Skip to main content
Log in

Study of the roles of cytochrome P450 (CYPs) in the metabolism and cytotoxicity of perhexiline

  • Toxicokinetics and Metabolism
  • Published:
Archives of Toxicology Aims and scope Submit manuscript

Abstract

Perhexiline is a prophylactic antianginal agent developed in the 1970s. Although, therapeutically, it remained a success, the concerns of its severe adverse effects including hepatotoxicity caused the restricted use of the drug, and eventually its withdrawal from the market in multiple countries. In the clinical setting, cytochrome P450 (CYP) 2D6 is considered as a possible risk factor for the adverse effects of perhexiline. However, the role of CYP-mediated metabolism in the toxicity of perhexiline, particularly in the intact cells, remains unclear. Using our previously established HepG2 cell lines that individually express 14 CYPs (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5, and 3A7) and human liver microsomes, we identified that CYP2D6 plays a major role in the hydroxylation of perhexiline. We also determined that CYP1A2, 2C19, and 3A4 contribute to the metabolism of perhexiline. The toxic effect of perhexiline was reduced significantly in CYP2D6-overexpressing HepG2 cells, in comparison to the control cells. In contrast, overexpression of CYP1A2, 2C19, and 3A4 did not show a significant protective effect against the toxicity of perhexiline. Pre-incubation with quinidine, a well-recognized CYP2D6 inhibitor, significantly attenuated the protective effect in CYP2D6-overexpressing HepG2 cells. Furthermore, perhexiline-induced mitochondrial damage, apoptosis, and ER stress were also attenuated in CYP2D6-overexpressing HepG2 cells. These findings suggest that CYP2D6-mediated metabolism protects the cells from perhexiline-induced cytotoxicity and support the clinical observation that CYP2D6 poor metabolizers may have higher risk for perhexiline-induced hepatotoxicity.

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

Similar content being viewed by others

References

  • Amoah AGB, Gould BJ, Parke DV, Lockhart JDF (1986) Further studies on the pharmacokinetics of perhexiline maleate in humans. Xenobiotica 16(1):63–68

    Article  CAS  Google Scholar 

  • Ashrafian H, Horowitz JD, Frenneaux MP (2007) Perhexiline. Cardiovasc Drug Rev 25(1):76–97

    Article  CAS  Google Scholar 

  • Chen S, Xuan J, Wan L, Lin H, Couch L, Mei N, Dobrovolsky VN, Guo L (2014) Sertraline, an antidepressant, induces apoptosis in hepatic cells through the mitogen-activated protein kinase pathway. Toxicol Sci 137(2):404–415

    Article  CAS  Google Scholar 

  • Chen M, Suzuki A, Borlak J, Andrade RJ, Lucena MI (2015) Drug-induced liver injury: interactions between drug properties and host factors. J Hepatol 63(2):503–514

    Article  CAS  Google Scholar 

  • Chen S, Ren Z, Yu D, Ning B, Guo L (2018a) DNA damage-induced apoptosis and mitogen-activated protein kinase pathway contribute to the toxicity of dronedarone in hepatic cells. Environ Mol Mutagen 59(4):278–289

    Article  CAS  Google Scholar 

  • Chen S, Wu Q, Ning B, Bryant M, Guo L (2018b) The role of hepatic cytochrome P450s in the cytotoxicity of dronedarone. Arch Toxicol 92(6):1969–1981

    Article  CAS  Google Scholar 

  • Chen S, Wu Q, Li X, Li D, Fan M, Ren Z, Bryant M, Mei N, Ning B, Guo L (2020) The role of hepatic cytochrome P450s in the cytotoxicity of sertraline. Arch Toxicol 94(7):2401–2411

    Article  Google Scholar 

  • Chen S, Wu Q, Li X, Li D, Mei N, Ning B, Puig M, Ren Z, Tolleson WH, Guo L (2021) Characterization of cytochrome P450s (CYP)-overexpressing HepG2 cells for assessing drug and chemical-induced liver toxicity. J Environ Sci Health C Toxicol Carcinog 39(1):68–86

    CAS  PubMed  PubMed Central  Google Scholar 

  • Cooper RG, Evans DA, Whibley EJ (1984) Polymorphic hydroxylation of perhexiline maleate in man. J Med Genet 21(1):27–33

    Article  CAS  Google Scholar 

  • Cooper RG, Evans DA, Price AH (1987) Studies on the metabolism of perhexiline in man. Eur J Clin Pharmacol 32(6):569–576

    Article  CAS  Google Scholar 

  • Davies BJ, Coller JK, James HM, Somogyi AA, Horowitz JD, Sallustio BC (2006) The influence of CYP2D6 genotype on trough plasma perhexiline and cis-OH-perhexiline concentrations following a standard loading regimen in patients with myocardial ischaemia. Br J Clin Pharmacol 61(3):321–325

    Article  CAS  Google Scholar 

  • Davies BJ, Coller JK, Somogyi AA, Milne RW, Sallustio BC (2007) CYP2B6, CYP2D6, and CYP3A4 catalyze the primary oxidative metabolism of perhexiline enantiomers by human liver microsomes. Drug Metab Dispos 35(1):128–138

    Article  CAS  Google Scholar 

  • Davies BJ, Herbert MK, Coller JK, Somogyi AA, Milne RW, Sallustio BC (2008) Steady-state pharmacokinetics of the enantiomers of perhexiline in CYP2D6 poor and extensive metabolizers administered Rac-perhexiline. Br J Clin Pharmacol 65(3):347–354

    Article  CAS  Google Scholar 

  • Dhakal B, Li CMY, Li R, Yeo K, Wright JA, Gieniec KA, Vrbanac L, Sammour T, Lawrence M, Thomas M, Lewis M, Perry J, Worthley DL, Woods SL, Drew P, Sallustio BC, Smith E, Horowitz JD, Maddern GJ, Licari G, Fenix K (2022) The antianginal drug perhexiline displays cytotoxicity against colorectal cancer cells in vitro: a potential for drug repurposing. Cancers 14(4):1043

    Article  CAS  Google Scholar 

  • He Y, Zhu L, Ma J, Lin G (2021) Metabolism-mediated cytotoxicity and genotoxicity of pyrrolizidine alkaloids. Arch Toxicol 95(6):1917–1942

    Article  CAS  Google Scholar 

  • Hofman J, Sorf A, Vagiannis D, Sucha S, Kammerer S, Küpper JH, Chen S, Guo L, Ceckova M, Staud F (2019) Brivanib exhibits potential for pharmacokinetic drug-drug interactions and the modulation of multidrug resistance through the inhibition of human ABCG2 drug efflux transporter and CYP450 biotransformation enzymes. Mol Pharm 16(11):4436–4450

    Article  CAS  Google Scholar 

  • Hofman J, Vagiannis D, Chen S, Guo L (2021) Roles of CYP3A4, CYP3A5 and CYP2C8 drug-metabolizing enzymes in cellular cytostatic resistance. Chem Biol Interac 340:109448

    Article  CAS  Google Scholar 

  • Inglis S, Stewart S (2006) Metabolic therapeutics in angina pectoris: history revisited with perhexiline. Eur J Cardiovasc Nurs 5(2):175–184

    Article  Google Scholar 

  • MacKenzie KR, Zhao M, Barzi M, Wang J, Bissig K-D, Maletic-Savatic M, Jung SY, Li F (2020) Metabolic profiling of norepinephrine reuptake inhibitor atomoxetine. Eur J Pharm Sci 153:105488

    Article  CAS  Google Scholar 

  • Marroquin LD, Hynes J, Dykens JA, Jamieson JD, Will Y (2007) Circumventing the Crabtree effect: replacing media glucose with galactose increases susceptibility of HepG2 cells to mitochondrial toxicants. Toxicol Sci 97(2):539–547

    Article  CAS  Google Scholar 

  • Midei MG, Darpo B, Ayers G, Brown R, Couderc J-P, Daly W, Ferber G, Sager PT, Camm AJ (2021) Electrophysiological and ECG effects of perhexiline, a mixed cardiac ion channel inhibitor, evaluated in nonclinical assays and in healthy subjects. J Clin Pharmacol 61(12):1606–1617

    Article  CAS  Google Scholar 

  • Mueller-Schoell A, Michelet R, Weinelt F, Kloft C, Mikus G (2021) CYP2D6 phenotype explains reported yohimbine concentrations in four severe acute intoxications. Arch Toxicol 95(8):2867–2870

    Article  CAS  Google Scholar 

  • Ren Z, Chen S, Zhang J, Doshi U, Li AP, Guo L (2016) Endoplasmic reticulum stress induction and ERK1/2 activation contribute to nefazodone-induced toxicity in hepatic cells. Toxicol Sci 154(2):368–380

    Article  CAS  Google Scholar 

  • Ren Z, Chen S, Seo JE, Guo X, Li D, Ning B, Guo L (2020) Mitochondrial dysfunction and apoptosis underlie the hepatotoxicity of perhexiline. Toxicol in Vitro 34:104987

    Article  Google Scholar 

  • Ren Z, Chen S, Pak S, Guo L (2021) A mechanism of perhexiline’s cytotoxicity in hepatic cells involves endoplasmic reticulum stress and p38 signaling pathway. Chem Biol Interac 334:109353

    Article  CAS  Google Scholar 

  • Sallustio BC, Westley IS, Morris RG (2002) Pharmacokinetics of the antianginal agent perhexiline: relationship between metabolic ratio and steady-state dose. Br J Clin Pharmacol 54(2):107–114

    Article  CAS  Google Scholar 

  • Shah RR, Oates NS, Idle JR, Smith RL, Lockhart JD (1982) Impaired oxidation of debrisoquine in patients with perhexiline neuropathy. Br Med J (clin Res Ed) 284(6312):295–299

    Article  CAS  Google Scholar 

  • Sørensen LB, Sørensen RN, Miners JO, Somogyi AA, Grgurinovich N, Birkett DJ (2003) Polymorphic hydroxylation of perhexiline in vitro. Br J Clin Pharmacol 55(6):635–638

    Article  Google Scholar 

  • Todorović Vukotić N, Đorđević J, Pejić S, Đorđević N, Pajović SB (2021) Antidepressants- and antipsychotics-induced hepatotoxicity. Arch Toxicol 95(3):767–789

    Article  Google Scholar 

  • Vagiannis D, Novotna E, Skarka A, Kammerer S, Küpper J-H, Chen S, Guo L, Staud F, Hofman J (2020) Ensartinib (X-396) effectively modulates pharmacokinetic resistance mediated by ABCB1 and ABCG2 drug efflux transporters and CYP3A4 biotransformation enzyme. Cancers 12(4):813

    Article  CAS  Google Scholar 

  • Wu Q, Ning B, Xuan J, Ren Z, Guo L, Bryant MS (2016) The role of CYP 3A4 and 1A1 in amiodarone-induced hepatocellular toxicity. Toxicol Lett 253:55–62

    Article  CAS  Google Scholar 

  • Xuan J, Chen S, Ning B, Tolleson WH, Guo L (2016) Development of HepG2-derived cells expressing cytochrome P450s for assessing metabolism-associated drug-induced liver toxicity. Chem Biol Interact 255:63–73

    Article  CAS  Google Scholar 

Download references

Funding

This study was funded by the National Center for Toxicological Research/U.S. Food and Drug Administration. Drs. Feng Li and Xuan Qin were supported by the National Institute of Diabetes and Digestive and Kidney Diseases (R01-DK121970) and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (R61HD099995) to Feng Li.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lei Guo.

Ethics declarations

Conflicts of interest

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

204_2022_3369_MOESM1_ESM.pptx

Supplementary file 1. Figs. S1 CYP2D6-mediated metabolism reduces the mitochondrial damage, apoptosis and ER stress induced by perhexiline. (A) CYP2D6-overexpressing HepG2 cells and EV control cells were changed into glucose-containing media or galactose-containing media and exposed to various concentrations of perhexiline for 24 h, and cell viability was measured. *p < 0.05 for comparison of cell viability in glucose medium and galactose medium within the same type of cell for the same concentration of perhexiline. (B and C) CYP2D6-overexpressing HepG2 cells and EV-control cells were exposed to 5, 7.5, or 10 μM perhexiline for 24 h and mitochondrial membrane potential by JC-1 staining (B) and caspase 3/7 activity (C) were measured. *p < 0.05 compared to EV control. (D) CYP2D6-overexpressing HepG2 cells and EV control cells were exposed to 10 μM perhexiline or DMSO for 24 h and mRNA levels of ER stress markers were measured using quantitative real-time PCR. (E) The protein levels of CHOP in EV control and CYP2D6-overexpressing HepG2 cells upon perhexiline treatment for 24 h were assessed using Western blots. GAPDH protein levels were used as internal control. The results shown are mean ± S.D. from 3 biological replicates. *p < 0.05 for comparison between perhexiline treatment and DMSO treatment in EV control cells. #p < 0.05 for comparison between perhexiline treated CYP2D6-overexpressing HepG2 cells and EV control cells. Fig. S2. CYP3A4-mediated metabolism marginally attenuates perhexiline-induced mitochondrial damage (A). CYP3A4-overexpressing HepG2 cells were changed into glucose-containing media or galactose-containing media and exposed to various concentrations of perhexiline for 24 h, and cell viability was measured. *p < 0.05 for comparison of cell viability in glucose medium and galactose medium for the same concentration of perhexiline treatment. (B and C) CYP3A4-overexpressing HepG2 cells and EV-control cells were exposed to 5–10 μM perhexiline for 24 h and mitochondrial membrane potential by JC-1 staining (B) and caspase 3/7 activity (C) were measured. The results shown are mean ± S.D. from 3 biological replicates. *p < 0.05 compared to EV control. Fig. S3. Generation of M1 and M3 in CYP2D6- and CYP3A4-overexpressing HepG2 cells upon 24 h exposure to perhexiline. CYP2D6- and 3A4-overexpressing HepG2 cell and EV control cells were treated with 5 μM perhexiline for 24 h. The cell extracts were then subjected to mass spectrometry to determine the abundance of the two types of trans-OH-Phx, M1 and M3, in CYP2D6 and CYP3A4 cells. The relative abundance of M1 and M3, measured as peak areas, were shown. The results shown are mean ± S.D. from 4 independent experiments. Fig. S4. CYP2B6-overexpressing HepG2 cells do not show protective effect against the cytotoxicity of perhexiline. CYP2B6-overexpressing HepG2 cells were exposed to 2.5 – 10 μM perhexiline for 24 h and the cytotoxicity of perhexiline was measured using cellular ATP contents (A) and LDH release (B). The results shown are mean ± S.D. from 3 biological replicates (PPTX 233 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ren, Z., Chen, S., Qin, X. et al. Study of the roles of cytochrome P450 (CYPs) in the metabolism and cytotoxicity of perhexiline. Arch Toxicol 96, 3219–3231 (2022). https://doi.org/10.1007/s00204-022-03369-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00204-022-03369-0

Keywords

Navigation