Skip to main content
Log in

Combined contributions of cytochrome P450s (CYPs) and non-enzymatic metabolism in the in vitro biotransformation of anaprazole, a novel proton pump inhibitor

  • Research
  • Published:
Naunyn-Schmiedeberg's Archives of Pharmacology Aims and scope Submit manuscript

Abstract

Anaprazole, a new proton pump inhibitor (PPI), is designed for the treatment of acid-related diseases, such as gastric ulcers and gastroesophageal reflux. This study explored the in vitro metabolic transformation of anaprazole. The metabolic stabilities of anaprazole in human plasma and human liver microsomes (HLM) were analyzed by liquid chromatography-tandem mass spectrometry (LC–MS/MS). Then, the contribution (%) of non-enzymatic and cytochrome P450s (CYPs) enzyme-mediated anaprazole metabolism was assessed. To obtain the metabolic pathways of anaprazole, the metabolites generated in HLM, thermal deactivated HLM, and cDNA-expressed recombinant CYPs incubation systems were identified by ultra-performance liquid chromatography/quadrupole-time-of-flight mass spectrometry (UPLC/Q-TOF–MS). Results showed that anaprazole was very stable in human plasma and unstable in HLM. The contribution (%) of non-enzymatic vs. CYPs enzyme-mediated metabolism was 49% vs. 51%. CYP3A4 was the major enzyme (48.3%), followed by CYP2C9 (17.7%) and CYP2C8 (12.3%), in responsible for the metabolism of anaprazole. Specific chemical inhibitors targeting CYP enzymes notably blocked the metabolic transformation of anaprazole. Six metabolites of anaprazole were identified in the non-enzymatic system, whereas 17 metabolites were generated in HLM. The biotransformation reactions mainly included sulfoxide reduction to thioether, sulfoxide oxidation to sulfone, deoxidation, dehydrogenation, O-dealkylation or O-demethylation of thioether, O-demethylation and dehydrogenation of thioether, O-dealkylation and dehydrogenation of thioether, thioether O-dealkylation and dehydrogenation of thioether, and O-dealkylation of sulfone. Both enzymatic and non-enzymatic metabolisms contribute to the clearance of anaprazole in human. Anaprazole is less likely to develop drug-drug interactions in clinical use compared to other PPIs.

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

Similar content being viewed by others

Data availability

The datasets used or analyzed during the current study are available from the corresponding author on reasonable request.

References

  • Bertilsson L (1995) Geographical/interracial differences in polymorphic drug oxidation Current state of knowledge of cytochromes P450 (CYP) 2D6 and 2C19. Clin Pharmacokinet 29(3):192–209

    Article  CAS  PubMed  Google Scholar 

  • Blume H et al (2006) Pharmacokinetic drug interaction profiles of proton pump inhibitors. Drug Saf 29(9):769–784

    Article  CAS  PubMed  Google Scholar 

  • Chaudhry AS, Kochhar R, Kohli KK (2008) Genetic polymorphism of CYP2C19 & therapeutic response to proton pump inhibitors. Indian J Med Res 127(6):521–530

    CAS  PubMed  Google Scholar 

  • Chawla PK et al (2015) Correlation of CYP2C19 genotype with plasma voriconazole levels: a preliminary retrospective study in Indians. Int J Clin Pharm 37(5):925–930

    Article  CAS  PubMed  Google Scholar 

  • Cheng DX et al (2016) Determination of anaprazole in human plasma by LC-MS/MS in pharmacokinetic study. Yao Xue Xue Bao 51(12):1885–1890

    PubMed  Google Scholar 

  • Croom EL, Wallace AD, Hodgson E (2010) Human variation in CYP-specific chlorpyrifos metabolism. Toxicology 276(3):184–191

    Article  CAS  PubMed  Google Scholar 

  • Davies B, Morris T (1993) Physiological parameters in laboratory animals and humans. Pharm Res 10(7):1093–1095

    Article  CAS  PubMed  Google Scholar 

  • De Morais SM et al (1994) Identification of a new genetic defect responsible for the polymorphism of (S)-mephenytoin metabolism in Japanese. Mol Pharmacol 46(4):594–598

    PubMed  Google Scholar 

  • Denisenko NP et al (2018) CYP3A and CYP2C19 activity in urine in relation to CYP3A4, CYP3A5, and CYP2C19 polymorphisms in Russian peptic ulcer patients taking omeprazole. Pharmgenomics Pers Med 11:107–112

    CAS  PubMed  PubMed Central  Google Scholar 

  • El Rouby N, Lima JJ, Johnson JA (2018) Proton pump inhibitors: from CYP2C19 pharmacogenetics to precision medicine. Expert Opin Drug Metab Toxicol 14(4):447–460

    Article  PubMed  PubMed Central  Google Scholar 

  • Fock KM et al (2008) Proton pump inhibitors. Clin Pharmacokinet 47(1):1–6

    Article  CAS  PubMed  Google Scholar 

  • Gao W et al (2021) Different dose of new generation proton pump inhibitors for the treatment of Helicobacter pylori infection: a meta-analysis. Int J Immunopathol Pharmacol 35:20587384211030396

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Gyawali CP (2017) Proton pump inhibitors in gastroesophageal reflux disease: friend or foe. Curr Gastroenterol Rep 19(9):46

    Article  PubMed  Google Scholar 

  • Harris DM et al (2021) Use of pharmacogenomics to guide proton pump inhibitor therapy in clinical practice. Dig Dis Sci 66(12):4120–4127

    Article  PubMed  Google Scholar 

  • Ishizaki T, Horai Y (1999) Review article: cytochrome P450 and the metabolism of proton pump inhibitors–emphasis on rabeprazole. Aliment Pharmacol Ther 13(Suppl 3):27–36

    Article  CAS  PubMed  Google Scholar 

  • Klotz U, Schwab M, Treiber G (2004) CYP2C19 polymorphism and proton pump inhibitors. Basic Clin Pharmacol Toxicol 95(1):2–8

    Article  CAS  PubMed  Google Scholar 

  • Li XQ et al (2004) Comparison of inhibitory effects of the proton pump-inhibiting drugs omeprazole, esomeprazole, lansoprazole, pantoprazole, and rabeprazole on human cytochrome P450 activities. Drug Metab Dispos 32(8):821–827

    Article  CAS  PubMed  Google Scholar 

  • Lima JJ et al (2021) Clinical pharmacogenetics implementation consortium (CPIC) guideline for CYP2C19 and proton pump inhibitor dosing. Clin Pharmacol Ther 109(6):1417–1423

    Article  PubMed  Google Scholar 

  • McColl KE, Kennerley P (2002) Proton pump inhibitors–differences emerge in hepatic metabolism. Dig Liver Dis 34(7):461–467

    Article  CAS  PubMed  Google Scholar 

  • Mori H, Suzuki H (2019) Role of acid suppression in acid-related diseases: proton pump inhibitor and potassium-competitive acid blocker. J Neurogastroenterol Motil 25(1):6–14

    Article  PubMed  PubMed Central  Google Scholar 

  • Neumann I, et al (2013) Comparison of different regimens of proton pump inhibitors for acute peptic ulcer bleeding. Cochrane Database Syst Rev (6):CD007999

  • Ogawa R, Echizen H (2010) Drug-drug interaction profiles of proton pump inhibitors. Clin Pharmacokinet 49(8):509–533

    Article  CAS  PubMed  Google Scholar 

  • Pallotta S, Pace F, Marelli S (2008) Rabeprazole: a second-generation proton pump inhibitor in the treatment of acid-related disease. Expert Rev Gastroenterol Hepatol 2(4):509–522

    Article  CAS  PubMed  Google Scholar 

  • Ray WA et al (2018) Association of oral anticoagulants and proton pump inhibitor cotherapy with hospitalization for upper gastrointestinal tract bleeding. JAMA 320(21):2221–2230

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Riu-Viladoms G et al (2019) Drug interactions with oral antineoplastic drugs: the role of the pharmacist. Eur J Cancer Care (engl) 28(1):e12944

    Article  PubMed  Google Scholar 

  • Rodrigues AD (1999) Integrated cytochrome P450 reaction phenotyping attempting to bridge the gap between cDAN-expressed cytochromes P450 and native human liver microsome. Biochem Pharmacol 57:465–480. https://doi.org/10.1016/s0006-2952(98)00268-8

    Article  CAS  PubMed  Google Scholar 

  • Sakaguchi M et al (2012) Comparison of PPIs and H2-receptor antagonists plus prokinetics for dysmotility-like dyspepsia. World J Gastroenterol 18(13):1517–1524

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Shi S, Klotz U (2008) Proton pump inhibitors: an update of their clinical use and pharmacokinetics. Eur J Clin Pharmacol 64(10):935–951

    Article  CAS  PubMed  Google Scholar 

  • Strand DS, Kim D, Peura DA (2017) 25 years of proton pump inhibitors: a comprehensive review. Gut Liver 11(1):27–37

    Article  CAS  PubMed  Google Scholar 

  • Tang C et al (2020) Qualitative and quantitative determination of anaprazole and its major metabolites in human plasma. J Pharm Biomed Anal 183:113146

    Article  CAS  PubMed  Google Scholar 

  • Tytgat GN (2001) Shortcomings of the first-generation proton pump inhibitors. Eur J Gastroenterol Hepatol 13(Suppl 1):S29-33

    PubMed  Google Scholar 

  • Vaezi MF, Yang YX, Howden CW (2017) Complications of proton pump inhibitor therapy. Gastroenterology 153(1):35–48

    Article  CAS  PubMed  Google Scholar 

  • Wedemeyer RS, Blume H (2014) Pharmacokinetic drug interaction profiles of proton pump inhibitors: an update. Drug Saf 37(4):201–211

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhu HPX, Zhang L, Sun H, Fan H, Pan Z, Huang C, Shi Z, Ding J, Wang Q, Du Y, Lyu N, Li Z (2022) Effect and safety of anaprazole in the treatment of duodenal ulcers: a randomized, rabeprazole-controlled, phase III non-inferiority study. Chin Med J (Engl) 12(24)

Download references

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Fei Liu, Yanjun Xu, and Xiaomei Zhuang; methodology and data analysis: Fei Liu, Li Wang, Zhen Zhang, and Xifeng Ma; writing—original draft preparation: Fei Liu; writing—review and editing: Xiaomei Zhuang. All authors have read and agreed to the published version of the manuscript. The authors confirm that all data were generated in-house and that no paper mill was used.

Corresponding author

Correspondence to Xiaomei Zhuang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethical approval

No applicable.

Conflict of interest

The authors declare no competing interests.

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.

Supplementary file1 (DOCX 3388 KB)

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, F., Xu, Y., Wang, L. et al. Combined contributions of cytochrome P450s (CYPs) and non-enzymatic metabolism in the in vitro biotransformation of anaprazole, a novel proton pump inhibitor. Naunyn-Schmiedeberg's Arch Pharmacol 396, 1759–1771 (2023). https://doi.org/10.1007/s00210-023-02415-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00210-023-02415-7

Keywords

Navigation