848
Views
40
CrossRef citations to date
0
Altmetric
Review

Signaling bias in drug discovery

Pages 321-333 | Received 11 Nov 2016, Accepted 16 Feb 2017, Published online: 09 Mar 2017

References

  • Kenakin TP, Ambrose JR, Irving PE. The relative efficiency of beta adrenoceptor coupling to myocardial inotropy and diastolic relaxation: organ-selective treatment for diastolic dysfunction. J Pharmacol Exp Ther. 1991;257:1189–1197. ​​​​
  • Roth BL, Chuang DM. Multiple mechanisms of serotonergic signal transduction. Life Sci. 1987;41:1051–1064.
  • Kenakin TP, Morgan PH. Theoretical effects of single and multiple transducer receptor coupling proteins on estimates of the relative potency of agonists. Mol Pharmacol. 1989;35:214–222.
  • Freire E. Statistical thermodynamic linkage between conformational and binding equilibria. Adv Protein Chem. 1998;51:255–279.
  • Frauenfelder H, Sligar SG, Wolynes PG. The energy landscapes and motions of proteins. Science. 1991;254:1598–1603.
  • Hilser VJ, Dowdy D, Oas TG, et al. The structural distribution of cooperative interactions in proteins: analysis of the native state ensemble. Proc Natl Acad Sci USA. 1998;95:9903–9908.
  • Hilser VJ, García-Moreno EB, Oas TG, et al. A statistical thermodynamic model of the protein ensemble. Chem Rev. 2006;106:1545–1558.
  • Hilser VJ, Thompson EB. Intrinsic disorder as a mechanism to optimizeallosteric coupling in proteins. Proc Natl Acad Sci USA. 2007;104:8311–8315.
  • Onaran HO, Costa T. Agonist efficacy and allosteric models of receptor action. Ann N Y Acad Sci. 1997;812:98–115.
  • Onaran HO, Scheer A, Cotecchia S, et al. A look at receptor efficacy. From the signaling network of the cell to the intramolecular motion of the receptor,in the pharmacology of functional, biochemical, and recombinant systems (handbook of experimental pharmacology). Kenakin TP, Angus JA, eds. Vol. 148:217–280. Heidelberg, Germany: Springer; 2000.
  • Hermans E. Biochemical and pharmacological control of the multiplicity of coupling at G-protein-coupled receptors. Pharmacol Ther. 2003;99:25–44.
  • Laugwitz K-L, Allgeier A, Offermanns S, et al. The human thyrotropic receptor: a heptahelical receptor capable of stimulating members of all four G protein families. Proc Natl Acad Sci USA. 1996;93:116–120.
  • Spengler D, Waeber C, Pantaloni C, et al. Differential signal transduction by five splice variants of the PACAP receptor. Nature. 1993;365:170–175.
  • Kenakin T. Agonist-receptor efficacy. II. Agonist trafficking of receptor signals. Trends Pharmacol Sci. 1995;16:232–238.
  • Jarpe MB, Knall C, Mitchell FM, et al. [D-Arg1, D-Phe5,D-Trp7,9,Leu11]Substance P acts as a biased agonist toward neuropeptide and chemokine receptors. J Biol Chem. 1998;273:3097–3104.
  • Lawler CP, Prioleau C, Lewis MM, et al. Interactions of the novel antipsychotic aripiprazole(OPC-14597) with dopamine and serotonin receptor subtypes. Neuropsychopharmacology. 1999;20:612–627.
  • Kenakin T. New concepts in drug discovery: collateral efficacy and permissive antagonism. Nat Rev Drug Discov. 2005;4:919–927.
  • Jl W, Von Zastrow M. Dissociation of functional roles of dynamin in receptor-mediated endocytosis and mitogenic signal transduction. J Biol Chem. 1999;274:24575–24578.
  • Kudlacek O, Waldhoer M, Kassack MU, et al. Biased inhibition by a suramin analogue of A1-adenosine eceptor/G protein coupling in fused receptor/G protein tandems: the A1-adenosine receptor is predominantly coupled to Go-alpha in human brain. Naunyn Schmiedebergs Arch Pharmacol. 2002;365:8–16.
  • Manning DR. Measures of efficacy using G proteins as endpoints: differential engagement of G proteins through single receptors. Mol Pharmacol. 2002;62:451–452.
  • Gonzalez E, Kulkarni H, Bolivar H, et al. The influence of CCL3L1 gene-containing segmental duplications on HIV-1/AIDS susceptibility. Science. 2005;307:1434–1440.
  • Muniz-Medina VM, Jones S, Maglich JM, et al. The relative activity of “function sparing” HIV-1 entry inhibitors on viral entry and CCR5 internalization: is allosteric functional selectivity a valuable therapeutic property? Mol Pharmacol. 2009;75:490–501.
  • Kenakin T, Watson C, Muniz-Medina V, et al. A simple method for quantifying functional selectivity and agonist bias. ACS Chem Neurosci. 2012;3:193–203.
  • Ferrari SL, Pierroz DD, Glatt V, et al. Bone response to intermittent parathyroid hormone is altered in mice null for β-Arrestin2. Endocrinology. 2005;146:1854–1862.
  • Gesty-Palmer D, Chen M, Reiter E, et al. Distinct beta-arrestin- and G protein-dependent pathways for parathyroid hormone receptor-stimulated ERK1/2 activation. J Biol Chem. 2006;281:10856–10864.
  • Gesty-Palmer D, Flannery P, Yuan L, et al. A b-arrestin-biased agonist of the parathyroid hormone receptor (PTH1R) promotes bone formation independent of G protein activation. Sci Transl Med. 2009;1:1ra1.
  • Sonoda N, Imamura T, Yoshizaki T, et al. β-Arrestin-1 mediates glucagon-like peptide-1 signaling to insulin secretion in cultured pancreatic b cells. Proc Natl Acad Sci USA. 2008;105:6614–6619.
  • Raehal KM, Walker JK, Bohn LM. Morphine side effects in beta-arrestin 2 knockout mice. J Pharmacol Exp Ther. 2005;314:1195–1201.
  • DeWire SM, Yamashita DS, Rominger DH, 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. 2013;344:708–717.
  • White KL, Scopton AP, Rives M-L, et al. Identification of novel functionally selective k-opioid receptor scaffolds. Mol Pharmacol. 2014;85:83–90.
  • Violin JD, Dewire SM, Barnes WG, et al. G protein-coupled receptor kinase and beta-arrestin-mediated desensitization of the angiotensin II type 1A receptor elucidated by diacylglycerol dynamics. J Biol Chem. 2006;281:36411–36419.
  • Violin JD, DeWire SM, Yamashita D, et al. Selectively engaging β-arrestins at the angiotensin II type 1 receptor reduces blood pressure and increases cardiac performance. J Pharmacol Exp Ther. 2010;335:572–579.
  • Watson C, Chen G, Irving P, et al. The use of stimulus-biased assay systems to detect agonist-specific receptor active states: implications for the trafficking of receptor stimulus by agonists. Mol Pharmacol. 2000;58:1230–1238.
  • Chen G, Way J, Armour S, et al. Use of constitutive G protein-coupled receptor activity for drug discovery. Mol Pharmacol. 2000;57:125–134.
  • Christmanson L, Westermark P, Betsholtz C. Islet amyloid polypeptide stimulates cyclic AMP accumulation via the porcine calcitonin receptor. Biochem Biophys Res Commun. 1994;205:1226–1235.
  • Felker M, Butler J, Collins S et al.: Biased ligand of the angiotensin receptor in acute heart failure (BLAST-AHF) Presented to the Heart Failure Association of the ESC, Florence Italy 2016: http://www.trevena.com/pdf/Phase2bBLAST-AHFdata.pdf
  • Black JW, Leff P. Operational models of pharmacological agonism. Proc R Soc Lond B Biol Sci. 1983;220:141–162.
  • Black JW, Leff P, Shankley NP, et al. An operational model of pharmacological agonism: the effect of E/[A] curve shape on agonist dissociation constant estimation. Br J Pharmacol. 1985;84:561–571.
  • Kenakin T. New concepts in pharmacological efficacy at 7TM receptors: IUPHAR review 2. Br J Pharmacol 168. 2013;168:554–575. ​​​​
  • Kenakin TP. The effective application of biased signaling to new drug discovery. Mol Pharmacol. 2015;88:1055–1061.
  • Stahl EL, Zhou L, Ehlert FJ, et al. A novel method for analyzing extremely biased agonism at G protein–coupled receptors. Mol Pharmacol. 2015;87:866–877.
  • Unett DJ, Gatlin J, Anthony TL, et al. Kinetics of 5-HT2B receptor signaling: profound agonist-dependent effects on signaling onset and duration. J Pharmacol Exp Ther. 2013;347:645–659.
  • Herrenbrink CK, Sykes DA, Donthamsetti P, et al. The role of kinetic context in apparent biased agonism at GPCRs Nature Commun. DOI:10.1038/ncomms10842
  • Rasmussen,S.G.F., DeVree, B.T., Zou,Y., Kruse, A.C., Chung, K.Y., Kobilka, T.S., et al. (2011a) Crystal structure of the β2 adrenergic receptor–Gs protein complex. Nature 477: 549–555.
  • Rasmussen SGF, Choi H-J, Fung JJ, et al. Structure of a nanobody-stabilized active state of the β2 adrenoceptor. Nature. 2011b;469:175–180.
  • Kruse AC, Ring AM, Manglik A, et al. Activation and allosteric modulatiojn of a muscarinic acetylcholine receptor. Nature. 2013;504:101–106.
  • Mary S, Damian M, Louet M, et al. Ligands and signaling proteins govern the conformational landscape explored by a G protein-coupled receptor. Proc Natl Acad Sci USA. 2012;109:8304–8309.
  • Yan F, Mosier PD, Westkaemper RB, et al. Gα-Subunits differentially alter the conformation and agonist affinity of κ-opioid receptors. Biochem. 2008;47:1567–1578.
  • Kenakin TP. What is pharmacological ‘affinity’? Relevance to biased agonism and antagonism. Trends Pharmacol Sci. 2014;35:434–441.
  • Strachan RT, Sciaky N, Cronan MR, et al. Genetic deletion of p90 ribosomal s6 kinase 2 alters patterns of 5-Hydroxytryptamine2A serotonin receptor functional selectivity. Mol Pharmacol. 2010;77:327–338.
  • Nijmeijer S, Vischer HF, Rosethorne EM, et al. Analysis of multiple histamine H4 receptor compound classes uncovers Gαi protein- and β-Arrestin2-biased ligands. Mol Pharmacol. 2012;82:1174–1182.
  • Baltos J-A, Paoletta S, Nguyen ATN, et al. Structure-activity analysis of biased agonism at the human adenosine A3 receptor. Mol Pharamol. 2016;90:11–12.
  • McPherson J, Rivero G, Baptist M, et al. μ-opioid receptors: correlation of agonist efficacy for signalling with ability to activate internalization. Mol Pharmacol. 2010;78:756–766.
  • Nickolls SA, Waterfield A, Williams RE, et al. Understanding the effect of different assay formats on agonist parameters: a study using the µ-opioid receptor. J Biomol Screen. 2011;16:706–716.
  • Thompson GL, Lane JR, Coudrat T, et al. Biased agonism of endogenous opioid peptides at the μ-opioid receptor. Mol Pharmacol. 2015;88:35–346.
  • D’Antona AM, Ahn KH, Kendall DA. Mutations of CB1 T219 produce active and inactive receptor forms: correlations with ligand affinity, receptor stability and cellular localization. Biocehm. 2006;45:5606–5617.
  • Belmer A, Doly S, Setola V, et al. Role of N-terminal region in G protein-coupled receptor functions: negative modulation revealed by 5-HT2B receptor polymorphisms. Mol Pharmacol. 2014;85:127–138.
  • Tschammer N, Bollinger S, Kenakin T, et al. Histidine 6.55 is a major determinant of ligand-biased signaling in dopamine D2L receptor. Mol Pharmacol. 2011;79(3):575–585.
  • Gregory KJ, Sexton PM, Tobin AB, et al. Stimulus bias provides evidence for conformational constraints in the structure of a G protein-coupled receptor. J Biol Chem. 2012;287:37066–37077.
  • Kenakin TP. The relative contribution of affinity and efficacy to agonist activity: organ selectivity of noradrenaline and oxymetazoline. Br J Pharmacol. 1984;81:131–141.
  • Just S, Illing S, Trester-Zedlitz M, et al. Differentiation of opioid drug effects by hierarchical multi-site phosphorylation. Mol Pharmacol. 2013;83:633–639.
  • Quoyera J, Janz JM, Luoc J, et al. Pepducin targeting the C-X-C chemokine receptor type 4 acts as a biased agonist favoring activation of the inhibitory G protein. Proc Natl Acad Sci USA. 2013;110:E5088–E5097.
  • Maillet EL, Pellegrini N, Valant C, et al. A novel, conformation-specific allosteric inhibitor of the tachykinin NK2 receptor (NK2R) with functionally selective properties. Faseb J. 2007;21:2124–2134.
  • Mathiesen JM, Ulven T, Martini L, et al. Identification of indole derivatives exclusively interfering with a G protein independent signaling pathway of the prostaglandin D2 receptor CRTH2. Mol Pharmacol. 2005;68:393–402.
  • Bradley SJ, Langmead CJ, Watson JM, et al. Quantitative analysis reveals multiple mechanisms of allosteric modulation of the mGlu5 receptor in rat astroglia. Mol Pharmacol. 2011;79:874–885.
  • Koole C, Wootten D, Simms J, et al. Allosteric ligands of the glucagon-like peptide 1 receptor (GLP-1R) differentially modulate endogenous and exogenous peptide responses in a pathway-selective manner: implications for drug screening. Mol Pharmacol. 2010;78:456–465.
  • Cook AE, Mistry SN, Gregory KJ, et al. Biased allosteric modulation at the CaS receptor engendered by structurally diverse calcimimetics. Br J Pharmacol. 2015;172:185–200.
  • Davey AE, Leach K, Valant C, et al. Positive and negative allosteric modulators promote biased signaling at the calcium-sensing receptor. Endocrinology. 2012;153:1232–1241.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.