IN VITRO ASSESSMENT OF REVERSIBLE AND METABOLISM-DEPENDENT INHIBITORY EFFECTS OF PROPOXAZEPAM ON CYP2C8 ACTIVITY

Authors

  • M. GOLOVENKO О.V. Bogatsky Physico-Chemical Institute of the National Academy of Sciences of Ukraine, Odesa, Ukraine
  • I. VALIVODZ О.V. Bogatsky Physico-Chemical Institute of the National Academy of Sciences of Ukraine, Odesa, Ukraine
  • A. REDER SLC «INTERCHEM», Odesa, Ukraine
  • V. LARIONOV О.V. Bogatsky Physico-Chemical Institute of the National Academy of Sciences of Ukraine, Odesa, Ukraine

DOI:

https://doi.org/10.15407/exp-oncology.2025.01.051

Keywords:

cancer, propoxazepam, CYP2C8, montelukast, gemfibrozil O-glucuronide, reversible inhibition, metabolism- dependent inhibition, DDI prediction

Abstract

Background. In oncology, drug-drug interactions (DDIs) are particularly relevant due to the complex medication regimens of cancer patients. These patients often require multiple drugs to manage both their disease and treatment-related side effects. Evaluating potential DDIs via the inhibition of CYP enzymes is crucial in drug discovery. This study aimed to assess the effect of propoxazepam on CYP2C8 activity in vitro by amodiaquine N-deethylation in human liver microsomes and to predict the likelihood of DDI through CYP activity reduction. Materials and Methods. Amodiaquine Ndeethylation was used as a marker of CYP2C8 activity. The positive controls included montelukast (1 μM) for rever sible inhibition and gemfibrozil O-glucuronide (40 μM) for metabolism-dependent inhibition. Propoxazepam was tested in both reversible and metabolism-dependent inhibition conditions being added with the substrate or pre-incubated with microsomes and NADPH, respectively. The metabolite formation was quantified by LC-MS/MS in a multiple reaction monitoring mode using the electrospray ionization technique. Results. Propoxazepam inhibited CYP2C8 activity in a concentration-dependent manner, with IC50 values of 20.5 ± 2.2 μM for reversible inhibition and 23.1 ± 3.2 μM for metabolism-dependent inhibition. Positive controls montelukast and gemfibrozil O-glucuronide showed expected inhibition (4.4% and 12.2% of control, respectively). Propoxazepam showed low binding to microsomal protein under the experimental conditions. Conclusion. Based on the indicators used (Ki, IC50, IC50 shift, and [I]/Ki ratios), propoxazepam is not expected to be a significant CYP2C8 inhibitor in vitro.

References

van den Beuken-van Everdingen MH, Hochstenbach LM, Joosten EA, et al. Update on prevalence of pain in patients with cancer: systematic review and meta-analysis. J Pain Symptom Manage. 2016;51(6):1070-1090. https://doi.org/ 10.1016/j.jpainsymman.2015.12.340

Kudoh A, Katagai H, Takazawa T. Anesthesia with ketamine, propofol, and fentanyl decreases the frequency of postoperative psychosis emergence and confusion in schizophrenic patients. J Clin Anesth. 2002;14(2): 107-110. https://doi.org/ 10.1016/s0952-8180(01)00363-4

Ye SY, Li JY, Li TH, et al. The efficacy and safety of celecoxib in addition to standard cancer therapy: a syste- matic review and meta-analysis of randomized controlled trials. Curr Oncol. 2022;29(9):6137-6153. https://doi.org/ 10.3390/curroncol29090482

Riechelmann RP, Tannock IF, Wang L, et al. Potential drug interactions and duplicate prescriptions among cancer patients. J Natl Cancer Inst. 2007;99(8):592-600. https://doi.org/10.1093/jnci/djk130

McLeod HL. Clinically relevant drug-drug interactions in oncology. Br J Clin Pharmacol. 1998; 45(6):539-544.

Palleria C, Di Paolo A, Giofrè C, et al. Pharmacokinetic drug-drug interaction and their implication in clinical management. J Res Med Sci. 2013;18(7):601-610.

Polasek TM, Lin FPY, Miners JO, Doogue MP. Perpetrators of pharmacokinetic drug-drug interactions arising from altered cytochrome P450 activity: a criteria-based assessment. Br J Clin Pharmacol. 2011;71(5):727-736.

Okuda H, Nishiyama T, Ogura K, et al. Lethal drug interactions of sorivudine, a new antiviral drug, with oral 5-fluorouracil prodrugs. Drug Metab Dispos. 1997;25(5):270-273.

Tornio A, Backman JT. Cytochrome P450 in pharmacogenetics: an update. Adv Pharmacol. 2018;83:151-185. https://doi.org/10.1016/bs.apha.2018.04.007

Hakkola J, Hukkanen J, Turpeinen M, Pelkonen O. Inhibition and induction of CYP enzymes in humans: an up- date. Arch Toxicol. 2020;94:3671-3722. https://doi.org/10.1007/s00204-020-02936-7

van Leeuwen RW, Swart EL, Boven E, et al. Potential drug interactions in cancer therapy: a prevalence study using an advanced screening method. Ann Oncol. 2011;22:2334-2341.

Moscato P, Cortelli L, Chiari L. Physiological responses to pain in cancer patients: A systematic review. Comput Methods Programs Biomed. 2022;217:1-16. https://doi.org/10.1016/j.cmpb.2022.106682

Golovenko MY. Propoxazepam is an innovative analgesic that inhibits acute and chronic pain and has a polymodal mechanism of action. Visn Nac Akad Nauk Ukr. 2021;(4):76-90.

Golovenko NYa, Larionov VB, Reder AS, Valivodz IP. An effector analysis of the interaction of propoxazepam with antagonists of GABA and glycine receptors. Neurochem J. 2017;11(4):302-308. https://doi.org/10.1134/ S1819712417040043

Golovenko MY, Reder A, Zupanets I, et al. Phase I study evaluating the pharmacokinetic profile of a novel oral analgesic propoxazepam. J Pre-Clin Clin Res. 2023;17(3):138-144. https://doi.org/10.26444/jpccr/169426

European Medicines Agency [EMA]. Guideline on the investigation of drug interactions. 21 June 2012. Available from: http://www.ema.europa.eu

Golovenko MYa, Babenko MM, Larionov VB, et al. Research in vitro drug interactions mediated by cytochrome P450 isoenzymes. Ministry of Health of Ukraine. State Expert Center. Kyiv; 2023. 54 p. Available from: https:// www.dec.gov.ua/wp-content/uploads/2023/11/metodrekomendacziyi__doslidzhennya-in-vitro-vzayemodiyi- likarskyh-zasobiv-oposeredkovanoyi-izofermentamy-czytohromu-r450.pdf

Backman JT, Filppula AM, Niemi M, Neuvonen PJ. Role of cytochrome P450 2C8 in drug metabolism and interac- tions. Pharmacol Rev. 2016;68:168-241. https://doi.org/ 10.1124/pr.115.011411

Kim KA, Chung J, Jung DH, Park JY. Identification of cytochrome P450 isoforms involved in the metabolism of loperamide in human liver microsomes. Eur J Clin Pharmacol. 2004;60:575-581.

Reder AS. Dispersed substance 7-bromo-5-(o-chlorophenyl)-3-propiloxy-1,2-dihydro-3H-1,4-benzodiazepine- 2-one (I) with at least 50% volume fraction of particles less than 30 μm for use as anticonvulsive and analgesic drug. Patent of Ukraine UA118626C2, published 11.02.2019.

Aquilante CL, Niemi M, Gong L, et al. PharmGKB summary: very important pharmacogene information for cytochrome P450, family 2, subfamily C, polypeptide 8. Pharmacogenet Genomics. 2013;23:721-728. https://doi. org/10.1097/FPC.0b013e3283653b27.

Golovenko M, Reder A, Larionov V, Andronati S. Metabolic profile and mechanisms reaction of receptor GABA- targeted propoxazepam in human hepatocytes. Biotechnologia Acta. 2022;15(1):25-33. https://doi.org/10.15407/ biotech15.01.043

Walsky RL, Obach RS, Gaman EA, Gleeson JP, Proctor WR. Selective inhibition of human cytochrome P4502C8 by montelukast. Drug Metab Dispos. 2005;33: 413-418. https://doi.org/10.1124/dmd.104.002766

Ma Y, Fu Y, Khojasteh SC, et al. Glucuronides as potential anionic substrates of human cytochrome P450 2C8 (CYP2C8). J Med Chem. 2017;60:8691-8705. https://doi.org/10.1021/acs.jmedchem.7b00510

Austin RP, Barton P, Cockroft SL, et al. The influence of nonspecific microsomal binding on apparent intrinsic clearance, and its prediction from physicochemical properties. Drug Metab Dispos. 2002;30(12):1497-1503. https:// doi.org/10.1124/dmd.30.12.1497.

Riechelmann RR, Girardi D. Drug interactions in cancer patients: A hidden risk? J Res Pharm Pract. 2016;5(2): 77-78. https://doi.org/10.4103/2279-042X.179560

Li XQ, Bjorkman A, Andersson TB, et al. Amodiaquine clearance and its metabolism to N-desethylamodiaquine is mediated by CYP2C8: a new high affinity and turnover enzyme-specific probe substrate. J Pharmacol Exp Ther. 2002;300(2):399-407. https://doi.org/10.1124/jpet.300.2.399

Schenkman JB, Jansson I. Spectral analyses of cytochromes P450. Methods Mol Biol. 2006;320:11-18. https://doi. org/10.1385/1-59259-998-2:11

Golovenko MYa, Larionov VB, Valivodz IP. Spectral characteristics of cytochrome P450 in the interaction with pro- poxazepam and its metabolite. Med Clin Chem. 2023;25(2):12-19. https://doi.org/10.11603/mcch.2410-681X.2023. i2.13854

Larionov VB, Golovenko MYa, Kuzmin VE, et al. Propoxazepam interaction with cytochromes CYP450 isoforms based on molecular docking-analysis. Dopov Nac Akad Nauk Ukr. 2023;(3):96-102. https://doi.org/10.15407/dopo- vidi2023.03.096 (in Ukrainian).

Bachmann KA, Lewis JD. Predicting inhibitory drug-drug interactions and evaluating drug interaction reports us- ing inhibition constants. Ann Pharmacother. 2005;39:1064-1072. https://doi.org/ 10.1345/aph.1E508

VandenBrink BM, Isoherranen N. The role of metabolites in predicting drug-drug interactions: focus on irrevers- ible cytochrome P450 inhibition. Curr Opin Drug Discov Devel. 2010;13:66-77.

Jin C, He X, Zhang F, et al. Inhibitory mechanisms of celastrol on human liver cytochrome P450 1A2, 2C19, 2D6, 2E1 and 3A4. Xenobiotica. 2015;45:571-577. https://doi.org/10.3109/00498254.2014.1003113

Golovenko M, Reder A, Larionov V, et al. Cross-species differential plasma protein binding of propoxazepam, a novel analgesic agent. Biopolymers Cell. 2021;37(6):459-468. https://doi.org/10.7124/bc.000A68

Busti AJ. The inhibitory constant (Ki) and its use in understanding drug interactions. Available from: https://www. ebmconsult.com/articles/inhibitory-constant-ki-drug-interactions. Accessed [25 Aug 2024].

Berry LM, Zhao Z. Dynamic modeling of cytochrome P450 inhibition in vitro: impact of inhibitor depletion on IC50 shift. Drug Metab Dispos. 2013;41(7):1374-1381. https://doi.org/10.1124/dmd.113.051508.

Downloads

Published

11.07.2025

How to Cite

GOLOVENKO, M., VALIVODZ, I., REDER, A., & LARIONOV, V. (2025). IN VITRO ASSESSMENT OF REVERSIBLE AND METABOLISM-DEPENDENT INHIBITORY EFFECTS OF PROPOXAZEPAM ON CYP2C8 ACTIVITY. Experimental Oncology, 47(1), 51–59. https://doi.org/10.15407/exp-oncology.2025.01.051

Issue

Section

Original contributions