Skip to main content

Advertisement

Log in

Novel Multiplexed High Throughput Screening of Selective Inhibitors for Drug-Metabolizing Enzymes Using Human Hepatocytes

  • Research Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

Selective chemical inhibitors are critical for reaction phenotyping to identify drug-metabolizing enzymes that are involved in the elimination of drug candidates. Although relatively selective inhibitors are available for the major cytochrome P450 enzymes (CYP), they are quite limited for the less common CYPs and non-CYPs. To address this gap, we developed a multiplexed high throughput screening (HTS) assay using 20 substrate reactions of multiple enzymes to simultaneously monitor the inhibition of enzymes in a 384-well format. Four 384-well assay plates can be run at the same time to maximize throughput. This is the first multiplexed HTS assay for drug-metabolizing enzymes reported. The HTS assay is technologically enabled with state-of-the-art robotic systems and highly sensitive modern LC-MS/MS instrumentation. Virtual screening is utilized to identify inhibitors for HTS based on known inhibitors and enzyme structures. Screening of ~4600 compounds generated many hits for many drug-metabolizing enzymes including the two time-dependent and selective aldehyde oxidase inhibitors, erlotinib and dibenzothiophene. The hit rate is much higher than that for the traditional HTS for biological targets due to the promiscuous nature of the drug-metabolizing enzymes and the biased compound selection process. Future efforts will focus on using this method to identify selective inhibitors for enzymes that do not currently have quality hits and thoroughly characterizing the newly identified selective inhibitors from our screen. We encourage colleagues from other organizations to explore their proprietary libraries using a similar approach to identify better inhibitors that can be used across the industry.

Graphical abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Abbreviations

ABT:

1-Aminobenzotriazole

ADME:

Absorption, distribution, metabolism, and excretion

AO:

Aldehyde oxidase

CBR:

Carbonyl reductase

CE:

Collision energy

CES:

Carboxylesterase

CO2 :

Carbon dioxide

CXP:

Collision cell exit potential

CYP:

Cytochrome P450

DDI:

Drug-drug interaction

DMSO:

Dimethyl sulfoxide

DP:

Declustering potential

EPHX:

Epoxide hydrolase

f m :

Fraction metabolized

FMO:

Flavin-containing monooxygenase

HEPES:

4-(2-Hydroxyethyl)-1-piperazineethanesulfonic acid

HHEP:

Human hepatocytes

HLM:

Human liver microsomes

HPLC:

High-performance liquid chromatography

HTS:

High throughput screening

IC50 :

Half-maximal inhibitory concentration

IVIVE:

In vitro-to-in vivo extrapolation

K I :

Inactivation rate constant

k inact :

The maximum rate of enzyme inactivation

K i,u :

Unbound inhibition constant

K m :

Michaelis constant

LC-MS/MS:

Liquid chromatography with tandem mass spectrometry

MAO:

Monoamine oxidase

MRM:

Multiple-reaction monitoring

4-MU:

4-Methylumbelliferone

M/Z:

Mass-to-charge ratio

Na2CO3 :

Sodium carbonate

NADPH:

Nicotinamide adenine dinucleotide phosphate, reduced form

NAT:

N-acetyltransferase

PK:

Pharmacokinetics

Q1:

First quadrupole mass filter

Q3:

Second quadrupole mass filter

TDI:

Time-dependent inhibition

UGT:

Uridine 5′-diphospho-glucuronosyltransferase

WEM:

Williams E medium

References

  1. Sun D, Gao W, Hu H, Zhou S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm Sin B. 2022;12(7):3049–62. https://doi.org/10.1016/j.apsb.2022.02.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Dickson M, Gagnon JP. Key factors in the rising cost of new drug discovery and development. Nat Rev Drug Discov. 2004;3(5):417–29. https://doi.org/10.1038/nrd1382.

    Article  CAS  PubMed  Google Scholar 

  3. Hughes JP, Rees S, Kalindjian SB, Philpott KL. Principles of early drug discovery. Br J Pharmacol. 2011;162(6):1239–49. https://doi.org/10.1111/j.1476-5381.2010.01127.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Doytchinova I. Drug design-Past, present, future. Molecules (Basel, Switzerland). 2022;27(5). doi: https://doi.org/10.3390/molecules27051496.

  5. Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrell S, Owen RM, et al. An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat Rev Drug Discov. 2015;14(7):475–86. https://doi.org/10.1038/nrd4609.

    Article  CAS  PubMed  Google Scholar 

  6. Bocci G, Carosati E, Vayer P, Arrault A, Lozano S, Cruciani G. ADME-Space: A new tool for medicinal chemists to explore ADME properties. Sci Rep. 2017;7(1):6359. https://doi.org/10.1038/s41598-017-06692-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Di L. Reaction phenotyping to assess victim drug-drug interaction risks. Expert Opin Drug Discov. 2017;12(11):1105–15. https://doi.org/10.1080/17460441.2017.1367280.

    Article  CAS  PubMed  Google Scholar 

  8. Zientek MA, Youdim K. Reaction phenotyping: Advances in the experimental strategies used to characterize the contribution of drug-metabolizing enzymes. Drug Metab Dispos. 2015;43(1):163–81. https://doi.org/10.1124/dmd.114.058750.

    Article  CAS  PubMed  Google Scholar 

  9. Bohnert T, Patel A, Templeton I, Chen Y, Lu C, Lai G, et al. Evaluation of a new molecular entity as a victim of metabolic drug-drug interactions-An industry perspective. Drug Metab Dispos. 2016;44(8):1399–423. https://doi.org/10.1124/dmd.115.069096.

    Article  CAS  PubMed  Google Scholar 

  10. Strelevitz TJ, Orozco CC, Obach RS. Hydralazine as a selective probe inactivator of aldehyde oxidase in human hepatocytes: Estimation of the contribution of aldehyde oxidase to metabolic clearance. Drug Metab Dispos. 2012;40(7):1441–8. https://doi.org/10.1124/dmd.112.045195.

    Article  CAS  PubMed  Google Scholar 

  11. Yang X, Johnson N, Di L. Evaluation of cytochrome P450 selectivity for hydralazine as an aldehyde oxidase inhibitor for reaction phenotyping. J Pharm Sci. 2019;108(4):1627–30. https://doi.org/10.1016/j.xphs.2018.11.007.

    Article  CAS  PubMed  Google Scholar 

  12. Yang X, Atkinson K, Di L. Novel cytochrome P450 reaction phenotyping for low-clearance compounds using the hepatocyte relay method. Drug Metab Dispos. 2016;44(3):460–5.

    Article  PubMed  Google Scholar 

  13. Tess D, Chang GC, Keefer C, Carlo A, Jones R, Di L. In vitro-in vivo extrapolation and scaling factors for clearance of human and preclinical species with liver microsomes and hepatocytes. AAPS J. 2023;25(3):40. https://doi.org/10.1208/s12248-023-00800-x.

    Article  CAS  PubMed  Google Scholar 

  14. Ramsden D, Smith D, Arenas R, Frederick K, Cerny MA. Identification and characterization of a selective human carbonyl reductase 1 substrate. Drug Metab Dispos. 2018;46(10):1434–40. https://doi.org/10.1124/dmd.118.082487.

    Article  CAS  PubMed  Google Scholar 

  15. Lee CA, Jones JP 3rd, Katayama J, Kaspera R, Jiang Y, Freiwald S, et al. Identifying a selective substrate and inhibitor pair for the evaluation of CYP2J2 activity. Drug Metab Dispos. 2012;40(5):943–51. https://doi.org/10.1124/dmd.111.043505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Li X, Jeso V, Heyward S, Walker GS, Sharma R, Micalizio GC, Cameron MD. Characterization of T-5 N-oxide formation as the first highly selective measure of CYP3A5 activity. Drug Metab Dispos. 2014;42(3):334–42. https://doi.org/10.1124/dmd.113.054726.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Kung P-P, Bingham P, Brooun A, Collins M, Deng Y-L, Dinh D, et al. Optimization of orally bioavailable enhancer of zeste homolog 2 (EZH2) inhibitors using ligand and property-based design strategies: Identification of development candidate (R)-5,8-dichloro-7-(methoxy (oxetan-3-yl)methyl)-2-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-3,4-dihydroisoquinolin-1(2H)-one (PF-06821497). J Med Chem. 2018;61(3):650–65. https://doi.org/10.1021/acs.jmedchem.7b01375.

    Article  CAS  PubMed  Google Scholar 

  18. Ghosal A. Chapter 8 - Evaluation of the clearance mechanism of non-CYP-mediated drug metabolism and DDI as a victim drug. In: Ma S, Chowdhury SK, editors. Identification and Quantification of Drugs, Metabolites, Drug Metabolizing Enzymes, and Transporters. Second ed. Amsterdam: Elsevier; 2020. p. 237–71.

    Chapter  Google Scholar 

  19. Zientek M, Youdim K. Simultaneous determination of multiple CYP inhibition constants using a cocktail-probe approach. Methods in molecular biology (Clifton, NJ). 2013;987:11–23. https://doi.org/10.1007/978-1-62703-321-3_2.

    Article  CAS  Google Scholar 

  20. Zientek M, Miller H, Smith D, Dunklee MB, Heinle L, Thurston A, et al. Development of an in vitro drug-drug interaction assay to simultaneously monitor five cytochrome P450 isoforms and performance assessment using drug library compounds. J Pharmacol Toxicol Methods. 2008;58(3):206–14. https://doi.org/10.1016/j.vascn.2008.05.131.

    Article  CAS  PubMed  Google Scholar 

  21. Yang X, Ryu S, Di L. Dasotraline as a selective cytochrome 2B6 inhibitor for reaction phenotyping. Biopharm Drug Dispos. 2019;40(9):358–61. https://doi.org/10.1002/bdd.2207.

    Article  CAS  PubMed  Google Scholar 

  22. Derringer G, Suich R. Simultaneous optimization of several response variables. J Qual Technol. 1980;12(4):214–9. https://doi.org/10.1080/00224065.1980.11980968.

    Article  Google Scholar 

  23. Graczyk PP. Gini Coefficient: A new way to express selectivity of kinase inhibitors against a family of kinases. J Med Chem 2007;50(23):5773-5779. doi: https://doi.org/10.1021/jm070562u.

  24. Margaillan G, Rouleau M, Klein K, Fallon JK, Caron P, Villeneuve L, et al. Multiplexed targeted quantitative proteomics predicts hepatic glucuronidation potential. Drug Metab Dispos. 2015;43(9):1331–5. https://doi.org/10.1124/dmd.115.065391.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dalvie D, Di L. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol Ther. 2019;201:137–80. https://doi.org/10.1016/j.pharmthera.2019.05.011.

    Article  CAS  PubMed  Google Scholar 

  26. Tan WK, Tan ARY, Sivanandam P, Goh EJH, Yap ZP, Saburulla NF, et al. In vitro inhibition of human aldehyde oxidase activity by clinically relevant concentrations of gefitinib and erlotinib: Comparison with select metabolites, molecular docking analysis, and impact on hepatic metabolism of zaleplon and methotrexate. J Pharmacol Exp Ther. 2020;374(2):295–307. https://doi.org/10.1124/jpet.120.265249.

    Article  CAS  PubMed  Google Scholar 

  27. Chen S, Austin-Muttitt K, Zhang LH, Mullins JGL, Lau AJ. In vitro and in silico analyses of the inhibition of human aldehyde oxidase by bazedoxifene, lasofoxifene, and structural analogues. J Pharmacol Exp Ther. 2019;371(1):75–86. https://doi.org/10.1124/jpet.119.259267.

    Article  CAS  PubMed  Google Scholar 

  28. Tunek A, Svensson LA. Bambuterol, a carbamate ester prodrug of terbutaline, as inhibitor of cholinesterases in human blood. Drug Metab Dispos. 1988;16(5):759–64.

    CAS  PubMed  Google Scholar 

  29. Pistolozzi M, Du H, Wei H, Tan W. Stereoselective inhibition of human butyrylcholinesterase by the enantiomers of bambuterol and their intermediates. Drug Metab Dispos. 2015;43(3):344–52. https://doi.org/10.1124/dmd.114.060251.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Authors greatly appreciate the help of Sophia M. Shi in editing the manuscript and many colleagues for their helpful discussion.

Author information

Authors and Affiliations

Authors

Contributions

Substantial contributions to the conception or design of the work; or the acquisition, analysis, or interpretation of data for the work: JL, DV, GB, AC, WB, SJ, LT, JY, YC, GC, MT, and LD. Drafting the work or revising it critically for important intellectual content: JL, DV, WB, SJ, LT, and LD. Final approval of the version to be published: JL, DV, GB, AC, WB, SJ, LT, JY, YC, GC, MT, and LD. Agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved: JL, DV, GB, AC, WB, SJ, LT, JY, YC, GC, MT, and LD.

Corresponding author

Correspondence to Li Di.

Ethics declarations

Conflict of Interest

Authors are employees of Pfizer Inc., New York, NY, USA, and may hold Pfizer stocks.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, J., Vernikovskaya, D., Bora, G. et al. Novel Multiplexed High Throughput Screening of Selective Inhibitors for Drug-Metabolizing Enzymes Using Human Hepatocytes. AAPS J 26, 36 (2024). https://doi.org/10.1208/s12248-024-00908-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12248-024-00908-8

Keywords

Navigation