716
Views
13
CrossRef citations to date
0
Altmetric
Review

Evolution of the drug-target residence time model

Pages 1441-1451 | Received 23 Apr 2021, Accepted 24 Jun 2021, Published online: 01 Jul 2021

References

  • Hill AV. The mode of action of nicotine and curari determined by the form of the contraction curve and the method of temperature coefficients. J Physiol. 1909;39(5):361–373.
  • Langley JN. On the reaction of cells and of nerve-endings to certain poisons, chiefly as regards the reaction of striated muscle to nicotine and curari. J Physiol. 1905;33(4–5):374–413.
  • Ehrlich P. Chemotherapeutics: scientific principles, methods and results. Lancet. 1913;182:445–451.
  • Maehle AH, Prüll CR, Halliwell RF. The emergence of the drug receptor theory. Nat Rev Drug Discov. 2002;1(8):637–641.
  • Swinney DC. Biochemical mechanisms of drug action: what does it take for success? Nat Rev Drug Discov. 2004;3(9):27–41.
  • Copeland RA. Evaluation of enzyme inhibitors in drug discovery: a guide for medicinal chemists and pharmacologists. 2nd ed. Hoboken, NJ, USA: Wiley; 2013.
  • Copeland RA, Pompliano DL, Meek TD. Drug-target residence time and its implications for lead optimization. Nat Rev Drug Discov. 2006;5(9):730–739.
  • Tummino PJ, Copeland RA. Residence time of receptor-ligand complexes and its effect on biological function. Biochemistry. 2008;47(20):5481–5492.
  • Vauquelin G, Van Leifde I. Slow antagonist dissociation and long-lasting in vivo receptor protection. Trends Pharmacol Sci. 2006;27(7):355–359.
  • Zhang R, Monsma F. The importance of drug-target residence time. Curr Opin Drug Discov Dev. 2009;12:488–496.
  • Copeland RA. The dynamics of drug-target interactions: drug-target residence time and its impact on efficacy and safety. Curr Opin Drug Discov Dev. 2010;5(4):305–310.
  • Lu H, Tonge PJ. Drug-target residence time: critical information for lead optimization. Curr Opin Chem Biol. 2010;14(4):467–474.
  • Swinney DC, Haubrich BA, Van Liefde I, et al. The role of binding kinetics in GPCR drug discovery. Curr Top Med Chem. 2015;15(24):2504–2522.
  • Copeland RA. The drug-target residence time model: a 10-year retrospective. Nat Rev Drug Discov. 2016;15(2):87–95.
  • van der Velden WJC, Heitman LH, Rosenkilde MM. Implications of ligand-receptor binding kinetics for therapeutic targeting of G protein-coupled receptors. ACS Pharmacol Transi Sci. 2020;3(2):179–189.
  • Markgren P-O, Schaal W, Hamalainen M, et al. Relationship between structure and interaction kinetics for HIV-1 protease inhibitors. J Med Chem. 2002;45(25):5430–5439.
  • Vauquelin G. Effects of target binding kinetics on in vivo drug efficacy: koff, kon and rebinding. Br J Pharmacol. 2016;173(15):2319–2334.
  • Copeland RA. Enzymes: a practical introduction to structure, mechanism and data analysis. 2nd ed. Hoboken, NJ, USA: Wiley; 2000.
  • Dahl G, Akerud T. Pharmacokinetics and the drug-target residence time concept. Drug DIscov Today. 2013;18(15–16):697–702.
  • deWitte WEA, Danhof M, van der Graff PH, et al. In vivo target residence time and kinetic selectivity: the association rate constant as determinant. <jtl>trends Pharmacol Sci. 2016;37(10):831–842.
  • Folmer RHA. Drug target residence time: a misleading concept. Drug Discov Today. 2018;23(1):12–16.
  • deWitte WEA, Danhof M, van der Graff PH, et al. The implications of target saturation for the use of drug-target residence time. Nat Rev Drug Discov. 2019;18(1):84.
  • Wood ER, Truesdale AT, McDonald OB, et al. A unique structure for epidermal growth factor receptor bound to GW572016 (lapatinib): relationship among protein conformation, inhibitor off-rate and receptor activity in tumor cells. Cancer Res. 2004;64(18):6652–6659.
  • Willemsen-Seegers N, Uitdehaag JCM, Prinsen MBW, et al. Compound selectivity and target residence time of kinase inhibitors studied with surface plasmon resonance. J Mol Biol. 2017;429(4):574–586.
  • Vauquelin G. Rebinding: or why drugs may act longer in vivo than expected from their in vitro target residence time. Expert Opin Drug Discov. 2010;5(10):927–941.
  • Vauquelin G. Link between a high kon for drug binding and a fast clinical action: to be or not to be? MedChemComm. 2018;9(9):1426–1438.
  • Gebre ST, Cameron SA, Li L, et al. Intracellular rebinding of transition-state analogues provides extended in vivo inhibition lifetimes on human purine nucleoside phosphorylase. J Biol Chem. 2017;292(38):15907–15915.
  • Georgi V, Schiele F, Berger B-T, et al. Binding kinetics survey of the drugged kinome. J Am Chem Soc. 2018;140(46):15774–15782.
  • van Liefe I, Vauquelin G. Sartan–AT1 receptor interactions: in vitro evidence for insurmountable antagonism and inverse agonism. Mol Cell Endocrinol. 2009;302(2):237–243.
  • Lacourciere Y, Asmer R. A comparison of efficacy and duration of action of candesartan cilexetil and losartan as assessed by clinic and ambulatory blood pressure after a missed dose, in truly hypertensive patients: a placebo-controlled forced titration study. Am J Hypertens. 1999;12(12):1181–1187.
  • Nederpelt I, Kuzikov M, deWitte WEA, et al. From receptor binding kinetics to signal transduction; a missing link in predicting in vivo drug-action. Sci Rep. 2017;7:14169.
  • Anderson K, Lai Z, McDonald OB, et al. Biochemical characterization of GSK1070916, a potent and selective inhibitor of Aurora B and Aurora C kinases with an extremely long residence time. Biochem J. 2009;420(2):259–265.
  • Utidehaag JCM, De Man J, Willemsen-Seegers N, et al. Target residence time-guided optimization on TTK kinase results in inhibitors with potent anti-proliferative activity. J Mol Biol. 2017;429(14):2211–2230.
  • Crawford JJ, Johnson AR, Misner DL, et al. Discovery of GDC-0853: a potent, selective, and noncovalent Burton;s tyrosine kinase inhibitor in early clinical development. J Med Chem. 2018;61(6):2227–2245.
  • Basavapathruni A, Jin L, Daigle SR, et al. Conformational adaptation drives potent, selective and durable inhibition of the human protein methyltransferase DOT1L. Chem Biol Drug Design. 2012;80(6):971–980.
  • Van Aller GS, Pappalardi MB, Ott HM, et al. Long residence time inhibition of EZH2 in activated polycomb repressive complex 2. ACS Chem Biol. 2014;9(3):622–629.
  • Knutson S, Kawano S, Minoshima Y, et al. Selective Inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-hodgkin lymphoma. Mol Cancer Ther. 2014;13(4):842–854.
  • Gooljarsingh LT, Fernandes C, Yan K, et al. A biochemical rationale for the anticancer effects of HSP90 inhibitors: slow, tight binding inhibition by geldanamycin and its analogues. Proc Natl Acad Sci USA. 2006;103(20):7625–7630.
  • Amaral M, Kokh DB, Bormke J, et al. Protein conformational flexibility modulates kinetics and thermodynamics of drug binding. Nature Commun. 2017;8(1):2276.
  • Yun Y, Chen J, Liu R, et al. Long residence time adenosine A1 receptor agonists produce sustained wash-resistant antilipolytic effect in rat adipocytes. Biochem Pharmacol. 2019;164:45–52.
  • Doornbos MLJ, Cid JM, Haubrich J, et al. Discovery and kinetic profiling of 7-Aryl-1,2,4-triazolo[4,3-a]pyridines: positive allosteric modulators of the metabotropic glutamate receptor 2. J Med Chem. 2017;60(15):6704–6720.
  • Xia L, Burger WAC, van Veldhoven JPD, et al. Structure–affinity relationships and structure–kinetics relationships of pyrido[2,1-f]purine-2,4-dione derivatives as human adenosine A3 receptor antagonists. J Med Chem. 2017;60(17):7555–7568.
  • Walkup GK, You Z, Ross PL, et al. Translating slow-binding kinetics into cellular and in vivo effects. Nat Chem Biol. 2015;11(6):416–423.
  • Daryaee F, Zhang Z, Goarty KR, et al. A quantitative mechanistic PK/PD model directly connects Btk target engagement and in vivo efficacy. J Chem Sci. 2017;8(5):3434–3443.
  • Lee KSS, Yang J, Niu J, et al. Drug-target residence time affects in vivo target occupancy through multiple pathways. ACS Central Sci. 2019;5(9):1614–1624.
  • Bradshaw JM, McFarland JM, Paavilainen VO, et al. Prolonged and tunable residence time using reversible covalent kinase inhibitors. Nat Chem Biol. 2015;11(7):521–531.
  • Spagnuolo LA, Eltschkner S, Yu W, et al. Evaluating the contribution of transition-state destabilization to changrs in the residence time of triazole-based InhA inhibitors. J Am Chem Soc. 2017;139(9):3417–3429.
  • Tiwary P, Limongelli V, Salvalaglio M, et al. Kinetics of protein-ligand unbinding: predicting pathways, rates and rate-limiting steps. Proc Natl Acad Sci USA. 2009;112:E386–E391.
  • Tonge PJ. Drug-target kinetics in drug discovery. ACS Chem Neurosci. 2018;9(1):29–39.
  • Bernetti M, Masetti M, Rocchia W, et al. Kinetics of drug binding and residence time. Annu Rev Phys Chem. 2019;70(1):143–171.
  • Supuran C. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov. 2008;7(2):168–181.
  • Segala E, Guo D, Cheng RKY, et al. Controlling the dissociation of ligands from the adenosine A2A receptor through modulation of salt bridge strength. J Med Chem. 2016;59(13):6470–6479.
  • Jin M, Petronella B, Cooke A, et al. Discovery of novel insulin-like growth factor-1 receptor inhibitors with unique time-dependent binding kinetics. ACS Med Chem Lett. 2013;4(7):627–631.
  • Vauquelin G, Van Liefe I, Swinney DC. On the different experimental manifestations of two-state “induced-fit” binding of drugs to their cellular targets. Br J Pharmacol. 2016;173(8):1268–1285.

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.