Gemfibrozil and its oxidative metabolites: quantification of aglycones, acyl glucuronides, and covalent adducts in samples from preclinical and clinical kinetic studies

https://doi.org/10.1016/S0378-4347(00)00041-4Get rights and content

Abstract

A gradient reversed-phase HPLC analysis for the direct measurement of gemfibrozil (GEM) and four oxidative metabolites in plasma and urine of humans and in tissue homogenates of rats was developed. The corresponding acyl glucuronides and the covalently bound protein adducts (in protein precipitates) were determined after liberation from the respective conjugates via alkaline hydrolysis. The limits of detection for the covalent adducts in human plasma are: 10 ng ml−1 (GEM), 20 ng ml−1 (M1), 0.5 ng ml−1 (M2, M4), and 5 ng ml−1 (M3). The method was validated with respect to selectivity, recovery, linearity, precision, and accuracy. It has been applied to the analysis of preclinical and clinical studies. Pharmacokinetic profiles of gemfibrozil, its metabolites, and covalent adducts in human plasma and rat tissue homogenates are given.

Introduction

Gemfibrozil, a fibrate lipid-regulating agent, is clinically effective in reducing serum cholesterol and triglyceride levels, decreasing LDL and increasing HDL levels [1]. Apart from new HMG CoA reductase inhibitors (e.g., atorvastatin), gemfibrozil is still a drug of choice in the treatment of hyperlipidaemias involving raised triglyceride levels [2] and has been effective in reducing the incidence of coronary heart disease (by 34%) [1]. Extensive phase-I metabolism yields at least four oxidative metabolites – three hydroxylated products (M1, M2, M4) and one dicarboxylic acid (M3) – and subsequent conjugation with glucuronic acid leads to the five respective acyl glucuronides (Fig. 1) [3], [4], [5], [6]. Renal excretion is the most important elimination pathway for the respective carboxylic acids as well as such glucuronides in man. Accordingly, a total of 60–70% of a gemfibrozil dose are found in urine, while only about 6% are recovered in faeces following peroral administration [4], [5]. As to be expected for lower molecular weight compounds, to which, upon glucuronide formation, almost 200 molecular weight units are added, considerable species differences were detected with respect to the extent of renal and biliary/faecal excretion [7], [8]. In rats, further oxidative metabolites were identified in rat urine [9], including a diol metabolite (both ring methyls hydroxylated) and the product of its further metabolism, the acid–alcohol derivative (ortho ring methyl hydroxylated, meta ring methyl further oxidized to acid), as well as ether and acyl (=ester) glucuronides of the same metabolite.

Acyl glucuronides are reactive intermediates which undergo typical chemical reactions such as hydrolysis, isomerization, and covalent binding to endogenous compounds. Hydrolysis of the ester glucuronides at physiological pH leads to back-formation of the aglycones and may be responsible for a potential increase of aglycone concentrations, also ex vivo/in vitro, i.e., after blood or urine sampling. Since the hydrolysis rate is temperature and pH-dependent, post-sampling degradation can be minimized by immediate cooling and pH-stabilization (pH 3–4) [10], [11], which is essential for the correct determination of aglycone and glucuronide concentrations. Isomerization – i.e., migration of the acyl residue from the C-1 position to the C-2, C-3, and C-4 positions of the glucuronic acid ring – leads to the generation of β-glucuronidase-resistant regioisomers. Like hydrolysis, it is pH-dependent and can hence be avoided. Covalent binding has been demonstrated for many acyl glucuronide-forming drugs in vivo [3], [12], [13], [14], [15], [16], [17], in vitro studies have verified the role of the acyl glucuronides in the formation of such covalent adducts [3], [15], [16], [17], [18], [19], which were proven to be formed via two different mechanisms, i.e., by nucleophilic displacement and condensation of the rearranged acyl glucuronide isomers with lysine residues (e.g. Lys-159), as shown for the acyl glucuronide of benoxaprofen [17], [20].

In addition to pH-dependent isomerization and hydrolysis, tissue homogenates include a further complicating source of instability, which is hydrolysis via lysosomal hydrolytic enzymes that are released upon the homogenization process and exhibit their highest activity at acidic pH, i.e. at the pH level where nonenzymatic hydrolysis and isomerization are minimized.

Analytical methods are described in literature for the quantification of gemfibrozil in plasma (HPLC) [21], [22], for gemfibrozil and its main metabolite M3 in plasma and urine (GC [23], HPLC [24]), and for the respective glucuronides (after hydrolysis). Another method, which was developed for the simultaneous determination (HPLC) of gemfibrozil and M1-M4 in plasma and urine requires the use of two detectors and two integrators as well as different stationary and mobile phases for the different biophases (plasma, urine) [4]. Moreover, it was only validated for fairly high plasma concentrations, so that the determination of the usually low concentrations of M2 and M4 was not possible. Furthermore, in the current studies, the respective procedure was expanded to include compounds released from the respective acyl glucuronides and covalent protein adducts.

Thus, aim of the present investigations was (a) to establish a direct gradient HPLC method for the simultaneous determination of gemfibrozil and M1–M4 in concentration ranges relevant for pharmacokinetic studies, and to validate the assay for the measurement of gemfibrozil, its major phase-I and phase-II metabolites, and the respective covalent adducts in different biophases (human plasma and urine, various rat tissues) as well as (b) to apply the methods to the elucidation of the pharmacokinetics of gemfibrozil, all its metabolites, and adducts in humans and rats.

Section snippets

Chemicals and reagents

Gemfibrozil, its metabolites M1–M4 [M1: 5-(4-Hydroxy-2,5-dimethyl-phenoxy)-2,2-dimethyl-pentanoic acid; M2: 5-(5-Hydroxymethyl-2-methyl-phenoxy)-2,2-dimethyl-pentanoic acid; M3: 3-(4-Carboxy-4-methyl-pentyloxy)-4-methyl-benzoic acid; M4: 5-(2-Hydoxymethyl-5-methyl-phenoxy)-2,2-dimethyl-pentanoic acid], and the structural analogues C-2 [4-(2,5-Dimethyl-phenoxy)-2,2-dimethyl-butyric acid] and C-5 [7-(2,5-Dimethyl-phenoxy)-2,2-dimethyl-heptanoic acid] were kindly provided by Parke Davis (Ann

Chromatography

The separation of gemfibrozil, M1–M4, and the used internal standards carried out under the conditions described above yielded appropriate peak separations as depicted in the representative chromatograms shown in Fig. 2. The determined retention times, capacity factors (k′), and resolution factors (Rs) of the compounds are listed in Table 2. Baseline separation was achieved on both, the Zorbax and the Spherisorb column, although the Spherisorb showed an improved performance regarding peak

Conclusions

A sensitive, reliable and accurate analytical method has been established and validated to investigate the kinetics of gemfibrozil, its phase-I and phase-II metabolites, and covalent protein adducts in humans and animals. The suitability of the method to measure pharmacokinetic studies has been confirmed by the application of the method on a preclinical study with rats and clinical trials with hyperlipidaemic and elderly patients. However, it needs to be emphasized again that a complex number

Acknowledgements

These investigations were supported by grants from the Doktor Robert Pfleger-Stiftung Bamberg and the Fonds der Chemischen Industrie Frankfurt/Main (to HSL).

References (31)

  • B.F. Thomas et al.

    Drug Metab. Dispos.

    (1999)
  • B.C. Sallustio et al.

    Biochem. Pharmacol.

    (1991)
  • E.J. Randinitis et al.

    J. Chromatogr.

    (1984)
  • E.J. Randinitis et al.

    J. Chromatogr.

    (1986)
  • M. Wang et al.

    Life Sci.

    (1998)
  • M.H. Frick et al.

    N. Engl. J. Med.

    (1987)
  • P.A. Todd et al.

    Am. J. Cardiol.

    (1985)
  • M.L. Hyneck et al.

    Clin. Pharmacol. Ther.

    (1988)
  • A. Nakagawa et al.

    Biomed. Chromatogr.

    (1991)
  • R.A. Okerholm et al.

    Proc. Roy. Soc. Med.

    (1976)
  • H. Knauf et al.

    Klin. Wochenschr.

    (1990)
  • K.J. Dix et al.

    Drug Metab. Dispos.

    (1999)
  • L. Sabordo et al.

    J. Pharmacol. Exp. Ther.

    (1999)
  • J. Hasegawa et al.

    Drug Metab. Dispos.

    (1982)
  • H. Spahn-Langguth et al.

    Drug Metab. Rev.

    (1992)
  • Cited by (35)

    • Comparative analysis of the removal and transformation of 10 typical pharmaceutical and personal care products in secondary treatment of sewage: A case study of two biological treatment processes

      2022, Journal of Environmental Chemical Engineering
      Citation Excerpt :

      Then, benzoic acid metabolite was obtained after it was cracked by lyase. However, its degradation pathway under anaerobic conditions had not been reported in detail [52–54]. Liu and Wong [30] found that aerobic biodegradation and anaerobic biodegradation had different effects on different types of PPCPs.

    • Biotransformation of trace organic compounds by activated sludge from a biological nutrient removal treatment system

      2016, Bioresource Technology
      Citation Excerpt :

      The EAWAG-BBD PPS predicts that the biotransformation of DEET under aerobic conditions proceeds by the hydrolysis of DEET to 3-methylbenzoate and diethylamine, followed by the metacleavage of 3-methylbenzoate metabolite to produce 2-hydroxy-6-oxo-hepta-2,4 dienoate (Rivera-Cancel et al., 2007). A suggested pathway for gemfibrozil biotransformation is by the hydroxylation of its meta-methyl group to yield a benzyl alcohol which rapidly oxidizes to a benzoic acid metabolite (Hermening et al., 2000). In oxygen deficient conditions, Group 3 TOrCs were observed to desorb over time from anaerobic (triclosan) and anoxic (gemfibrozil, naproxen, ibuprofen, and triclosan) sludge solids (Figs. 2 and S4).

    • Performance of suspended and attached growth bioreactors for the removal of cationic and anionic pharmaceuticals

      2016, Chemical Engineering Journal
      Citation Excerpt :

      Gemfibrozil contains aromatic-aliphatic ether and the degradation occurs by the cleavage of ether, thereby forming phenol derivative and aldehyde as reported elsewhere [43]. Degradation can also be due to the conversion of aromatic methyl group to alcohol [44]. Presence of additional carbon source increase the biomass activity and therefore increases the pharmaceutical degradation [45].

    • Eco-directed sustainable prescribing: Feasibility for reducing water contamination by drugs

      2014, Science of the Total Environment
      Citation Excerpt :

      Despite the ready availability of limited PK data for drugs, comprehensive PK data (sufficient to estimate API levels that would reach the environment after metabolism) can be surprisingly difficult to locate; the pharmacokinetics for many drugs are still not even sufficiently understood. This is because PK data needed for clinical trials and drug registration purposes do not need to account for the portion of a dose that passes directly through the gut unabsorbed and unmetabolized (sometimes exceeding the majority of a dose) or for reversible metabolic conjugates (those that can undergo deglucuronidation, via microbial or abiotic hydrolysis) versus total conjugates, which include non-reversible conjugates formed from phase I metabolites; conjugates of phase I metabolites do not yield the parent API upon hydrolysis (Hermening et al., 2000). The data cited in studies involving predicted environmental concentrations (PECs) for APIs often simply state that an API is “extensively metabolized” or “extensively excreted” and are insufficient to rule out whether the API has potential for occurrence in the environment via excretion.

    • Mechanistic role of acyl glucuronides

      2013, Drug-Induced Liver Disease
    View all citing articles on Scopus
    View full text