Abstract
Aims
Our objective was to elucidate further the underlying mechanism responsible for therapeutic failures observed with concomitant administration of the oral contraceptive 17α-ethinyloestradiol (EE2) and rifampicin.
Methods
We investigated both oxidative and direct conjugative [3H]-EE2 metabolism by human liver S9 fraction and the effect of known enzyme-inducing drugs using a human hepatocyte induction model in vitro.
Results
Cofactor dependent [3H]-EE2 metabolism by human liver S9 fraction produced 2-hydroxy- [3H]-EE2, 2-methoxy- [3H]-EE2, and direct [3H]-EE2sulphate and glucuronide conjugates. Only two detectable metabolites of [3H]-EE2 were produced by the S9 fraction in the presence of all cofactors: [3H]-EE2–3-sulphate (75.7±7.6% s.d.) and 2-methoxy-3H-EE2 (2.6%±0.5% s.d.). Human hepatocytes extensively metabolized [3H]-EE2 to its glucuronide and sulphate conjugates. Small amounts of a 2-methoxy- [3H]-EE2 3-conjugate, ≤10%, was observed but no. 2-hydroxy- [3H]-EE2 was detected. An unexpected finding in our study was increased [3H]-EE2–3-sulphate production (1.5–3.3 fold, n =3 donor livers) by hepatocytes pretreated with rifampicin compared to control hepatocytes. No statistically significant increase in [3H]-EE2–3-sulphation was observed in hepatocytes pretreated with 3-methylcholanthrene, phenobarbitone, dexamethasone, or omeprazole over nontreated hepatocytes. To our knowledge, this is the first observation of sulphotransferase induction by rifampicin in human hepatocytes in vitro resulting in increased [3H]-EE2sulphation.
Conclusions
Our data indicate that the major EE2 metabolic products formed by human hepatocytes in vitro are direct EE2 conjugates with EE2 oxidation representing minor pathways. Further studies are required to establish the mechanism of sulphotransferase induction and the clinical relevance of our findings.
Keywords: conjugation, drug–drug interactions, ethinyloestradiol, glucuronidation, hepatocytes, human, induction, metabolism, sulphation
Introduction
Co-administration of certain drugs with oral contraceptives containing 50 μg or less of 17α-ethinyloestradiol; 17α-ethynyl-1,3,5 [10]-estratriene-3,17β-diol; EE2 [1] have resulted in menstrual bleeding irregularities and pregnancies. Much of the evidence reported for oral contraceptive failure with concurrent drug use is anecdotal and there is considerable controversy concerning which drugs are involved, the mechanism and clinical relevance of this type of drug–drug interaction [2–6].
There are two main mechanisms by which concurrent drug use are believed to reduce oral contraceptive steroid efficacy; enzyme-induction and/or impaired enterohepatic circulation. The two drug classes most commonly implicated are anticonvulsants and certain antibiotics [2–4, 7, 8]. Following administration of radioactive EE2 to humans, the drug is primarily recovered in bile or urine as glucuronide or sulphate conjugates (Figure 1). Urinary recovery of EE2 derived radioactivity reported ranges from 16 to 54% of an EE2 dose [9–15] and from 28 to 59% is excreted in the faeces [12–15]. Ring ‘A’ hydroxylation, of which 2-hydroxy-EE2 is probably the most important primary oxidative metabolite, can be followed by methylation by catechol-O-methyltransferase to form a secondary metabolite, 2-methoxy-EE2. These two metabolites are excreted in urine and bile as both glucuronide and sulphate conjugates but EE2–3-glucuronide and EE2–3-sulphate are the predominant species found in these two body fluids following an EE2 dose [9, 16, 17].
Figure 1.
Major human ethinyloestradiol (EE2) metabolic pathways.
There is considerable evidence that the antibiotics rifampicin [18–20] and rifabutin [21] and the anticonvulsant drugs phenytoin [22], phenobarbitone [23], carbamazepine [22] and oxcarbazepine [24] induce metabolism of EE2 and significantly lower oestrogen plasma concentrations in humans. However, none of these studies reported the metabolic products produced. Individual CYP450 human liver enzymes reported to oxidize EE2 include: CYP3A, CYP2C, and CYP2E [25, 26]. The work of Guengerich [25] highlighted CYP3A4, known to be induced by numerous drugs, as the major CYP450 enzyme responsible for EE2 oxidation. Although EE2 is known to be predominantly excreted in urine and bile as EE2–3-glucuronide and EE2–3-sulphate, to our knowledge no human liver studies in vitro have been reported concerning the relative effects of drugs known to induce liver enzymes on the individual parallel human EE2 metabolic pathways shown in Figure 1.
Human primary hepatocytes represent a relevant experimental system for the evaluation of pharmacokinetic drug–drug interactions. In our laboratory human hepatocytes are routinely used as an experimental model for toxicology, metabolism and pharmacokinetic drug interactions [27–30]. We have previously reported that treatment of primary human hepatocytes with rifampicin led to duration and dose-dependent induction of the rate of CYP3A-dependent metabolic pathways: lignocaine dealkylation [31] and testosterone 6β-hydroxylation [30].
In this report the disposition of [3H]-EE2 was investigated in human liver S9 fractions, freshly isolated human hepatocytes, and freshly isolated hepatocytes following exposure to the known P450 enzyme inducer drugs rifampicin (CYP3A4, CYP2C9 and CYP2C19), phenobarbitone (CYP3A4), omeprazole (CYP1A2), methylcholanthrene (CYP1A2), and dexamethasone (CYP3 A4) [32] in vitro. Unlike most previous human liver studies in vitro which used EE2 at μm concentrations, our study used therapeutically relevant [3H]-EE2 (1 nm) concentrations [21, 24]. In addition we report the results of [3H]-EE2 metabolism at clinically relevant concentrations by human liver S9 fractions in the presence or absence of several individual cofactors.
This study was presented in part at the 8th North American ISSX Meeting, Hilton Head, SC, October 26–30, 1997.
Methods
Chemicals
Vitrogen was obtained from Collagen Corp., Palo Alto, CA. Fungizone, l-glutamine, and phenol red were procured from Life Technologies, Inc., Grand Island, NY. The BCA Protein Assay Reagent™kit (232225), containing Pierce working solution, was purchased from Pierce, Rockford IL. 2α-hydroxytestosterone, 7β-hydroxytestosterone, 16α-hydroxytestosterone, 2β- hydroxytestosterone, 6β-hydroxytestosterone, 16β-hydroxytestosterone, and androstenedione were obtained from Steraloids, Inc., Eilton, NH. Collagenase was purchased from Worthington Biochemicals, Freehold, NJ. 17α- [6,7– [3H](N)]-ethinyloestradiol ( [3H]-EE2), Sp. Ac. =49.1 Ci mmol−1 was obtained from New England Nuclear, MA. Reference EE2 metabolites: 2-hydroxy-EE2, 2-methoxy-EE2, EE2–3-sulphate, EE2–3,17-di-sulphate, EE2–17-sulphate, and EE2–17- glucuronide were kindly provided by Dr W. Slikker, National Center for Toxicology (NCTR), AK. All other chemicals were obtained from Sigma Chemical Co., St Louis MO.
Human liver source
Human donor livers unsuitable for transplantation used in this study were acquired from the International Institute for the Advancement of Medicine (IIAM, coordinated from Exton, PA) or the Washington Transplant Consortium (Washington DC). The human donor demographics and medical histories are listed in Table 1.
Table 1.
Human donor demographics and medical history.
Preparation of human liver hepatocytes and S9 fractions
Human hepatocytes were isolated according to the two-step collagenase perfusion procedure of Li et al. [33]. Isolated hepatocytes were used either as suspension cultures for metabolism studies or as monolayer cultures for enzyme induction studies. Human liver homogenates were centrifuged at 9000 g according to the methods described [34, 35]. The resulting supernatant S9 fraction containing both cytosolic and microsomal enzymes was aliquoted and stored frozen at −70° C until used.
Human liver S9 fraction incubations
Human liver S9 fractions prepared from donor livers 1, 2, 3, and 4 were incubated in the presence of individual cofactors. Each S9 incubation contained 1 mg S9 protein and [3H]-EE2 (1 nm, 9.2×104 d min−1). The following cofactors were added to individual incubations: NADPH generating system [glucose-6-phosphate (10 mm), NADP+ (1 mm) and glucose-6-phosphate dehydrogenase (1.2 units ml−1)]; SAM (1 mm)+NADPH generating system; UDPGA (2.5 mm); or PAPS (100 μm); in a 6 ml 100 mm Tris buffer pH 7.8. Incubations were carried out at 37° C in air for 40 min in a shaking metabolic water bath. The reaction was terminated and protein precipitated by adding 2 ml acetone to the reaction mixture. Incubation media were pelleted and the supernatant analysed by h.p.l.c. for quantification of individual radioactive compounds. Irreversibly bound [3H]-EE2 derived radioactivity in the pellet was determined as described below.
Hepatocyte enzyme induction procedure
Primary human induction studies were performed using the following protocol developed by Li et al. [28, 30, 31]: day 1, freshly isolated human liver hepatocytes were transferred to Vitrogen-coated 24-well plates containing Dulbecco’s modified Eagle’s media (DMEM) supplemented with insulin, hydrocortisone, l-glutamine, and MEM nonessential amino acids, BSA, fructose, gentamicin, and amikacin. The culture medium was replaced with sandwich medium after 2–4 h of attachment. The cells were sandwiched with collagen and incubated at 37° C in a 95% air:5% CO2. Media changes were carried out daily. On day 3 and day 4 a selected enzyme inducer drug for CYP1A (OMP, 3-MC) or CYP3A (DEX, RIF, or PB) was added and on day 5 the medium was replaced with Krebs-Hensleit buffer and individual drug experiments were performed.
Hepatocyte drug induction experiments
The hepatocyte induction model was validated following pretreatment of freshly isolated hepatocytes (0.7×106 cells ml−1 density per well) with 3-MC (1 μm), OMP (33.3 μm), DEX (33.3 μm), or RIF (33.3 μm), and non treated hepatocytes. On day 5, media were removed and the hepatocytes were then incubated with KHB containing either 2 μm ethoxyresorufin or 250 μm testosterone for 60 min at 37° C under a 90% air:5% CO2 atmosphere. Following incubations, the CYP1A enzyme activities were determined by measuring the ethoxyresorufin-O-deethylase activity according to the method reported by Burke et al. [36]. CYP3A enzyme activity determinations were based on the methods reported by Sonderfan et al. [37] and Fuane et al. [38].
Hepatocytes isolated from donor livers 1, 2, and 3 were used to investigate the induction of [3H]-EE2 metabolism after pretreatment of hepatocytes (0.7×106 cells ml−1 per well) with known enzyme inducer drugs at the following concentrations: for DEX, RIF, or OMP at 0, 0.33, 1.0, 3.3, 10, and 33.3 μm and for 3-MC or PB at 0, 0.01, 0.033, 0.1, 0.33, and 1 mm. The concentrations used were previously shown by Li et al. [28, 30, 31] to be near optimal in the induction of the corresponding CYP isoenzymes. The metabolism of [3H]-EE2 was determined in pretreated and non treated hepatocytes following incubation with [3H]-EE2 (1 nm, 1.1×105 d min−1/well) for 15 min.
[3H]-EE2 metabolic recovery experiments were conducted with freshly isolated hepatocytes in suspension from donor liver 5 and with non treated hepatocytes isolated from donor livers 6 and 7 following pretreatment with rifampicin (33.3 μm). Hepatocytes in suspension (donor liver 5) were incubated with [3H]-EE2 (1 nm or 100 nm, 1.1×105 d min−1/well) in the presence or absence of 5 μm ketoconazole, a known CYP3 A4 competitive inhibitor (Ki =0.7 μm) [39], for 0.1, 1, 4, and 8 h. The rifampicin pretreated and non treated hepatocyte experiments (donor livers 6 and 7) were incubated with [3H]-EE2 (1 nm, 1.1×105 d min−1/well) for 0.1, 1, 4, and 8 h in the presence or absence of ketoconazole (5 μm).
Quantification of EE2 metabolism after treatment with CYP inducers
Both the incubation mixture and cells were stored at −80° C until analysed by h.p.l.c. To analyse the medium, a 500 μl aliquot of medium was added to 2 ml acetone and centrifuged to remove insoluble material. The solvent was then evaporated using first a stream of dry nitrogen and then a vacuum, the residue was reconstituted with 150 μl 15% acetonitrile in water, and 100 μl (10–50 nCi 3H) was analysed by h.p.l.c. Cells were analysed after releasing them from the flask with 1 ml 0.25% trypsin/0.02% EDTA at 37° C for 10 min. This was added to 2 ml acetone, centrifuged, and dried as above.
Protein bound radioactivity was determined from the radioactivity adhering to the acetone insoluble pellet obtained above. These pellets were washed twice with 5 ml methanol and once with 5 ml water, centrifuging in between. 100 μl acetone washed rat liver homogenate was added to each tube to help maintain the small pellet during the washing procedure. The washed pellets were then disrupted with 500 μl 6 m guanidinium chloride, 10 ml Flo-Scint IV scintillation cocktail was added and the solution was counted for tritium on a Packard Tri-Carb 2500 TR liquid scintillation analyser.
H.p.l.c. conditions consisted of a Hewlett-Packard 1050 h.p.l.c. system with variable wavelength detector. A 250×4.6 mm Alltech Adsorbosphere HS C18 column was used with a mobile phase of 10 mm ammonium phosphate pH 2.7 progressing to 70% acetonitrile/30% 10 mm ammonium phosphate pH 2.7 over 25 min (holding at this composition for 10 min) at a flow rate of 1 ml min−1. Detection was by radioactivity using a Radiomatic Flo-One/Beta detector set for tritium and Ultima-Flo M scintillation cocktail or by u.v. at 265 nm.
Measurement of hepatocyte homogenate protein concentration
Protein content of the freshly isolated human hepatocytes was determined with using a BCA Protein Assay (Pierce Chemical Co., 1993) using BSA as protein standards.
Statistical analysis
Statistical differences between control and induction experiments were determined using the Student’s t-test. Pairwise comparisons of non treated and treated hepatocyte induction experiments in the presence and absence of ketoconazole were analysed using one way anova and the Tukey test using a confidence interval of 95%. Statistical analyses were performed using SigmaStat for Windows version 2.0 (SPPS Inc., San Rafael CA).
Results
Identification of EE2 metabolites
EE2 and its metabolites were identified after h.p.l.c. analysis of the incubation medium by comparing the individual metabolite elution times with synthesized reference compounds (figure 2a). Following separate incubations of [3H]-EE2 with human S9 fractions in the presence of either NADPH, PAPS, or NADPH plus SAM, single h.p.l.c. metabolite peaks were observed with elution times corresponding to 2-OH-EE2 (24.2 min), EE2–3-sulphate (21.7 min), or 2-methoxy-EE2 (27.2 min), respectively. Incubation of [3H]-EE2 with S9 fractions in the presence of UDPGA produced a single peak with an h.p.l.c. elution time of 19.9 min. Although reference EE2–3-glucuronide was not available and the elution time of the expected [3H]-EE2–3-glucuronide peak would not be resolved from EE2–3,17-di-sulphate, the latter compound would not be produced by the S9 fraction in the absence of PAPS cofactor.
Figure 2.
H.p.l.c. chromatograms of (a) synthesized oestrogen compounds, u.v. detection at 265 nm; (b) extract from 4 h EE2 (1 nm) incubation with isolated human hepatocytes in vitro, radiochemical detection.
Figure 2b is a h.p.l.c. radio-chromatogram of a medium extract following a 4 h incubation of [3H]-EE2 with human isolated hepatocytes. Peaks observed with elution times of 21.7 min and 26.3 min corresponded to EE2–3-sulphate and EE2 reference compounds, respectively. The peak at 19.9 min was identified as EE2–3-glucuronide similar to that when [3H]-EE2 was incubated with S9 fraction in the presence of UDPGA. Although this peak could represent EE2–3,17-sulphate, it is highly unlikely since this compound was not observed following incubation of [3H]-EE2 with S9 fraction in the presence of PAPS cofactor. Finally, the 2-methoxy-EE2-conjugate was identified following enzyme hydrolysis by glucuronidase and sulphatase enzymes. The resultant hydrolysed compound had an elution time of 27.2 min corresponding to 2-methoxy-EE2 (Figure 2a).
Human liver S9 fraction incubations in vitro
[3H]-EE2 drug metabolism by human liver S9 fractions prepared from donor livers 1, 2, and 3 were investigated in the presence or absence of individual cofactors. Formation of [3H]-EE2 metabolites was quantified by h.p.l.c. [3H]-EE2 incubation with three individual donor liver S9 fractions (Table 2) in the presence of NADPH produced 3.5–5.1% 2-hydroxy-EE2 and 4.6–9.5% of [3H]-EE2 derived radioactivity was covalently bound to S9 protein. When [3H]-EE2 was incubated with human liver S9 fractions in the presence of UDPGA, 5.7–8.7% of the added [3H]-EE2 was converted directly to [3H]-EE2–3-glucuronide. In the presence of PAPS [3H]-EE2 was rapidly and extensively converted, 67.2–78.7%, to [3H]-EE2-sulphate. Minimal covalent binding was observed in the presence of either UDPGA (0.9–1.4%) or PAPS (1.1–1.8%).
Table 2.
EE2 (1 nm) incubation (40 min) with human liver S9 fraction. Individual metabolites formed as percent total added radioactivity: cofactor dependence.
Further cofactor incubation experiments were carried out using human liver S9 fraction prepared from donor liver 4 to investigate cofactor dependence on the disposition of [3H]-EE2. The results listed in Table 3 for incubations of [3H]-EE2 (1 nm) in the presence of three individual cofactors NADPH, UDPGA, or PAPS produced similar results to those listed in Table 2 for donor livers 1, 2, and 3. When SAM and NADPH were added to the media, 2-methoxy- [3H]-EE2 was the predominate molecular species observed (mean =15.6±3.1% s.d.) and covalent binding was reduced to a mean of 7.3±0.1% s.d. compared with 15.2±7.3% s.d. when only NADPH was present. When SAM and NADPH were present, 2-hydroxy- [3H]-EE2 represented a mean 1.9%±0.7% s.d. of metabolized [3H]-EE2. When all four cofactors were incubated with [3H]-EE2, small amounts of covalently bound radioactivity (1.7±0.1% s.d.) and 2-methoxy- [3H]-EE2 (2.6±0.5% s.d) were observed, however, similar to the data shown in Table 2 the major metabolite formed was [3H]-EE2–3-sulphate (mean =75.7±7.6% s.d.). No 2-methoxy- [3H]-EE2–3-conjugate was detected.
Table 3.
EE2 (1 nm) incubation (40 min) with human liver* S9 fraction. Individual metabolites as percent total radioactivity added: cofactor dependence.
Validation of human hepatocyte induction model
The isolated liver hepatocyte induction model was tested using freshly prepared hepatocytes from donor liver 1. Production rates of 6β-hydroxytestosterone from testosterone by human liver hepatocytes pretreated with rifampicin or dexamethasone were significantly greater (P <0.001) than that observed for control hepatocytes as shown in Figure 3a. Likewise Figure 3b shows that pretreatment of hepatocytes with 3-methylcholanthrene or omeprazole significantly increased the conversion rate of ethoxyresorufin to desethyl ethoxyresorufin (P <0.001) compared with control hepatocytes.
Figure 3.
Rates of metabolite formation following pretreatment of hepatocytes prepared from Donor 1 with 3-methylcholanthrene (1 μm), omeprazole (33 μm), dexamethasone (33 μm), rifampicin (33.3 μm) or non treated. (a) conversion rates of testosterone (250 μm) to 6β-hydroxy-testosterone, 60 min incubation; (b) conversion rates of ethoxyresorufin (2 μm) to resorufin, 60 min incubation. Each data point represents three individual determinations. Error bars are ±s.d.
Rates of [3H]-EE2 metabolism by inducer pretreated and non treated isolated human hepatocytes
Freshly prepared hepatocytes from the same individual donor livers 1, 2, and 3 used in the S9 experiments were either non treated or pretreated with varying concentrations of the P450 enzyme inducer, rifampicin. Following the treatment period, [3H]-EE2 (1 nm) conversion rates to [3H]-EE2–3-glucuronide and [3H]-EE2–3-sulphate were determined. As shown in Figure 4a, pretreatment of hepatocytes with rifampicin at 3.3, 10, and 33.3 μm concentrations resulted in significant increases in the conversion rates of [3H]-EE2– [3H]-EE2–3-sulphate compared with non treated hepatocytes (P <0.01). With the exception of 0.3 μm rifampicin treated sample, P =0.03, no significant differences (P 0.2) were observed between treated and non treated hepatocytes in the production of [3H]-EE2–3- glucuronide (Figure 4b).
Figure 4.
Rates of (a) [3H]-EE2–3-sulphate or (b) [3H]-EE2–3-glucuronide formation by non treated or pretreated human hepatocytes with varying rifampicin concentrations and following a 15 min [3H]2-EE2 (1 nm) incubation with: (▪) Donor liver 1; () Donor liver 2; (
) Donor liver 3. n =3 determinations ±s.d. for each experiment.
In addition to the rifampicin experiments, human liver hepatocytes prepared from donor livers 3 and 4 were pretreated with varying concentrations of 3-methylcholanthrene, omeprazole, phenobarbitone or dexamethasone. Although some individual experiments showed statistical differences between rates of [3H]-EE2 conversion to [3H]-EE2–3-glucuronide and [3H]-EE2–3- sulphate for pretreated compared with non treated hepatocytes, no consistent significant pattern was established.
[3H]-EE2 metabolic time profiles for rifampicin treated and non treated human hepatocyte experiments
Mass balance experiments were conducted with non treated freshly prepared hepatocyte suspensions from liver donor 5 and with rifampicin pretreated or non-treated hepatocytes from donor livers 6 and 7 to determine the fate of [3H]-EE2 derived radioactivity added to the medium. The recovery of [3H]-EE2 derived radioactivity was examined following incubation of [3H]-EE2 (1 nm) with hepatocytes for 0.1, 1.0, 4.0, or 8.0 h in the presence or absence of ketoconazole (5 μm). H.p.l.c. analysis of media extracts using radiochemical detection provided metabolic time profiles for the recovery experiments. Results of this analysis are shown for donor liver 5 and donor liver 6 in Figure 5 and Figure 6, respectively. Results obtained for donor liver 7 were similar to those observed for donor liver 6 (data for donor liver 7 not shown). [3H]-EE2 metabolic peaks identified in the h.p.l.c. radiochromatograms were [3H]-EE2–3-sulphate, [3H]-EE2–3-glucuronide, and a 2-methoxy- [3H]-EE2–3-conjugate. no 2-hydroxy- [3H]-EE2 or its conjugates were observed in any of the [3H]-EE2 hepatocyte incubations.
Figure 5.
Media metabolites recovered after incubations of [3H]-EE2 (1 nm) on day 0 with human hepatocytes in suspension prepared from Donor liver 5. a) Hepatocytes and [3H]-EE2 (1 nm) b) Hepatocytes, [3H]-EE2 (1 nm), and ketoconazole (5 μm). Symbols represent: (○) EE2; (▿) 2-methoxy-EE2–3-conjugate; (▵) EE2–3-glucuronide; (□) EE2–3-sulphate. Each data point represents three individual determinations and error bars are s.d.
Figure 6.
Media metabolites recovered after incubation of [3H]-EE2 (1 nm) with human liver hepatocytes prepared from Donor liver 6: a) Non treated hepatocytes b) Rifampicin treated (33 μm). c) Non treated hepatocytes and ketoconazole (5 μm). d) Rifampicin treated (33 μm) and ketoconazole (5 μm). Symbols represent: (○) EE2; (▿) 2-methoxy-EE2–3-conjugate; (▵) EE2–3-glucuronide; (□) EE2–3-sulphate. Each data point represents three individual determinations and error bars are s.d.
One way anova and pairwise comparison using the Tukey Test were performed on the 1 h and 8 h time points to establish the effect of rifampicin pretreatment and addition of 5 μm ketoconazole to each set of donor liver experiments. Parent drug remaining in the media following a 8 h incubation was ≤10% for all incubations shown in Figure 5 and Figure 6. No statistical differences were observed in metabolic profiles obtained for donor liver 5 hepatocytes following incubations of [3H]-EE2 on day 0 in the absence (Figure 5a) or presence (Figure 5b) of 5 μm ketoconazole. Pretreatment of hepatocytes prepared from donor liver 6 (Figure 6b) with rifampicin resulted in a statistically significant increase in production of [3H]-EE2–3-sulphate from [3H]-EE2 compared with untreated hepatocytes (Figure 6a). Similarly, in the presence of 5 μm ketoconazole, increased amounts of [3H]-EE2–3-sulphate were observed for rifampicin treated hepatocyte (Figure 6d) compared with non rifampicin treated hepatocytes (Figure 6c). Addition of ketoconazole (5 μm) inhibitor appeared to have no effect on the induction of [3H]-EE2 sulphation by rifampicin. There was a statistically significant decrease in the production of [3H]-EE2–3-glucuronide in the rifampicin pretreated hepatocyte experiments compared with controls. Ketoconazole appeared to have no effect on either sulphation or glucuronidation.
Production of the 2-methoxy- [3H]-EE2–3-conjugate was minimal (less than 5%) in hepatocytes prepared from all three donor livers under all incubation conditions. A statistical analysis of the [3H]-2-methoxy-EE2– 3-conjugate was not attempted due to the low number of radioactive counts observed in the h.p.l.c. radiochromatograms. Unlike the S9 studies, there was no detectable [3H]-EE2 derived radioactivity covalently bound to cellular protein.
Discussion
Numerous reports in the literature have implicated drug–drug interactions for decreased effectiveness of EE2 and therapeutic failures of oral contraceptives (for recent reviews see Helms et al. [2]; Shenfield & Griffin [3], and Orme et al. [4]. The two main mechanisms by which simultaneous administration of oral contraceptive pills and other drugs are believed to reduce oral contraceptive steroid efficacy are enzyme induction and impaired enterohepatic circulation. EE2 is excreted in urine and bile after oxidation and direct conjugation reactions, however, most investigations have focused on induction of the major CYP450 oxidative pathway that converts EE2 to the catechol, 2-hydroxy-EE2. Individual CYP450 enzymes reported to catalyse this reaction include: CYP2E, CYP2C, and CYP3A [25, 26]. The work of Guengerich [23] highlighted CYP3A4, known to be induced by numerous drugs, as the major CYP450 enzyme responsible for EE2 oxidation. In this regard, evidence exists in the literature implicating that coadministration of rifampicin and oral contraceptives can result in therapeutic failures [18–20].
Urinary and biliary excretion of radioactivity following human ingestion of [3H]-EE2 has been studied by a few investigators and their results suggest that EE2 oxidation may represent a minor elimination pathway compared to direct EE2 conjugation [13–16]. The available data suggest that factors other than induction of EE2 oxidative enzymes could contribute to the observed therapeutic failures when rifampicin is coadministered with oral contraceptive pills.
Our investigation examined the fate of [3H]-EE2 when incubated with human liver S9 fractions and freshly isolated human hepatocytes and points out the importance of using intact liver cells when evaluating relative contributions of parallel metabolic pathways that include direct conjugation of the parent drug. Similar to other authors, we demonstrated that human liver fractions in the presence of NADPH are capable of producing both 2-methoxy-EE2 and 2-hydroxy-EE2 and that during EE2 oxidation, a highly reactive species is formed which covalently binds to cellular protein [40–46]. Formation of EE2–3-glucuronide or EE2–3-sulphate, was also observed when the appropriate cofactor was added to the medium as reported by others [47, 48] (Tables 2 and 3). However, when all cofactors including NADPH were added to the medium EE2–3-sulphate predominated representing 76% of the metabolized EE2 (Table 3). Although our results with S9 human liver fractions in vitro suggest the importance of EE2 conjugation, the artificial nature of the system, i.e. addition of cofactors, limits reliable extrapolation of our results to humans in vivo.
Freshly isolated human hepatocytes represent a more relevant model for predicting the relative contributions of parallel metabolic pathways than sub cellular liver fractions since hepatocytes contain the full complement of metabolizing enzymes and cofactors present in the intact liver. In addition, they offer the capability to investigate induction of liver enzymes and the resulting effect on a drug’s metabolism. The results of our EE2 human liver hepatocyte metabolism experiments in vitro are consistent with a number of observations reported in humans and demonstrate the advantage of hepatocytes for these types of drug metabolism studies. Firstly, we observed that the EE2 oxidation leading to 2-hydroxy-EE2 and a secondary metabolite, 2-methoxy-EE2, followed by conjugation may represent a minor metabolic pathway similar to that observed in human subjects. Under our experimental conditions, no 2-hydroxy-EE2 was detected. Although it is possible that some 2-hydroxy- [3H]-EE2 was formed and then reacted to form covalently bound species, the mass balance indicates that it could be only a very small fraction of total metabolism. Also, less than 10% of metabolized [3H]-EE2 was observed as a 2-methoxy- [3H]-EE2 conjugate (Figures 5 and 6). Secondly, we have demonstrated that human liver hepatocytes are capable of forming EE2–3-glucuronide and EE2–3-sulphate. A comparison of the data in Figure 5a and Figure 6a demonstrates the individual variation in the relative amounts of EE2–3-glucronide and EE2-sulphate produced and is similar to observations reported for EE2 conjugates excreted in human urine and bile following EE2 administration [13]. The rapid metabolism of EE2 to EE2–3-sulphate by human liver hepatocytes shown in Figures 5 and 6 points out the importance of obtaining early time time points during hepatocyte incubation experiments.
Direct conjugation and excretion of EE2 as opposed to EE2 oxidation represents the predominant elimination pathway for this drug in both human hepatocytes in vitro and humans in vivo. These observations suggest that mechanisms other than induction of P450 enzymes by known inducer drugs may be partly responsible for therapeutic failures observed with administration of oral contraceptives.
We observed no induction of EE2–3-glucuronide formation in rifampicin treated hepatocytes over non treated. Although Li et al. [49] observed a slight increase (0.3 fold) in zidovudine glucuronidation by rifampicin treated hepatocytes, this was only observed in one of two livers examined. A novel finding of our investigation supports this hypothesis in that metabolism of EE2 to EE2–3-sulphate by human liver hepatocytes prepared from three individual donor livers was induced following pretreatment with the known enzyme inducer drug, rifampicin. Kern et al. [50] observed increased sulphation of p-nitrophenol following rifampicin (50 μm) pretreatment of rat or human hepatocytes for 7 and 12 days. Increases in the sulphation of p-nitrophenol by pretreated hepatocytes were 2.0–2.5 fold (rats) and 2.0 fold (human) compared with controls. Our study did not directly address the individual enzyme responsible for EE2 sulphation and there has been some controversy over the identity of human ST(s) responsible for the sulphation of oestrogens and EE2. Falany et al. [51] reported that EE2 at 1.5 μm was sulphated to form EE2–3-sulphate by human cytosolic phenol sulphotransferase (P-PST) expressed in bacteria. A distinct human oestrogen sulphotransferase (hEST-1) was isolated by Falany et al. [52] from human liver and expressed in two bacterial expression systems. This enzyme which maximally sulphates beta estradiol and estrone at concentrations of 20 nm is also capable of sulphating EE2. Frobes-Bamforth & Coughtrie [53] presented evidence for the existence of a sulphotranseferase in adult human liver which sulphates both estrone and EE2 but not planar phenols. Based on this information, it appears likely that at 1 nm EE2 concentrations the enzyme described by Frobes-Bamforth is responsible for the conversion of EE2 to its sulphated conjugate observed in our study. If this assumption is correct, this is the first demonstration of induction of oestrogen sulphotransferase by rifampicin in human hepatocytes, however, further studies will be required to confirm this hypothesis.
In summary, our results show that primary human hepatocyte cultures possess extensive sulphation and glucuronidation activities and therefore can be applied as an experimental model for the evaluation of drug–drug interactions involving phase II conjugating pathways. Our study represents the first to suggest induction of EE2–3-sulphation by pretreatment of human hepatocytes with rifampicin. The major EE2 metabolic products observed in human hepatocytes in vitro are direct EE2 conjugates with EE2 oxidation representing minor pathways. Further studies are required to establish the clinical relevance of our findings.
References
- 1.Murad F, Kuret JA. Estrogens and progestins. In: Gilman AG, Rall TW, Nies AS, editors. The Pharmacological Basis of Therapeutics. 8. New York: Pergamon Press; 1990. pp. 1384–1412. [Google Scholar]
- 2.Helms SE, Bredle DL, Zajic J, Jarjoura D, Brodell RT, Krishnarao I. Oral contraceptive failure rates and oral antibiotics. J Am Acad Dermatol. 1997;36:705–710. doi: 10.1016/s0190-9622(97)80322-2. [DOI] [PubMed] [Google Scholar]
- 3.Shenfield GM, Griffin JM. Clinical pharmacokinetics of contraceptive steroids. Clin Pharmacokinet. 1991;20:15–37. doi: 10.2165/00003088-199120010-00002. [DOI] [PubMed] [Google Scholar]
- 4.Orme ML, Back DJ, Breckenridge AM. Clinical pharmacokinetics of oral contraceptive steroids. Clin Pharmacokinet. 1983;8:95–136. doi: 10.2165/00003088-198308020-00001. [DOI] [PubMed] [Google Scholar]
- 5.Van Dijke CP, Weber JC. Interaction between oral contraceptives and griseofulvin. Br Med J (Clin Res. eds) 1984;288:1125–1126. doi: 10.1136/bmj.288.6424.1125-a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.McDaniel PA, Caldroney RD. Oral contraceptives and griseofulvin interactions. Drug Intell Clin Pharm. 1986;20:384. doi: 10.1177/106002808602000511. [DOI] [PubMed] [Google Scholar]
- 7.Back DJ, Orme ML. Pharmacokinetic drug interactions with oral contraceptives. Clin Pharmacokinet. 1990;18:472–484. doi: 10.2165/00003088-199018060-00004. [DOI] [PubMed] [Google Scholar]
- 8.Back DJ, Grimmer SF, Orme ML, Proudlove C, Mann RD, Breckenridge AM. Evaluation of committee on safety of medicines yellow card reports on antibiotics. Br J Clin Pharmacol. 1988;25:527–532. doi: 10.1111/j.1365-2125.1988.tb03341.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Abdel-Aziz MT, Williams KI. Metabolism of radioactive 17α-ethynylestradiol by women. Steroids. 1970;15:695–710. doi: 10.1016/s0039-128x(70)80074-5. [DOI] [PubMed] [Google Scholar]
- 10.Kamyab S, Fotherby K, Steele SJ. Metabolism of 4–14C-ethynyl oestradiol in women. Nature. 1969;221:360–361. doi: 10.1038/221360a0. [DOI] [PubMed] [Google Scholar]
- 11.Kulkarni BD, Goldzieher JW. A preliminary report on urinary excretion pattern and method of isolation of 14C-ethynylestradiol metabolites in women. Contraception. 1970;1:47–55. [Google Scholar]
- 12.Reed MJ, Steele SJ. Metabolism of ethynyloestradiol in man. J Endocrinol. 1972;55:351–361. doi: 10.1677/joe.0.0550351. [DOI] [PubMed] [Google Scholar]
- 13.Maggs JL, Park BK. A comparative study of biliary and urinary 2-hydroxylated metabolites of [6, 7–3H] 17′-ethynylestradiol in women. Contraception. 1985;32:173–182. doi: 10.1016/0010-7824(85)90105-2. [DOI] [PubMed] [Google Scholar]
- 14.Maggs JL, Grimmer SFM, Orme ML, Breckenridge AM, Park BK, Gilmore IT. The biliary and urinary metabolites of [3H]17α-ethynylestradiol in women. Xenobiotica. 1983;13:421–431. doi: 10.3109/00498258309052280. [DOI] [PubMed] [Google Scholar]
- 15.Cargill DI, Steinetz BG, Gosnell E, et al. Fate of ingested radiolabeled ethynylestradiol and its 3-cyclopentyl ether in patients with bile fistulas. J Clin Endocrinol. 1969;29:1051–1061. doi: 10.1210/jcem-29-8-1051. [DOI] [PubMed] [Google Scholar]
- 16.Williams MC, Helton ED, Goldzieher JW. The urinary metabolites of 17α-ethynylestradiol-9α,11χ-3H in women. Chromatographic profiling and identification of ethynyl and non-ethynyl compounds. Steroids. 1975;25:229–246. doi: 10.1016/s0039-128x(75)90135-x. [DOI] [PubMed] [Google Scholar]
- 17.Williams MC, Goldzieher JW. Chromatographic patterns of urinary ethynyl estrogen metabolites in various populations. Steroids. 1980;36:255–281. doi: 10.1016/0039-128x(80)90001-x. [DOI] [PubMed] [Google Scholar]
- 18.Back DJ, Breckenridge AM, Crawford FE, et al. The effect of rifampicin on the pharmacokinetics of ethynylestradiol in women. Contraception. 1980;21:135–143. doi: 10.1016/0010-7824(80)90125-0. [DOI] [PubMed] [Google Scholar]
- 19.Bolt HM, Kappus H, Bolt M. Effect of rifampicin treatment on the metabolism of oestradiol and 17alpha-ethinyloestradiol by human liver microsomes. Eur J Clin Pharmacol. 1975;8:301–307. doi: 10.1007/BF00562654. [DOI] [PubMed] [Google Scholar]
- 20.Bolt HM, Bolt M, Kappus R. Interaction of rifampicin treatment with pharmacokinetics and metabolism of ethinyloestradiol in man. Acta Endocrinol (Copenh) 1977;85:189–197. doi: 10.1530/acta.0.0850189. [DOI] [PubMed] [Google Scholar]
- 21.Barditch-Crovo P, Trapnell CB, Ette E, et al. The effects of rifampin and rifabutin on the pharmacokinetics and pharmacodynamics of a combination oral contraceptive. Clin Pharmacol Ther. 1999;65:428–438. doi: 10.1016/S0009-9236(99)70138-4. [DOI] [PubMed] [Google Scholar]
- 22.Crawford P, Chadwick DJ, Martin C, Tjia J, Back DJ, Orme ML. The interaction of phenytoin and carbamazepine with combined oral contraceptive steroids. Br J Clin Pharmacol. 1990;30:892–896. doi: 10.1111/j.1365-2125.1990.tb05457.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Back DJ, Bates M, Bowden A, et al. The interaction of phenobarbital and other anticonvulsants with oral contraceptive steroid therapy. Contraception. 1980;22:495–503. doi: 10.1016/0010-7824(80)90102-x. [DOI] [PubMed] [Google Scholar]
- 24.Fattore C, Cipolla G, Gatti G. Induction of ethinylestradiol and levonorgestrel metabolism by oxcarbazepine in healthy women. Epilepsia. 1999;40:783–787. doi: 10.1111/j.1528-1157.1999.tb00779.x. [DOI] [PubMed] [Google Scholar]
- 25.Guengerich F. Oxidation of 17α-ethynylestradiol by human liver cytochrome P-450. Mol Pharmacol. 1988;33:500–508. [PubMed] [Google Scholar]
- 26.Ball SE, Forrester LM, Wolf CR, Back DJ. Differences in the cytochrome P-450 isoenzymes involved in the 2-hydroxylation of oestradiol and 17α-ethinyloestradiol. Biochem J. 1990;267:221–226. doi: 10.1042/bj2670221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Li AP, Beck DJ, Loretz LJ. Primary human hepatocytes as an in vitro toxicology system. In: Salem H, Katz SA, editors. Advances in Animal Alternatives for Safety and Efficacy. New York NY: Taylor and Francis; 1998. [Google Scholar]
- 28.Li AP. Drug–Drug Interactions: Scientific and Regulatory Perspectives. Vol. 43. San Diego: Academic Press; 1997. Primary human hepatocytes as an experimental system for the evaluation of pharmacokinetic drug–drug interactions; pp. 103–130. [Google Scholar]
- 29.Li AP. Primary hepatocyte culture as an in vitro toxicological system. In: Gad S, editor. In Vitro Toxicology. New York NY: Raven Press; 1994. pp. 195–220. [Google Scholar]
- 30.Li AP, Jurima-Romet M. Applications of primary human hepatocytes in the evaluation of pharmacokinetic drug–drug interactions: evaluation of model drugs terfenadine and rifampin. Cell Biol Toxicol. 1997;13:365–374. doi: 10.1023/a:1007451911843. [DOI] [PubMed] [Google Scholar]
- 31.Li AP, Rasmussen A, Xu L, Kaminski DL. Rifampicin induction of lidocaine metabolism in cultured human hepatocytes. J Pharmacol Exp Ther. 1995;274:673–677. [PubMed] [Google Scholar]
- 32.Rendic S, Di Carlo FJ. Human cytochrome P450 enzymes: a status report summarizing their reactions, substrates, inducers, and inhibitors. Drug Metab Rev. 1997;29:413–581. doi: 10.3109/03602539709037591. [DOI] [PubMed] [Google Scholar]
- 33.Li AP, Roque MA, Beck DJ, Kaminiski DL. Isolation and culturing of hepatocytes from human liver. J Tissue Culture Meth. 1992;14:139–146. [Google Scholar]
- 34.Guengerich FP, Distlerath LM, Reilly PEB, et al. Human liver cytochromes P-450 involved in polymorphism of drug oxidations. Xenobiotica. 1985;16:367–378. doi: 10.3109/00498258609050245. [DOI] [PubMed] [Google Scholar]
- 35.Ludden LK, Strong JM, Kohn EC, Collins JM. Similarity of metabolism for CAI (NSC 609974) in human liver tissue in vitro and in humans in vivo. Clin Cancer Res. 1995;1:399–405. [PubMed] [Google Scholar]
- 36.Burke MD, Thompson S, Elcombe CR, Halpert J, Haaparanta T, Mayer RT. Ethoxy-, pentoxy-, and benzyloxyphenoxazones and homologues: a series of substrates to distinguish between different induced cytochromes P450. Biochem Pharmacol. 1985;34:3337–3345. doi: 10.1016/0006-2952(85)90355-7. [DOI] [PubMed] [Google Scholar]
- 37.Sonderfan AJ, Arlotto MP, Dutton DR, McMillen SK, Parkinson A. Regulation of testosterone hydroxylation by rat liver microsomal cytochrome P450. Arch Biochem Biophys. 1987;255:27–41. doi: 10.1016/0003-9861(87)90291-8. [DOI] [PubMed] [Google Scholar]
- 38.Funae Y, Imaoka S. Purification and characterization of liver microsomal cytochrome P450 from untreated male rats. Biochim Biophys Acta. 1987;926:349–358. doi: 10.1016/0304-4165(87)90221-2. [DOI] [PubMed] [Google Scholar]
- 39.Pichard L, Fabre I, Fabre G, et al. Screening for inducers and inhibitors of cytochrome P-450 (cyclosporin A oxidase) in primary cultures of human hepatocytes and in liver micromsomes. Drug Metab Dispos. 1990;18:595–606. [PubMed] [Google Scholar]
- 40.Orme ML, Back DJ, Ball S. Interindividual variation in the metabolism of ethynylestradiol. Pharmac Ther. 1989;43:251–260. doi: 10.1016/0163-7258(89)90121-6. [DOI] [PubMed] [Google Scholar]
- 41.Purba HS, Maggs JL, Orme ML, Back DJ, Park BK. The metabolism of 17α-ethinyloestradiol by human liver microsomes: formation of catechol and chemically reactive metabolites. Br J Clin Pharmac. 1987;23:447–453. doi: 10.1111/j.1365-2125.1987.tb03074.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Stramentinoli G, Gualano M, Rovagnati P, Padova C. Influence of S-adenosyl-l-methionine on irreversible binding of ethynylestradiol to rat liver microsomes, and its implication in bile secretion. Biochem Pharmacol. 1779;28:981–984. doi: 10.1016/0006-2952(79)90290-9. [DOI] [PubMed] [Google Scholar]
- 43.Horning EC, Thenot JP, Helton ED. Toxic agents resulting from the oxidative metabolism of steroid hormones and drugs. J Toxicol Environ Health. 1978;4:341–361. doi: 10.1080/15287397809529665. [DOI] [PubMed] [Google Scholar]
- 44.Kitteringham NR, Lambert C, Maggs JL, Colbert J, Park BK. A comparative study of the formation of chemically reactive drug metabolites by human liver microsomes. Br J Clin Pharmacol. 1988;26:13–21. doi: 10.1111/j.1365-2125.1988.tb03358.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kappus H, Bolt HM, Remmer H. Irreversible protein binding of metabolites of ethynylestradiol in vivo and in vitro. Steroids. 1973;22:203–205. doi: 10.1016/0039-128x(73)90086-x. [DOI] [PubMed] [Google Scholar]
- 46.Bolt HM, Kappus H, Kasbohrer R. Metabolism of 17α-ethinylestradiol by human liver microsomes in vitro: aromatic hydroxylation and irreversible protein binding of metabolites. Endrocrinol Metab. 1974;39:1072–1080. doi: 10.1210/jcem-39-6-1072. [DOI] [PubMed] [Google Scholar]
- 47.Pacifici GM, Back DJ. Sulphation and glucuronidation of ethinyloestradiol in human liver in vitro. J Steroid Biochem. 1988;31:345–349. doi: 10.1016/0022-4731(88)90360-3. [DOI] [PubMed] [Google Scholar]
- 48.Temellini A, Giuliani L, Pacifici GM. Interindividual variability in the glucuronidation and sulphation of ethinyloestradiol in human liver. Br J Clin Pharmacol. 1991;31:661–664. doi: 10.1111/j.1365-2125.1991.tb05589.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Li AP, Reith MK, Rasmussen A, et al. Primary human hepatocytes as a tool for the evaluation of structure-activity relationship in cytochrome P450 induction potential of xenobiotics: evaluation of rifampin, rifapentine, and rifabutin. Chem Biol Interact. 1997;107:17–30. doi: 10.1016/s0009-2797(97)00071-9. [DOI] [PubMed] [Google Scholar]
- 50.Kern A, Bader A, Pichlmayr R, Sewing K-F. Drug metabolism in hepatocyte sandwich cultures of rats and humans. Biochem Pharmacol. 1997;54:761–772. doi: 10.1016/s0006-2952(97)00204-9. [DOI] [PubMed] [Google Scholar]
- 51.Falany CN, Wheeler J, Oh TS, Falany JL. Steroid sulfation by expressed human cytosolic sulfotransferases. J Steroid Biochem Molec Biol. 1994;48:369–375. doi: 10.1016/0960-0760(94)90077-9. [DOI] [PubMed] [Google Scholar]
- 52.Falany CN, Krasnykh H, Falany JL. Bacterial expression and characterization of a cDNA for human liver estrogen sulfotransferase. J Steroid Biochem Mol Biol. 1995;52:529–539. doi: 10.1016/0960-0760(95)00015-r. [DOI] [PubMed] [Google Scholar]
- 53.Frobes-Bamforth KJ, Coughtrie MWH. Identification of new adult human liver sulfotransferase with specificity for endogenous and xenobiotic estrogens. Biochem Biophys Res Comm. 1994;198:707–711. doi: 10.1006/bbrc.1994.1102. [DOI] [PubMed] [Google Scholar]