardaga ENDOSOMAL GENERATION OF cAMP in GPCR SIGNALING , 2014, Nature chemical biology.

[69]  P. Sexton,et al.  Allosteric modulation of G protein-coupled receptors: A pharmacological perspective , 2011, Neuropharmacology.

[70]  M. Dowling,et al.  Exploring the Mechanism of Agonist Efficacy: A Relationship between Efficacy and Agonist Dissociation Rate at the Muscarinic M3 Receptor , 2009, Molecular Pharmacology.

[71]  Y. Peterson,et al.  The conformational signature of arrestin3 predicts its trafficking and signaling functions , 2016, Nature.

[72]  Kunhong Xiao,et al.  Multiple ligand-specific conformations of the β2-adrenergic receptor. , 2011, Nature chemical biology.

[73]  P. Sexton,et al.  A Monod-Wyman-Changeux Mechanism Can Explain G Protein-coupled Receptor (GPCR) Allosteric Modulation* , 2011, The Journal of Biological Chemistry.

[74]  I. Gaidarov,et al.  Kinetics of 5-HT2B Receptor Signaling: Profound Agonist-Dependent Effects on Signaling Onset and Duration , 2013, The Journal of Pharmacology and Experimental Therapeutics.

[75]  P. Sexton,et al.  Allosteric GPCR modulators: taking advantage of permissive receptor pharmacology. , 2007, Trends in pharmacological sciences.

[76]  Tamara L. Kinzer-Ursem,et al.  Both Ligand- and Cell-Specific Parameters Control Ligand Agonism in a Kinetic Model of G Protein–Coupled Receptor Signaling , 2007, PLoS Comput. Biol..

[77]  Albert C. Pan,et al.  Pathway and mechanism of drug binding to G-protein-coupled receptors , 2011, Proceedings of the National Academy of Sciences.

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

[79]  R. Huber,et al.  Structure–kinetic relationship study of CDK8/CycC specific compounds , 2013, Proceedings of the National Academy of Sciences.

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

[81]  G. Cottrell,et al.  Endosomes: A legitimate platform for the signaling train , 2009, Proceedings of the National Academy of Sciences.

[82]  R. Stevens,et al.  Structural Features for Functional Selectivity at Serotonin Receptors , 2013, Science.

[83]  Kurt Wüthrich,et al.  Biased Signaling Pathways in β2-Adrenergic Receptor Characterized by 19F-NMR , 2012, Science.

[84]  F. Ehlert,et al.  Estimation of the affinities of allosteric ligands using radioligand binding and pharmacological null methods. , 1988, Molecular pharmacology.

[85]  F. Ehlert Analysis of Allosterism in Functional Assays , 2005, Journal of Pharmacology and Experimental Therapeutics.

[86]  K. Jacobson,et al.  Structure-Activity Analysis of Biased Agonism at the Human Adenosine A3 Receptor , 2016, Molecular Pharmacology.

[87]  Arthur Christopoulos,et al.  The role of kinetic context in apparent biased agonism at GPCRs , 2016, Nature Communications.

[88]  S. Charlton,et al.  Long Receptor Residence Time of C26 Contributes to Super Agonist Activity at the Human β2 Adrenoceptor , 2016, Molecular Pharmacology.

[89]  R. Copeland The drug–target residence time model: a 10-year retrospective , 2015, Nature Reviews Drug Discovery.

[90]  P. Sexton,et al.  Molecular Mechanisms of Action and In Vivo Validation of an M4 Muscarinic Acetylcholine Receptor Allosteric Modulator with Potential Antipsychotic Properties , 2010, Neuropsychopharmacology.

[91]  L. Mahan,et al.  Correction to “The Kinetics of Competitive Radioligand Binding Predicted by the Law of Mass Action” , 2014, Molecular Pharmacology.

[92]  J. Violin,et al.  Biased ligands at G-protein-coupled receptors: promise and progress. , 2014, Trends in pharmacological sciences.

[93]  D. Hall,et al.  Modeling the functional effects of allosteric modulators at pharmacological receptors: an extension of the two-state model of receptor activation. , 2000, Molecular pharmacology.

[94]  S. Lazareno,et al.  Analogs of WIN 62,577 define a second allosteric site on muscarinic receptors. , 2002, Molecular pharmacology.

[95]  Robert J. Lefkowitz,et al.  Transduction of Receptor Signals by ß-Arrestins , 2005, Science.

[96]  Aashish Manglik,et al.  Propagation of conformational changes during μ-opioid receptor activation , 2015, Nature.

[97]  J. Proska,et al.  Mechanisms of steric and cooperative actions of alcuronium on cardiac muscarinic acetylcholine receptors. , 1994, Molecular pharmacology.