Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Discovery of new GPCR ligands to illuminate new biology

Abstract

Although a plurality of drugs target G-protein-coupled receptors (GPCRs), most have emerged from classical medicinal chemistry and pharmacology programs and resemble one another structurally and functionally. Though effective, these drugs are often promiscuous. With the realization that GPCRs signal via multiple pathways, and with the emergence of crystal structures for this family of proteins, there is an opportunity to target GPCRs with new chemotypes and confer new signaling modalities. We consider structure-based and physical screening methods that have led to the discovery of new reagents, focusing particularly on the former. We illustrate their use against previously untargeted or orphan GPCRs, against allosteric sites, and against classical orthosteric sites that selectively activate one downstream pathway over others. The ligands that emerge are often chemically novel, which can lead to new biological effects.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: GPCR drugs as a percentage of all drugs, by decade of introduction.
Figure 2: GPCRs may activate multiple downstream signaling pathways: role of biased signaling.
Figure 3: Multiple allosteric sites for GPCRs.
Figure 4: Physical and docking screens to deorphanize MRGPRX2.
Figure 5: Novel biased agonists for the μ-opioid receptor.

References

  1. Dixon, R.A. et al. Cloning of the gene and cDNA for mammalian β-adrenergic receptor and homology with rhodopsin. Nature 321, 75–79 (1986). This paper closed the curtains on classical pharmacology, ushering in its molecular era.

    Article  CAS  PubMed  Google Scholar 

  2. Mason, J.S., Bortolato, A., Congreve, M. & Marshall, F.H. New insights from structural biology into the druggability of G protein-coupled receptors. Trends Pharmacol. Sci. 33, 249–260 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Roth, B.L., Sheffler, D.J. & Kroeze, W.K. Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophrenia. Nat. Rev. Drug Discov. 3, 353–359 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Hopkins, A.L. Network pharmacology: the next paradigm in drug discovery. Nat. Chem. Biol. 4, 682–690 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. DeWire, S.M., Ahn, S., Lefkowitz, R.J. & Shenoy, S.K. β-arrestins and cell signaling. Annu. Rev. Physiol. 69, 483–510 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Rosenbaum, D.M. et al. GPCR engineering yields high-resolution structural insights into beta2-adrenergic receptor function. Science 318, 1266–1273 (2007). This paper begins GPCR pharmacology and ligand discovery at atomic resolution.

    Article  CAS  PubMed  Google Scholar 

  7. Kenakin, T.P. Biased signalling and allosteric machines: new vistas and challenges for drug discovery. Br. J. Pharmacol. 165, 1659–1669 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Latorraca, N.R., Venkatakrishnan, A.J. & Dror, R.O. GPCR dynamics: structures in motion. Chem. Rev. 117, 139–155 (2017). A seminal review on GPCR molecular dynamics simulations and what we can and cannot infer from them.

    Article  CAS  PubMed  Google Scholar 

  9. Irwin, J.J. & Shoichet, B.K. Docking screens for novel ligands conferring new biology. J. Med. Chem. 59, 4103–4120 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hert, J., Irwin, J.J., Laggner, C., Keiser, M.J. & Shoichet, B.K. Quantifying biogenic bias in screening libraries. Nat. Chem. Biol. 5, 479–483 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Powers, R.A., Morandi, F. & Shoichet, B.K. Structure-based discovery of a novel, noncovalent inhibitor of AmpC β-lactamase. Structure 10, 1013–1023 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Doman, T.N. et al. Molecular docking and high-throughput screening for novel inhibitors of protein tyrosine phosphatase-1B. J. Med. Chem. 45, 2213–2221 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Grüneberg, S., Wendt, B. & Klebe, G. Subnanomolar inhibitors from computer screening: a model study using human carbonic anhydrase II. Angew. Chem. Int. Edn Engl. 40, 389–393 (2001).

    Article  Google Scholar 

  14. Evers, A. & Klebe, G. Ligand-supported homology modeling of g-protein-coupled receptor sites: models sufficient for successful virtual screening. Angew. Chem. Int. Edn Engl. 43, 248–251 (2004).

    Article  CAS  Google Scholar 

  15. Schapira, M., Abagyan, R. & Totrov, M. Nuclear hormone receptor targeted virtual screening. J. Med. Chem. 46, 3045–3059 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Hermann, J.C. et al. Structure-based activity prediction for an enzyme of unknown function. Nature 448, 775–779 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Teotico, D.G. et al. Docking for fragment inhibitors of AmpC β-lactamase. Proc. Natl. Acad. Sci. USA 106, 7455–7460 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen, Y. & Shoichet, B.K. Molecular docking and ligand specificity in fragment-based inhibitor discovery. Nat. Chem. Biol. 5, 358–364 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rodríguez, D., Ranganathan, A. & Carlsson, J. Discovery of GPCR ligands by molecular docking screening: novel opportunities provided by crystal structures. Curr. Top. Med. Chem. 15, 2484–2503 (2015).

    Article  PubMed  CAS  Google Scholar 

  20. Wootten, D., Christopoulos, A. & Sexton, P.M. Emerging paradigms in GPCR allostery: implications for drug discovery. Nat. Rev. Drug Discov. 12, 630–644 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Neubig, R.R., Spedding, M., Kenakin, T. & Christopoulos, A. International union of pharmacology committee on receptor nomenclature and drug classification. XXXVIII. Update on terms and symbols in quantitative pharmacology. Pharmacol. Rev. 55, 597–606 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Pert, C.B., Pasternak, G. & Snyder, S.H. Opiate agonists and antagonists discriminated by receptor binding in brain. Science 182, 1359–1361 (1973).

    Article  CAS  PubMed  Google Scholar 

  23. Fenalti, G. et al. Molecular control of δ-opioid receptor signalling. Nature 506, 191–196 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Liu, W. et al. Structural basis for allosteric regulation of GPCRs by sodium ions. Science 337, 232–236 (2012). The first direct structural evidence for a GPCR allosteric modulator.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Garcia-Barrantes, P.M. et al. Lead optimization of the VU0486321 series of mGlu1 PAMs. Part 1: SAR of modifications to the central aryl core. Bioorg. Med. Chem. Lett. 25, 5107–5110 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, C. et al. Structure of the human smoothened receptor bound to an antitumour agent. Nature 497, 338–343 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kruse, A.C. et al. Structure and dynamics of the M3 muscarinic acetylcholine receptor. Nature 482, 552–556 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Thal, D.M. et al. Crystal structures of the M1 and M4 muscarinic acetylcholine receptors. Nature 531, 335–340 (2016). From the structures of two muscarinic receptors, the difficulties in generating selective orthosteric ligands for the family are highlighted.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nickols, H.H. & Conn, P.J. Development of allosteric modulators of GPCRs for treatment of CNS disorders. Neurobiol. Dis. 61, 55–71 (2014).

    Article  PubMed  CAS  Google Scholar 

  30. Ahn, S. et al. Allosteric “beta-blocker” isolated from a DNA-encoded small molecule library. Proc. Natl. Acad. Sci. USA 114, 1708–1713 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Oswald, C. et al. Intracellular allosteric antagonism of the CCR9 receptor. Nature 540, 462–465 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Zheng, Y. et al. Structure of CC chemokine receptor 2 with orthosteric and allosteric antagonists. Nature 540, 458–461 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cheng, R.K.Y. et al. Structural insight into allosteric modulation of protease-activated receptor 2. Nature 545, 112–115 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Lu, J. et al. Structural basis for the cooperative allosteric activation of the free fatty acid receptor GPR40. Nat. Struct. Mol. Biol. 24, 570–577 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Roth, B.L. & Chuang, D.M. Multiple mechanisms of serotonergic signal transduction. Life Sci. 41, 1051–1064 (1987).

    Article  CAS  PubMed  Google Scholar 

  36. Urban, J.D. et al. Functional selectivity and classical concepts of quantitative pharmacology. J. Pharmacol. Exp. Ther. 320, 1–13 (2007). Crystallization of the concept of GPCR functional selectivity and biased signaling.

    Article  CAS  PubMed  Google Scholar 

  37. Whistler, J.L. & von Zastrow, M. Morphine-activated opioid receptors elude desensitization by β-arrestin. Proc. Natl. Acad. Sci. USA 95, 9914–9919 (1998). One of the first demonstrations that drugs may differ in their degree of G-protein and arrestin bias, along with its functional consequences.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Xiao, R.P., Ji, X. & Lakatta, E.G. Functional coupling of the β 2-adrenoceptor to a pertussis toxin-sensitive G protein in cardiac myocytes. Mol. Pharmacol. 47, 322–329 (1995).

    CAS  PubMed  Google Scholar 

  39. Masuho, I. et al. Distinct profiles of functional discrimination among G proteins determine the actions of G protein-coupled receptors. Sci. Signal. 8, ra123 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Kroeze, W.K. et al. PRESTO-Tango as an open-source resource for interrogation of the druggable human GPCRome. Nat. Struct. Mol. Biol. 22, 362–369 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Carlsson, J. et al. Ligand discovery from a dopamine D3 receptor homology model and crystal structure. Nat. Chem. Biol. 7, 769–778 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Allen, J.A. et al. Discovery of β-arrestin-biased dopamine D2 ligands for probing signal transduction pathways essential for antipsychotic efficacy. Proc. Natl. Acad. Sci. USA 108, 18488–18493 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wacker, D. et al. Structural features for functional selectivity at serotonin receptors. Science 340, 615–619 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lansu, K. et al. In silico design of novel probes for the atypical opioid receptor MRGPRX2. Nat. Chem. Biol. 13, 529–536 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Huang, X.P. et al. Allosteric ligands for the pharmacologically dark receptors GPR68 and GPR65. Nature 527, 477–483 (2015). Combination of physical screening with modeling and docking to discover ligands for orphan GPCRs, using the probe molecules that emerge to illuminate in vivo function.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Roth, B.L. DREADDs for neuroscientists. Neuron 89, 683–694 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wess, J., Nakajima, K. & Jain, S. Novel designer receptors to probe GPCR signaling and physiology. Trends Pharmacol. Sci. 34, 385–392 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lee, M.H. et al. The conformational signature of β-arrestin2 predicts its trafficking and signalling functions. Nature 531, 665–668 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wells, J.A. & McClendon, C.L. Reaching for high-hanging fruit in drug discovery at protein-protein interfaces. Nature 450, 1001–1009 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Tan, Q. et al. Structure of the CCR5 chemokine receptor-HIV entry inhibitor maraviroc complex. Science 341, 1387–1390 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Foord, S.M. et al. International Union of Pharmacology. XLVI. G protein-coupled receptor list. Pharmacol. Rev. 57, 279–288 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Taipale, J. & Beachy, P.A. The Hedgehog and Wnt signalling pathways in cancer. Nature 411, 349–354 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Robarge, K.D. et al. GDC-0449-a potent inhibitor of the hedgehog pathway. Bioorg. Med. Chem. Lett. 19, 5576–5581 (2009).

    Article  CAS  PubMed  Google Scholar 

  54. Yauch, R.L. et al. Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 326, 572–574 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wang, C. et al. Structural basis for Smoothened receptor modulation and chemoresistance to anticancer drugs. Nat. Commun. 5, 4355 (2014).

    Article  CAS  PubMed  Google Scholar 

  56. Lacroix, C. et al. Identification of novel smoothened ligands using structure-based docking. PLoS One 11, e0160365 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Sharpe, H.J., Wang, W., Hannoush, R.N. & de Sauvage, F.J. Regulation of the oncoprotein Smoothened by small molecules. Nat. Chem. Biol. 11, 246–255 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Roth, B.L. & Kroeze, W.K. Integrated approaches for genome-wide interrogation of the druggable non-olfactory G protein-coupled receptor superfamily. J. Biol. Chem. 290, 19471–19477 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sterling, T. & Irwin, J.J. ZINC 15—ligand discovery for everyone. J. Chem. Inf. Model. 55, 2324–2337 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Yanai, T. et al. Identification and molecular docking studies for novel inverse agonists of SREB, super conserved receptor expressed in brain. Genes Cells 21, 717–727 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Wardman, J.H. et al. Identification of a small-molecule ligand that activates the neuropeptide receptor GPR171 and increases food intake. Sci. Signal. 9, ra55 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Ngo, T. et al. Orphan receptor ligand discovery by pickpocketing pharmacological neighbors. Nat. Chem. Biol. 13, 235–242 (2017).

    Article  CAS  PubMed  Google Scholar 

  63. Gloriam, D.E., Foord, S.M., Blaney, F.E. & Garland, S.L. Definition of the G protein-coupled receptor transmembrane bundle binding pocket and calculation of receptor similarities for drug design. J. Med. Chem. 52, 4429–4442 (2009).

    Article  CAS  PubMed  Google Scholar 

  64. Munk, C., Harpsøe, K., Hauser, A.S., Isberg, V. & Gloriam, D.E. Integrating structural and mutagenesis data to elucidate GPCR ligand binding. Curr. Opin. Pharmacol. 30, 51–58 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Shehata, M.A. et al. Novel agonist bioisosteres and common structure-activity relationships for the orphan G protein-coupled receptor GPR139. Sci. Rep. 6, 36681 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Bassilana, F. et al. Target identification for a Hedgehog pathway inhibitor reveals the receptor GPR39. Nat. Chem. Biol. 10, 343–349 (2014).

    Article  CAS  PubMed  Google Scholar 

  67. Christopoulos, A., Lanzafame, A. & Mitchelson, F. Allosteric interactions at muscarinic cholinoceptors. Clin. Exp. Pharmacol. Physiol. 25, 185–194 (1998).

    Article  CAS  PubMed  Google Scholar 

  68. Shirey, J.K. et al. An allosteric potentiator of M4 mAChR modulates hippocampal synaptic transmission. Nat. Chem. Biol. 4, 42–50 (2008).

    Article  CAS  PubMed  Google Scholar 

  69. Gould, R.W. et al. State-dependent alterations in sleep/wake architecture elicited by the M4 PAM VU0467154 - Relation to antipsychotic-like drug effects. Neuropharmacology 102, 244–253 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Ghoshal, A. et al. Potentiation of M1 muscarinic receptor reverses plasticity deficits and negative and cognitive symptoms in a schizophrenia mouse model. Neuropsychopharmacology 41, 598–610 (2016).

    Article  CAS  PubMed  Google Scholar 

  71. Besnard, J. et al. Automated design of ligands to polypharmacological profiles. Nature 492, 215–220 (2012).

    Article  CAS  PubMed  Google Scholar 

  72. Bridges, T.M. et al. Chemical lead optimization of a pan Gq mAChR M1, M3, M5 positive allosteric modulator (PAM) lead. Part II: development of a potent and highly selective M1 PAM. Bioorg. Med. Chem. Lett. 20, 1972–1975 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Blaney, J.M., Hansch, C., Silipo, C. & Vittoria, A. Structure-activity relationships of dihydrofolate reductase inhibitors. Chem. Rev. 84, 333–407 (1984).

    Article  CAS  Google Scholar 

  74. Pinto, D.J. et al. Discovery of 1-(4-methoxyphenyl)-7-oxo-6-(4-(2-oxopiperidin-1-yl)phenyl)-4,5,6,7-tetrahydro-1H-pyrazolo[3,4-c]pyridine-3-carboxamide (apixaban, BMS-562247), a highly potent, selective, efficacious, and orally bioavailable inhibitor of blood coagulation factor Xa. J. Med. Chem. 50, 5339–5356 (2007).

    Article  CAS  PubMed  Google Scholar 

  75. Kolb, P. et al. Structure-based discovery of β2-adrenergic receptor ligands. Proc. Natl. Acad. Sci. USA 106, 6843–6848 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kruse, A.C. et al. Muscarinic receptors as model targets and antitargets for structure-based ligand discovery. Mol. Pharmacol. 84, 528–540 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kaczor, A.A. et al. Structure-based virtual screening for dopamine D2 receptor ligands as potential antipsychotics. ChemMedChem 11, 718–729 (2016).

    Article  CAS  PubMed  Google Scholar 

  78. Barelier, S., Sterling, T., O'Meara, M.J. & Shoichet, B.K. The recognition of identical ligands by unrelated proteins. ACS Chem. Biol. 10, 2772–2784 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Bohn, L.M., Gainetdinov, R.R., Lin, F.T., Lefkowitz, R.J. & Caron, M.G. μ-opioid receptor desensitization by β-arrestin-2 determines morphine tolerance but not dependence. Nature 408, 720–723 (2000). The first paper to demonstrate that arrestin signaling may have in vivo consequences relevant to drug actions.

    Article  CAS  PubMed  Google Scholar 

  80. DeWire, S.M. et al. A G protein-biased ligand at the μ-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine. J. Pharmacol. Exp. Ther. 344, 708–717 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Manglik, A. et al. Crystal structure of the μ-opioid receptor bound to a morphinan antagonist. Nature 485, 321–326 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Manglik, A. et al. Structure-based discovery of opioid analgesics with reduced side effects. Nature 537, 185–190 (2016). A recent success for structure-based discovery, leading to chemically novel agonists for the μ-opioid receptor. The novelty of the chemotype was echoed in new biological activities.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Broom, D.C. et al. Comparison of receptor mechanisms and efficacy requirements for delta-agonist-induced convulsive activity and antinociception in mice. J. Pharmacol. Exp. Ther. 303, 723–729 (2002).

    Article  CAS  PubMed  Google Scholar 

  84. Boerrigter, G. et al. Cardiorenal actions of TRV120027, a novel β-arrestin-biased ligand at the angiotensin II type I receptor, in healthy and heart failure canines: a novel therapeutic strategy for acute heart failure. Circ Heart Fail 4, 770–778 (2011).

    Article  CAS  PubMed  Google Scholar 

  85. Chen, X. et al. Structure-functional selectivity relationship studies of β-arrestin-biased dopamine D2 receptor agonists. J. Med. Chem. 55, 7141–7153 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Morgenweck, J., Frankowski, K.J., Prisinzano, T.E., Aubé, J. & Bohn, L.M. Investigation of the role of βarrestin2 in kappa opioid receptor modulation in a mouse model of pruritus. Neuropharmacology 99, 600–609 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. White, K.L. et al. The G protein-biased κ-opioid receptor agonist RB-64 is analgesic with a unique spectrum of activities in vivo. J. Pharmacol. Exp. Ther. 352, 98–109 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Conroy, J.L., Free, R.B. & Sibley, D.R. Identification of G protein-biased agonists that fail to recruit β-arrestin or promote internalization of the D1 dopamine receptor. ACS Chem. Neurosci. 6, 681–692 (2015).

    Article  CAS  PubMed  Google Scholar 

  89. Yadav, P.N., Kroeze, W.K., Farrell, M.S. & Roth, B.L. Antagonist functional selectivity: 5-HT2A serotonin receptor antagonists differentially regulate 5-HT2A receptor protein level in vivo. J. Pharmacol. Exp. Ther. 339, 99–105 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Schmid, C.L., Streicher, J.M., Meltzer, H.Y. & Bohn, L.M. Clozapine acts as an agonist at serotonin 2A receptors to counter MK-801-induced behaviors through a βarrestin2-independent activation of Akt. Neuropsychopharmacology 39, 1902–1913 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Marion, S., Weiner, D.M. & Caron, M.G. RNA editing induces variation in desensitization and trafficking of 5-hydroxytryptamine 2c receptor isoforms. J. Biol. Chem. 279, 2945–2954 (2004).

    Article  CAS  PubMed  Google Scholar 

  92. Clarke, W.P., Chavera, T.A., Silva, M., Sullivan, L.C. & Berg, K.A. Signalling profile differences: paliperidone versus risperidone. Br. J. Pharmacol. 170, 532–545 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Canal, C.E. et al. Molecular pharmacology and ligand docking studies reveal a single amino acid difference between mouse and human serotonin 5-HT2A receptors that impacts behavioral translation of novel 4-phenyl-2-dimethylaminotetralin ligands. J. Pharmacol. Exp. Ther. 347, 705–716 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. de Graaf, C. et al. Crystal structure-based virtual screening for fragment-like ligands of the human histamine H1 receptor. J. Med. Chem. 54, 8195–8206 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Wacker, D. et al. Conserved binding mode of human β2 adrenergic receptor inverse agonists and antagonist revealed by X-ray crystallography. J. Am. Chem. Soc. 132, 11443–11445 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Katritch, V. et al. Structure-based discovery of novel chemotypes for adenosine A2A receptor antagonists. J. Med. Chem. 53, 1799–1809 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Carlsson, J. et al. Structure-based discovery of A2A adenosine receptor ligands. J. Med. Chem. 53, 3748–3755 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Lane, J.R. et al. Structure-based ligand discovery targeting orthosteric and allosteric pockets of dopamine receptors. Mol. Pharmacol. 84, 794–807 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Jang, J.W. et al. Novel scaffold identification of mGlu1 receptor negative allosteric modulators using a hierarchical virtual screening approach. Chem. Biol. Drug Des. 87, 239–256 (2016).

    Article  CAS  PubMed  Google Scholar 

  100. Vass, M. et al. Dynamics and structural determinants of ligand recognition of the 5-HT6 receptor. J. Comput. Aided Mol. Des. 29, 1137–1149 (2015).

    Article  CAS  PubMed  Google Scholar 

  101. Kiss, R. & Keseru, G.M. Novel histamine H4 receptor ligands and their potential therapeutic applications: an update. Expert Opin. Ther. Pat. 24, 1185–1197 (2014).

    Article  CAS  PubMed  Google Scholar 

  102. Kolb, P. et al. Limits of ligand selectivity from docking to models: in silico screening for A(1) adenosine receptor antagonists. PLoS One 7, e49910 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Negri, A. et al. Discovery of a novel selective kappa-opioid receptor agonist using crystal structure-based virtual screening. J. Chem. Inf. Model. 53, 521–526 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Keiser, M.J. et al. Relating protein pharmacology by ligand chemistry. Nat. Biotechnol. 25, 197–206 (2007).

    Article  CAS  PubMed  Google Scholar 

  105. Huang, X.P. et al. Parallel functional activity profiling reveals valvulopathogens are potent 5-hydroxytryptamine2B receptor agonists: implications for drug safety assessment. Mol. Pharmacol. 76, 710–722 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Support was given by NIH grants U01104974, and R35GM122481, and the NIMH-PDSP. We thank T. Kenakin for discussions of allostery, J. Pottel and A. Levit for reading this manuscript, and the latter for chemoinformatic assistance.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Bryan L Roth or Brian K Shoichet.

Ethics declarations

Competing interests

B.K.S. and B.L.R. are co-founders of Epiodyne, a GPCR ligand discovery company. B.K.S. and J.J.I. are co-founders of Blue Dolphin, a library docking CRO.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roth, B., Irwin, J. & Shoichet, B. Discovery of new GPCR ligands to illuminate new biology. Nat Chem Biol 13, 1143–1151 (2017). https://doi.org/10.1038/nchembio.2490

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2490

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research